Network


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

Hotspot


Dive into the research topics where Jinjin Feng is active.

Publication


Featured researches published by Jinjin Feng.


Cancer Research | 2009

Identification of MCAM/CD146 as the target antigen of a human monoclonal antibody that recognizes both epithelioid and sarcomatoid types of mesothelioma.

Scott Bidlingmaier; Jiang He; Yong Wang; Feng An; Jinjin Feng; Dario Barbone; Dong-Wei Gao; Ben Franc; V. Courtney Broaddus; Bin Liu

The prognosis for patients diagnosed with mesothelioma is generally poor, and currently available treatments are usually ineffective. Therapies that specifically target tumor cells hold much promise for the treatment of cancers that are resistant to current approaches. We have previously selected phage antibody display libraries on mesothelioma cell lines to identify a panel of internalizing human single chain (scFv) antibodies that target mesothelioma-associated, clinically represented cell surface antigens and further exploited the internalizing function of these scFvs to specifically deliver lethal doses of liposome-encapsulated small molecule drugs to both epithelioid and sarcomatous subtypes of mesothelioma cells. Here, we report the identification of MCAM/MUC18/CD146 as the surface antigen bound by one of the mesothelioma-targeting scFvs using a novel cloning strategy based on yeast surface human proteome display. Immunohistochemical analysis of mesothelioma tissue microarrays confirmed that MCAM is widely expressed by both epithelioid and sarcomatous types of mesothelioma tumor cells in situ but not by normal mesothelial cells. In addition, quantum dot-labeled anti-MCAM scFv targets primary meosthelioma cells in tumor fragment spheroids cultured ex vivo. As the first step in evaluating the therapeutic potential of MCAM-targeting antibodies, we performed single-photon emission computed tomography studies using the anti-MCAM scFv and found that it recognizes mesothelioma organotypic xenografts in vivo. The combination of phage antibody library selection on tumor cells and rapid target antigen identification by screening the yeast surface-displayed human proteome could be a powerful method for mapping the targetable tumor cell surface epitope space.


The Journal of Nuclear Medicine | 2010

Targeting Prostate Cancer Cells In Vivo Using a Rapidly Internalizing Novel Human Single-Chain Antibody Fragment

Jiang He; Yong Wang; Jinjin Feng; Xiaodong Zhu; Xiaoli Lan; Arun K. Iyer; Niu Zhang; Youngho Seo; Henry F. VanBrocklin; Bin Liu

Human antibodies targeting prostate cancer cell surface epitopes may be useful for imaging and therapy. The objective of this study was to evaluate the tumor targeting of an internalizing human antibody fragment, a small-size platform, to provide high contrast in a mouse model of human prostate carcinoma. Methods: A prostate tumor–targeting single-chain antibody fragment (scFv), UA20, along with a nonbinding control scFv, N3M2, were labeled with 99mTc and evaluated for binding and rapid internalization into human prostate tumor cells in vitro and tumor homing in vivo using xenograft models. For the in vitro studies, the labeled UA20 scFv was incubated at 37°C for 1 h with metastatic prostate cancer cells (DU145) to assess the total cellular uptake versus intracellular uptake. For the animal studies, labeled UA20 and N3M2 scFvs were administered to athymic mice implanted subcutaneously with DU145 cells. Mice were imaged with small-animal SPECT/CT with concomitant biodistribution at 1 and 3 h after injection. Results: The UA20 scFv was labeled in 55%−65% yield and remained stable in phosphate buffer within 24 h. The labeled UA20 scFv was taken up specifically by prostate tumor cells. Internalization was rapid, because incubation at 37°C for less than 1 h resulted in 93% internalization of total cell-associated scFvs. In animal studies, SPECT/CT showed significant tumor uptake as early as 1 h after injection. At 3 h after injection, tumor uptake was 4.4 percentage injected dose per gram (%ID/g), significantly greater than all organs or tissues studied (liver, 2.7 %ID/g; other organs or tissues, <1 %ID/g), except the kidneys (81.4 %ID/g), giving tumor-to-blood and tumor-to-muscle ratios of 12:1 and 70:1, respectively. In contrast, the control antibody exhibited a tumor uptake of only 0.26 %ID/g, similar to that of muscle and fat. Tumor-specific targeting was evidenced by reduced tumor uptake of nearly 70% on administration of a 10-fold excess of unlabeled UA20 scFv. Kidney uptake was nonspecific, consistent with the route of excretion by scFvs. Conclusion: The UA20 scFv showed rapid and specific internalization in prostate tumor cells in vitro and accumulation in prostate tumor xenografts in vivo, demonstrating the potential for future development for prostate cancer imaging and targeted therapy.


Biomaterials | 2011

The effect of internalizing human single chain antibody fragment on liposome targeting to epithelioid and sarcomatoid mesothelioma

Arun K. Iyer; Yang Su; Jinjin Feng; Xiaoli Lan; Xiaodong Zhu; Yue Liu; Dong-Wei Gao; Youngho Seo; Henry F. VanBrocklin; V. Courtney Broaddus; Bin Liu; Jiang He

Immunoliposomes (ILs) anchored with internalizing human antibodies capable of targeting all subtypes of mesothelioma can be useful for targeted imaging and therapy of this malignant disease. The objectives of this study were to evaluate both the in vitro and in vivo tumor targeted internalization of novel internalizing human single chain antibody (scFv) anchored ILs on both epithelioid (M28) and sarcomatoid (VAMT-1) subtypes of human mesothelioma. ILs were prepared by post-insertion of mesothelioma-targeting human scFv (M1) onto preformed liposomes and radiolabeled with (111)In ((111)In-IL-M1), along with control non-targeted liposomes ((111)In-CL). Incubation of (111)In-IL-M1 with M28, VAMT-1, and a control non-tumorigenic cell line (BPH-1) at 37 °C for 24 h revealed efficient binding and rapid internalization of ILs into both subtypes of tumor cells but not into the BPH-1 cells; internalization accounted for approximately 81-94% of total cell accumulation in mesothelioma cells compared to 37-55% in control cells. In tumor-bearing mice intravenous (i.v.) injection of (111)In-IL-M1 led to remarkable tumor accumulation: 4% and 4.7% injected dose per gram (% ID/g) for M28 and VAMT-1 tumors, respectively, 48 h after injection. Furthermore, tumor uptake of (111)In-IL-M1 in live xenograft animal models was verified by single photon emission computed tomography (SPECT/CT). In contrast, i.v. injection of (111)In-CL in tumor-bearing mice revealed very low uptake in both subtypes of mesothelioma, 48 h after injection. In conclusion, M1 scFv-anchored ILs showed selective tumor targeting and rapid internalization into both epithelioid and sarcomatoid subtypes of human mesothelioma, demonstrating its potential as a promising vector for enhanced tumor drug targeting.


PLOS ONE | 2010

Targeted In Vivo Extracellular Matrix Formation Promotes Neovascularization in a Rodent Model of Myocardial Infarction

Shirley S. Mihardja; Dong-Wei Gao; Richard E. Sievers; Qizhi Fang; Jinjin Feng; Jianming Wang; Henry F. VanBrocklin; James W. Larrick; Manley Huang; Michael W. Dae; Randall J. Lee

Background The extracellular matrix plays an important role in tissue regeneration. We investigated whether extracellular matrix protein fragments could be targeted with antibodies to ischemically injured myocardium to promote angiogenesis and myocardial repair. Methodology/Principal Findings Four peptides, 2 derived from fibronectin and 2 derived from Type IV Collagen, were assessed for in vitro and in vivo tendencies for angiogenesis. Three of the four peptides—Hep I, Hep III, RGD—were identified and shown to increase endothelial cell attachment, proliferation, migration and cell activation in vitro. By chemically conjugating these peptides to an anti-myosin heavy chain antibody, the peptides could be administered intravenously and specifically targeted to the site of the myocardial infarction. When administered into Sprague-Dawley rats that underwent ischemia-reperfusion myocardial infarction, these peptides produced statistically significantly higher levels of angiogenesis and arteriogenesis 6 weeks post treatment. Conclusions/Significance We demonstrated that antibody-targeted ECM-derived peptides alone can be used to sufficiently alter the extracellular matrix microenvironment to induce a dramatic angiogenic response in the myocardial infarct area. Our results indicate a potentially new non-invasive strategy for repairing damaged tissue, as well as a novel tool for investigating in vivo cell biology.


Cancer Research | 2011

Novel Human Single Chain Antibody Fragments That Are Rapidly Internalizing Effectively Target Epithelioid and Sarcomatoid Mesotheliomas

Arun K. Iyer; Xiaoli Lan; Xiaodong Zhu; Yang Su; Jinjin Feng; Xiaoju Zhang; Dong-Wei Gao; Youngho Seo; Henry F. VanBrocklin; V. Courtney Broaddus; Bin Liu; Jiang He

Human antibodies targeting all subtypes of mesothelioma could be useful to image and treat this deadly disease. Here we report tumor targeting of a novel internalizing human single chain antibody fragment (scFv) labeled with (⁹⁹m)Tc ((⁹⁹m)Tc-M40) in murine models of mesothelioma of both epithelioid (M28) and sarcomatoid (VAMT-1) origins. (⁹⁹m)Tc-M40 was taken up rapidly and specifically by both subtype tumor cells in vitro, with 68% to 92% internalized within 1 hour. The specificity of binding was evidenced by blocking (up to 95%) with 10-fold excess of unlabeled M40. In animal studies, tumors of both subtypes were clearly visualized by SPECT/CT as early as 1 hour postinjection of (⁹⁹m)Tc-M40. Tumor uptake measured as percent of injected dose per gram tissue (%ID/g) at 3 hours was 4.38 and 5.84 for M28 and VAMT-1 tumors, respectively, significantly greater than all organs or tissues studied (liver, 2.62%ID/g; other organs or tissues <1.7%ID/g), except the kidneys (130.7%ID/g), giving tumor-to-blood ratios of 5:1 and 7:1 and tumor-to-muscle ratios of 45:1 and 60:1, for M28 and VAMT-1, respectively. The target-mediated uptake was confirmed by a nearly 70% reduction in tumor activity following administration of 10-fold excess of unlabeled scFv. Taken together, these results indicate that M40 can rapidly and specifically target epithelioid and sarcomatoid tumor cells, demonstrating the potential of this agent as a versatile targeting ligand for imaging and therapy of all subtypes of mesothelioma.


Cancer Research | 2010

Abstract 4411: Novel internalizing human scFvs targeting pancreatic tumor cells residing in their natural tissue microenvironment

Xiaodong Zhu; Jiang He; Xiaoju Zhang; Yue Liu; Guixi Zheng; Jinjin Feng; Yang Su; Grace E. Kim; Margaret A. Tempero; Bin Liu

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer death in the United States with a five-year survival rate of about 4%. Current diagnostic and therapeutic regimens are limited and ineffective. The goal of this project is to develop novel internalizing human antibodies that target pancreatic tumor cells residing within their natural tissue microenvironment, and possess the ability to translocate through the cell membrane to reach the inside of live tumor cells. Antibodies with these two properties, i.e., being able to target tumor cells in human tissue specimen and to penetrate live tumor cells, can in the future be used to develop minimally invasive imaging strategy for earlier detection and targeted therapy that requires efficient and specific accumulation of payloads inside tumor cells. To achieve this objective, we have selected a large naive phage antibody display library that we have recently constructed on microdissected human PDAC tissue specimens. We have identified a panel of internalizing human single chain antibodies (scFvs) that target pancreatic tumor cells in situ residing in their natural tissue microenvironment. Immunohistochemistry analysis showed that these scFvs bound to human PDAC, thereby recognizing clinically represented tumor antigens as opposed to cell line artifacts. All antibodies studied are internalized by PDAC cells in vitro. Using single photon emission computed tomography / computed tomography (SPECT/CT), we showed that technetium-labeled scFvs target human xenograft PDAC cells in vivo. Interestingly, a subset of these scFvs also target murine pancreatic tumor cell lines derived from transgenic PDAC mice, indicating that they target tumor epitopes conserved between human and mouse. Antibodies targeting tumor antigens across species would allow future imaging and therapy studies to be performed not only in xenograft but also transgenic PDAC models. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4411.


Molecular Imaging and Biology | 2009

Biodistributions of 177Lu- and 111In-labeled 7E11 antibodies to prostate-specific membrane antigen in xenograft model of prostate cancer and potential use of 111In-7E11 as a pre-therapeutic agent for 177Lu-7E11 radioimmunotherapy.

Mei-Hsiu Pan; Dong-Wei Gao; Jinjin Feng; Jiang He; Youngho Seo; John Tedesco; John G. Wolodzko; Bruce H. Hasegawa; Benjamin L. Franc


Society of Nuclear Medicine Annual Meeting Abstracts | 2010

111In-labeled immunoliposomes targeting both epithelioid and sarcomatoid mesothelioma

Arun K. Iyer; Xiaoli Lan; Jinjin Feng; Xiaodong Zhu; Yang Su; Yue Liu; Henry VanBrocklin; Bin Liu; Jiang He


Society of Nuclear Medicine Annual Meeting Abstracts | 2007

Biodistribution of an activatable cell-penetrating SPECT imaging probe for prostate cancer in mice study

Mei Hsiu Pan; Ella F. Jones; Gregory Watkins; Jinjin Feng; Jiang He; M. Scott Shell; Mustafa Janabi; Henry F. VanBrocklin; Thomas F. Budinger; Rajesh Shinde; Ken A. Dill; Christopher H. Contag; Benjamin L. Franc


Society of Nuclear Medicine Annual Meeting Abstracts | 2013

Effects of size and targeting ligand on biodistribution of liposome nanoparticles in tumor mice

Jinjin Feng; Arun K. Iyer; Youngho Seo; Curtney Broaddus; Bin Liu; Henry VanBrocklin; Jiang He

Collaboration


Dive into the Jinjin Feng's collaboration.

Top Co-Authors

Avatar

Jiang He

University of California

View shared research outputs
Top Co-Authors

Avatar

Bin Liu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dong-Wei Gao

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiaodong Zhu

University of California

View shared research outputs
Top Co-Authors

Avatar

Yang Su

University of California

View shared research outputs
Top Co-Authors

Avatar

Youngho Seo

University of California

View shared research outputs
Top Co-Authors

Avatar

Xiaoli Lan

University of California

View shared research outputs
Top Co-Authors

Avatar

Yue Liu

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge