Network


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

Hotspot


Dive into the research topics where Edward K. Fung is active.

Publication


Featured researches published by Edward K. Fung.


The Journal of Nuclear Medicine | 2017

Curative multi-cycle radioimmunotherapy monitored by quantitative SPECT/CT-based theranostics, using bispecific antibody pretargeting strategy in colorectal cancer

Sarah M. Cheal; Edward K. Fung; Miteshkumar Patel; Blesida Punzalan; Hong Xu; Hongfen Guo; Pat Zanzonico; Sebastien Monette; Karl Dane Wittrup; Nai-Kong Cheung; Steven M. Larson

Radioimmunotherapy of solid tumors using antibody-targeted radionuclides has been limited by low therapeutic indices (TIs). We recently reported a novel 3-step pretargeted radioimmunotherapy (PRIT) strategy based on a glycoprotein A33 (GPA33)–targeting bispecific antibody and a small-molecule radioactive hapten, a complex of 177Lu and S-2-(4-aminobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (177Lu-DOTA-Bn), that leads to high TIs for radiosensitive tissues such as blood (TI = 73) and kidney (TI = 12). We tested our hypothesis that a fractionated anti-GPA33 DOTA-PRIT regimen calibrated to deliver a radiation absorbed dose to tumor of more than 100 Gy would lead to a high probability of tumor cure while being well tolerated by nude mice bearing subcutaneous GPA33-positive SW1222 xenografts. Methods: We treated groups of nude mice bearing 7-d-old SW1222 xenografts with a fractionated 3-cycle anti-GPA33 DOTA-PRIT regimen (total administered 177Lu-DOTA-Bn activity, 167 MBq/mouse; estimated radiation absorbed dose to tumor, 110 Gy). In randomly selected mice undergoing treatment, serial SPECT/CT imaging was used to monitor treatment response and calculate radiation absorbed doses to tumor. Necropsy was done on surviving animals 100–200 d after treatment to determine frequency of cure and assess select normal tissues for treatment-related histopathologies. Results: Rapid exponential tumor progression was observed in control treatment groups (i.e., no treatment or 177Lu-DOTA-Bn only), leading to euthanasia due to excessive tumor burden, whereas 10 of 10 complete responses were observed for the DOTA-PRIT–treated animals within 30 d. Treatment was well tolerated, and 100% histologic cure was achieved in 9 of 9 assessable animals without detectable radiation damage to critical organs, including bone marrow and kidney. Radiation absorbed doses to tumor derived from SPECT/CT (102 Gy) and from biodistribution (110 Gy) agreed to within 6.9%. Of the total dose of approximately 100 Gy, the first dose contributes 30%, the second dose 60%, and the third dose 10%. Conclusion: In a GPA33-positive human colorectal cancer xenograft mouse model, we validated a SPECT/CT-based theranostic PRIT regimen that led to 100% complete responses and 100% cures without any treatment-related toxicities, based on high TIs for radiosensitive tissues. These studies support the view that anti-GPA33 DOTA-PRIT will be a potent radioimmunotherapy regimen for GPA33-positive colorectal cancer tumors in humans.


Theranostics | 2018

Theranostic pretargeted radioimmunotherapy of internalizing solid tumor antigens in human tumor xenografts in mice: Curative treatment of HER2-positive breast carcinoma

Sarah M. Cheal; Hong Xu; Hongfen Guo; Mitesh Patel; Blesida Punzalan; Edward K. Fung; Sang-gyu Lee; Meghan Bell; Manisha Singh; Achim A. Jungbluth; Pat Zanzonico; Alessandra Piersigilli; Steven M. Larson; Nai-Kong V. Cheung

In recent reports, we have shown that optimized pretargeted radioimmunotherapy (PRIT) based on molecularly engineered antibody conjugates and 177Lu-DOTA chelate (DOTA-PRIT) can be used to cure mice bearing human solid tumor xenografts using antitumor antibodies to minimally internalizing membrane antigens, GPA33 (colon) and GD2 (neuroblastoma). However, many solid tumor membrane antigens are internalized after antibody binding and it is generally believed that internalizing tumor membrane antigens are not suitable targets for PRIT. In this study, we tested the hypothesis that DOTA-PRIT can be performed successfully to target HER2, an internalizing membrane antigen widely expressed in breast, ovarian, and gastroesophageal junction cancers. Methods: DOTA-PRIT was carried out in athymic nude mice bearing BT-474 xenografts, a HER2-expressing human breast cancer, using a three-step dosing regimen consisting of sequential intravenous administrations of: 1) a bispecific IgG-scFv (210 kD) format (BsAb) carrying the IgG sequence of the anti-HER2 antibody trastuzumab and the scFv “C825” with high-affinity, hapten-binding antibody for Bn-DOTA (metal) (BsAb: anti-HER2-C825), 2) a 500 kD dextran-based clearing agent, followed by 3) 177Lu-DOTA-Bn. At the time of treatment, athymic nude mice bearing established subcutaneous BT-474 tumors (medium- and smaller-sized tumors with tumor volumes of 209 ± 101 mm3 and ranging from palpable to 30 mm3, respectively), were studied along with controls. We studied single- and multi-dose regimens. For groups receiving fractionated treatment, we verified quantitative tumor targeting during each treatment cycle using non-invasive imaging with single-photon emission computed tomography/computed tomography (SPECT/CT). Results: We achieved high therapeutic indices (TI, the ratio of radiation-absorbed dose in tumor to radiation-absorbed dose to critical organs, such as bone marrow) for targeting in blood (TI = 28) and kidney (TI = 7), while delivering average radiation-absorbed doses of 39.9 cGy/MBq to tumor. Based on dosimetry estimates, we implemented a curative fractionated therapeutic regimen for medium-sized tumors that would deliver approximately 70 Gy to tumors, which required treatment with a total of 167 MBq 177Lu-DOTA-Bn/mouse (estimated absorbed tumor dose: 66 Gy). This regimen was well tolerated and achieved 100% complete responses (CRs; defined herein as tumor volume equal to or smaller than 4.2 mm3), including 62.5% histologic cure (5/8) and 37.5% microscopic residual disease (3/8) at 85 days (d). Treatment controls showed tumor progression to 207 ± 201% of pre-treatment volume at 85 d and no CRs. Finally, we show that treatment with this curative 177Lu regimen leads to a very low incidence of histopathologic abnormalities in critical organs such as bone marrow and kidney among survivors compared with non-treated controls. Conclusion: Contrary to popular belief, we demonstrate that DOTA-PRIT can be successfully adapted to an internalizing antigen-antibody system such as HER2, with sufficient TIs and absorbed tumor doses to achieve a high probability of cures of established human breast cancer xenografts while sparing critical organs of significant radiotoxicity.


Molecular Pharmaceutics | 2018

ImmunoPET Imaging of Endogenous and Transfected Prolactin Receptor Tumor Xenografts

Sarah M. Cheal; Shutian Ruan; Darren R. Veach; Valerie A. Longo; Blesida Punzalan; Jiong Wu; Edward K. Fung; Marcus P. Kelly; Jessica R. Kirshner; Jason T. Giurleo; George Ehrlich; Amy Qi Han; Gavin Thurston; William C. Olson; Pat Zanzonico; Steven M. Larson; Jorge A. Carrasquillo

Antibodies labeled with positron-emitting isotopes have been used for tumor detection, predicting which patients may respond to tumor antigen-directed therapy, and assessing pharmacodynamic effects of drug interventions. Prolactin receptor (PRLR) is overexpressed in breast and prostate cancers and is a new target for cancer therapy. We evaluated REGN2878, an anti-PRLR monoclonal antibody, as an immunoPET reagent. REGN2878 was labeled with Zr-89 after conjugation with desferrioxamine B or labeled with I-131/I-124. In vitro determination of the half-maximal inhibitory concentration (IC50) of parental REGN2878, DFO-REGN2878, and iodinated REGN2878 was performed by examining the effect of the increasing amounts of these on uptake of trace-labeled I-131 REGN2878. REGN1932, a non-PRLR binding antibody, was used as a control. Imaging and biodistribution studies were performed in mice bearing tumor xenografts with various expression levels of PRLR, including MCF-7, transfected MCF-7/PRLR, PC3, and transfected PC3/PRLR and T4D7v11 cell lines. The specificity of uptake in tumors was evaluated by comparing Zr-89 REGN2878 and REGN1932, and in vivo competition compared Zr-89 REGN2878 uptake in tumor xenografts with and without prior injection of 2 mg of nonradioactive REGN2878. The competition binding assay of DFO-REGN2878 at ratios of 3.53-5.77 DFO per antibody showed IC50 values of 0.4917 and 0.7136 nM, respectively, compared to 0.3455 nM for parental REGN2878 and 0.3343 nM for I-124 REGN2878. Imaging and biodistribution studies showed excellent targeting of Zr-89 REGN2878 in PRLR-positive xenografts at delayed times of 189 h (presented as mean ± 1 SD, percent injected activity per mL (%IA/mL) 74.6 ± 33.8%IA/mL). In contrast, MCF-7/PRLR tumor xenografts showed a low uptake (7.0 ± 2.3%IA/mL) of control Zr-89 REGN1932 and a very low uptake and rapid clearance of I-124 REGN2878 (1.4 ± 0.6%IA/mL). Zr-89 REGN2878 has excellent antigen-specific targeting in various PRLR tumor xenograft models. We estimated, using image-based kinetic modeling, that PRLR antigen has a very rapid in vivo turnover half-life of ∼14 min from the cell membrane. Despite relatively modest estimated tumor PRLR expression numbers, PRLR-expressing cells have shown final retention of the Zr-89 REGN2878 antibody, with an uptake that appeared to be related to PRLR expression. This reagent has the potential to be used in clinical trials targeting PRLR.


Medical Physics | 2014

TU‐F‐12A‐01: Quantitative Non‐Linear Compartment Modeling of 89Zr‐ and 124I‐ Labeled J591 Monoclonal Antibody Kinetics Using Serial Non‐Invasive Positron Emission Tomography Imaging in a Pre‐Clinical Human Prostate Cancer Mouse Model

Edward K. Fung; Sarah M. Cheal; Sandhya Chalasani; Shoaib B. Fareedy; B Otto; Blesida Punzalan; John L. Humm; Neil H. Bander; Joseph R. Osborne; S. M. Larson; Pat Zanzonico

PURPOSE To examine the binding kinetics of human IgG monoclonal antibody J591 which targets prostate-specific membrane antigen (PSMA) in a pre-clinical mouse cancer model using quantitative PET compartmental analysis of two radiolabeled variants. METHODS PSMA is expressed in normal human prostate, and becomes highly upregulated in prostate cancer, making it a promising therapeutic target. Two forms of J591, radiolabeled with either 8 9 Zr or 12 4 I, were prepared. 8 9 Zr is a radiometal that becomes trapped in the cell upon internalization by the antigen-antibody complex, while radioiodine leaves the cell. Mice with prostate cancer xenografts underwent non-invasive serial imaging on a Focus 120 microPET up to 144 hours post-injection of J591. A non-linear compartmental model describing the binding and internalization of antibody in tumor xenograft was developed and applied to the PET-derived time-activity curves. The antibody-antigen association rate constant (ka), total amount of antigen per gram tumor (Ag_total), internalization rate of antibody-antigen complex, and efflux rate of radioisotope from tumor were fitted using the model. The surface-bound and the internalized activity were also estimated. RESULTS Values for ka, Ag_total, and internalization rate were found to be similar regardless of radiolabel payload used. The efflux rate, however, was ∼ 9-fold higher for 12 4 I-J591 than for 8 9 Zr-J591. Time-dependent surface-bound and internalized radiotracer activity were similar for both radiolabels at early times post-injection, but clearly differed beyond 24 hours. CONCLUSION Binding and internalization of J591 to PSMA-expressing tumor xenografts were similar when radiolabeled with either 8 9 Zr or 12 4 I payload. The difference in efflux of radioactivity from tumor may be attributable to differential biological fate intracellularly of the radioisotopes. This has great significance for radioimmunotherapy and antibody-drug conjugates. Further exploration using the model will examine binding and radioisotope residence as antibody dose is increased to antigen saturation. The Center for Targeted Radioimmunotherapy and Theranostics, Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center (MSK), NIH (R25-CA096945). Technical services provided by the MSK Small-Animal Imaging Core Facility were supported by the NIH (R24-CA83084, P30-CA08748, and P50-CA92629; Zanzonico). NCI, Center to Reduce Cancer Health Disparity (R21 CA153177-03; Osborne).


European Journal of Nuclear Medicine and Molecular Imaging | 2016

Theranostic pretargeted radioimmunotherapy of colorectal cancer xenografts in mice using picomolar affinity ⁸⁶Y- or ¹⁷⁷Lu-DOTA-Bn binding scFv C825/GPA33 IgG bispecific immunoconjugates.

Sarah M. Cheal; Hong Xu; Hongfen Guo; Sang-gyu Lee; Blesida Punzalan; Sandhya Chalasani; Edward K. Fung; Achim A. Jungbluth; Pat Zanzonico; Jorge A. Carrasquillo; Joseph O'Donoghue; Peter Smith-Jones; Karl Dane Wittrup; Nai-Kong Cheung; S. M. Larson


EJNMMI research | 2016

Targeting of radiolabeled J591 antibody to PSMA-expressing tumors: optimization of imaging and therapy based on non-linear compartmental modeling

Edward K. Fung; Sarah M. Cheal; Shoaib B. Fareedy; Blesida Punzalan; Volkan Beylergil; Jawaria Amir; Sandhya Chalasani; Wolfgang A. Weber; Daniel E. Spratt; Darren R. Veach; Neil H. Bander; Steven M. Larson; Pat Zanzonico; Joseph R. Osborne


Archive | 2016

Systems and methods for determining optimum patient-specific antibody dose for tumor targeting

Pat Zanzonico; Sarah M. Cheal; Steven M. Larson; Joseph R. Osborne; Edward K. Fung; David Ulmert


The Journal of Nuclear Medicine | 2018

Pretargeted radioimmunotherapy with 225Ac-proteus-DOTA hapten.

Sarah M. Cheal; Michael R. McDevitt; Miteshkumar Patel; Guangbin Yang; Nagavarakishore Pillarsetty; Hong Xu; Hongfen Guo; Edward K. Fung; Nai-Kong Cheung; Ouathek Ouerfelli; Steven M. Larson


The Journal of Nuclear Medicine | 2016

Serial dynamic and static preclinical PET/CT image-based immunokinetic modeling of 86Y-hapten localization in HER2-expressing SKOV3 xenograft during anti-HER2/antilanthanide DOTA pretargeted radioimmunotherapy

Edward K. Fung; Sarah M. Cheal; Pat Zanzonico; Steven M. Larson


The Journal of Nuclear Medicine | 2016

Curative theranostic pretargeted radioimmunotherapy (PRIT) of GPA33-expressing colorectal cancer (CRC) using a bispecific antibody with high affinity for GPA33 and DOTA (D) metal complex (DPRIT).

Sarah M. Cheal; Hong Xu; Hongfen Guo; Miteshkumar Patel; Blesida Punzalan; Edward K. Fung; Pat Zanzonico; K. Dane Wittrup; Nai-Kong Cheung; Steven M. Larson

Collaboration


Dive into the Edward K. Fung's collaboration.

Top Co-Authors

Avatar

Sarah M. Cheal

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Steven M. Larson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Blesida Punzalan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hong Xu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hongfen Guo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pat Zanzonico

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nai-Kong Cheung

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sandhya Chalasani

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge A. Carrasquillo

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Joseph R. Osborne

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge