39 results on '"Yung Hua Wang"'
Search Results
2. Defining the Magnetic Resonance Features of Renal Lesions and Their Response to Everolimus in a Transgenic Mouse Model of Tuberous Sclerosis Complex
- Author
-
Shubhangi Agarwal, Emilie Decavel-Bueff, Yung-Hua Wang, Hecong Qin, Romelyn Delos Santos, Michael J. Evans, and Renuka Sriram
- Subjects
TSC ,kidney ,everolimus ,mTOR ,MRI ,mp-MRI ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Tuberous sclerosis complex (TSC) is an inherited genetic disorder characterized by mutations in TSC1 or TSC2 class of tumor suppressers which impact several organs including the kidney. The renal manifestations are usually in the form of angiomyolipoma (AML, in 80% of the cases) and cystadenomas. mTOR inhibitors such as rapamycin and everolimus have shown efficacy in reducing the renal tumor burden. Early treatment prevents the progression of AML; however, the tumors regrow upon cessation of therapy implying a lifelong need for monitoring and management of this morbid disease. There is a critical need for development of imaging strategies to monitor response to therapy and progression of disease which will also facilitate development of newer targeted therapy. In this study we evaluated the potential of multiparametric 1H magnetic resonance imaging (mpMRI) to monitor tumor response to therapy in a preclinical model of TSC, the transgenic mouse A/J Tsc2+/-. We found 2-dimensional T2-weighted sequence with 0.5 mm slice thickness to be optimal for detecting renal lesions as small as 0.016 mm3. Baseline characterization of lesions with MRI to assess physiological parameters such as cellularity and perfusion is critical for distinguishing between cystic and solid lesions. Everolimus treatment for three weeks maintained tumor growth at 36% from baseline, while control tumors displayed steady growth and were 70% larger than baseline at the end of therapy. Apparent diffusion coefficient, T1 values and normalized T2 intensity changes were also indictive of response to treatment. Our results indicate that standardization and implementation of improved MR imaging protocols will significantly enhance the utility of mpMRI in determining the severity and composition of renal lesions for better treatment planning.
- Published
- 2022
- Full Text
- View/download PDF
3. Targeting a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) for RAS-driven cancers
- Author
-
Shion A. Lim, Jie Zhou, Alexander J. Martinko, Yung-Hua Wang, Ekaterina V. Filippova, Veronica Steri, Donghui Wang, Soumya G. Remesh, Jia Liu, Byron Hann, Anthony A. Kossiakoff, Michael J. Evans, Kevin K. Leung, and James A. Wells
- Subjects
Therapeutics ,Medicine - Abstract
Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal “AND” gate for improving the therapeutic index.
- Published
- 2022
- Full Text
- View/download PDF
4. Measuring Dynamic Changes in the Labile Iron Pool in Vivo with a Reactivity-Based Probe for Positron Emission Tomography
- Author
-
Ryan K. Muir, Ning Zhao, Junnian Wei, Yung-hua Wang, Anna Moroz, Yangjie Huang, Ying-Chu Chen, Renuka Sriram, John Kurhanewicz, Davide Ruggero, Adam R. Renslo, and Michael J. Evans
- Subjects
Chemistry ,QD1-999 - Published
- 2019
- Full Text
- View/download PDF
5. Supplementary Table from CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease
- Author
-
Michael J. Evans, Rahul Aggarwal, James A. Wells, Jonathan Chou, Eric J. Small, Felix Y. Feng, Youngho Seo, Emily Chan, Renuka Sriram, Adam Foye, Li Zhang, Jun Zhu, Emily A. Egusa, Kevin K. Leung, Shion A. Lim, Jie Zhou, Hyunjung Kim, Nima Hooshdaran, Sasank Sakhamuri, Yung-Hua Wang, Kai Trepka, Shalini Chopra, and Ning Zhao
- Abstract
Supplementary Table from CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease
- Published
- 2023
6. Data from Theranostic Targeting of CUB Domain Containing Protein 1 (CDCP1) in Pancreatic Cancer
- Author
-
Michael J. Evans, James A. Wells, Kimberly S. Kirkwood, Eric A. Collisson, Kevin K. Leung, Sean Carlin, Youngho Seo, Lydia H. Zhang, Shion A. Lim, Jie Zhuo, Alexander J. Martinko, Zhuo Chen, Yangjie Huang, Ning Zhao, Junnian Wei, Jeremy M. Sharib, Yung-Hua Wang, and Anna Moroz
- Abstract
Purpose:The recent emergence of radioligand therapies for cancer treatment has increased enthusiasm for developing new theranostic strategies coupling both imaging and cytotoxicity in the same entity. In this study, we evaluated whether CUB domain containing protein 1 (CDCP1), a single-pass transmembrane protein highly overexpressed in diverse human cancers, might be a target for cancer theranostics.Experimental Design:The ectodomain of CDCP1 was targeted using radiolabeled forms of 4A06, a potent and specific recombinant human antibody that we developed. Imaging and antitumor assessment studies were performed in animal models of pancreatic cancer, including two patient-derived xenograft models we developed for this study. For antitumor assessment studies, the endpoints were death due to tumor volume >3,000 mm3 or ≥20% loss in body weight. Specific tracer binding or antitumor effects were assessed with an unpaired, two-tailed Student t test and survival advantages were assessed with a log rank (Mantel–Cox) test. Differences at the 95% confidence level were interpreted to be significant.Results:89Zr-4A06 detected a broad dynamic range of full length or cleaved CDCP1 expression on seven human pancreatic cancer tumors (n = 4/tumor). Treating mice with single or fractionated doses of 177Lu-4A06 significantly reduced pancreatic cancer tumor volume compared with mice receiving vehicle or unlabeled 4A06 (n = 8; P < 0.01). A single dose of 225Ac-4A06 also inhibited tumor growth, although the effect was less profound compared with 177Lu-4A06 (n = 8; P < 0.01). A significant survival advantage was imparted by 225Ac-4A06 (HR = 2.56; P < 0.05).Conclusions:These data establish that CDCP1 can be exploited for theranostics, a finding with widespread implications given its breadth of overexpression in cancer.
- Published
- 2023
7. Data from Molecular Imaging of Prostate Cancer Targeting CD46 Using ImmunoPET
- Author
-
Jiang He, Robert R. Flavell, Bin Liu, Youngho Seo, Michael J. Evans, Rahul Aggarwal, Felix Y. Feng, Jonathan Chou, Emily A. Egusa, Renuka Sriram, Robert Dreicer, Daniel Gioeli, Sui Shen, Fujun Qin, Yangjie Huang, Yung-hua Wang, Ning Zhao, Tony L. Huynh, Mayuri Jayaraman, Walter Zhao, Denis R. Beckford-Vera, Yang Su, Jun Hua, Jun Li, and Sinan Wang
- Abstract
Purpose:We recently identified CD46 as a novel therapeutic target in prostate cancer. In this study, we developed a CD46-targeted PET radiopharmaceutical, [89Zr]DFO-YS5, and evaluated its performance for immunoPET imaging in murine prostate cancer models.Experimental Design:[89Zr]DFO-YS5 was prepared and its in vitro binding affinity for CD46 was measured. ImmunoPET imaging was conducted in male athymic nu/nu mice bearing DU145 [AR−, CD46+, prostate-specific membrane antigen–negative (PSMA−)] or 22Rv1 (AR+, CD46+, PSMA+) tumors, and in NOD/SCID gamma mice bearing patient-derived adenocarcinoma xenograft, LTL-331, and neuroendocrine prostate cancers, LTL-331R and LTL-545.Results:[89Zr]DFO-YS5 binds specifically to the CD46-positive human prostate cancer DU145 and 22Rv1 xenografts. In biodistribution studies, the tumor uptake of [89Zr]DFO-YS5 was 13.3 ± 3.9 and 11.2 ± 2.5 %ID/g, respectively, in DU145 and 22Rv1 xenografts, 4 days postinjection. Notably, [89Zr]DFO-YS5 demonstrated specific uptake in the PSMA- and AR-negative DU145 model. [89Zr]DFO-YS5 also showed uptake in the patient-derived LTL-331 and -331R models, with particularly high uptake in the LTL-545 neuroendocrine prostate cancer tumors (18.8 ± 5.3, 12.5 ± 1.8, and 32 ± 5.3 %ID/g in LTL-331, LTL-331R, and LTL-545, respectively, at 4 days postinjection).Conclusions:[89Zr]DFO-YS5 is an excellent PET imaging agent across a panel of prostate cancer models, including in both adenocarcinoma and neuroendocrine prostate cancer, both cell line- and patient-derived xenografts, and both PSMA-positive and -negative tumors. It demonstrates potential for clinical translation as an imaging agent, theranostic platform, and companion biomarker in prostate cancer.
- Published
- 2023
8. Supplementary Figure from CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease
- Author
-
Michael J. Evans, Rahul Aggarwal, James A. Wells, Jonathan Chou, Eric J. Small, Felix Y. Feng, Youngho Seo, Emily Chan, Renuka Sriram, Adam Foye, Li Zhang, Jun Zhu, Emily A. Egusa, Kevin K. Leung, Shion A. Lim, Jie Zhou, Hyunjung Kim, Nima Hooshdaran, Sasank Sakhamuri, Yung-Hua Wang, Kai Trepka, Shalini Chopra, and Ning Zhao
- Abstract
Supplementary Figure from CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease
- Published
- 2023
9. Data from CUB Domain-Containing Protein 1 (CDCP1) Is a Target for Radioligand Therapy in Castration-Resistant Prostate Cancer, including PSMA Null Disease
- Author
-
Michael J. Evans, Rahul Aggarwal, James A. Wells, Jonathan Chou, Eric J. Small, Felix Y. Feng, Youngho Seo, Emily Chan, Renuka Sriram, Adam Foye, Li Zhang, Jun Zhu, Emily A. Egusa, Kevin K. Leung, Shion A. Lim, Jie Zhou, Hyunjung Kim, Nima Hooshdaran, Sasank Sakhamuri, Yung-Hua Wang, Kai Trepka, Shalini Chopra, and Ning Zhao
- Abstract
Purpose:With the improvement in overall survival with 177Lu-PSMA 617, radioligand therapy (RLT) is now a viable option for patients with metastatic castration-resistant prostate cancer (mCRPC). However, responses are variable, in part due to low PSMA expression in 30% of patients. Herein, we evaluated whether the cell surface protein CUB domain-containing protein 1 (CDCP1) can be exploited to treat mCRPC with RLT, including in PSMA-low subsets.Experimental Design:CDCP1 levels were evaluated using RNA sequencing from 119 mCRPC biopsies. CDCP1 levels were assessed in 17 post–enzalutamide- or abiraterone-treated mCRPC biopsies, 12 patient-derived xenografts (PDX), and prostate cancer cell lines. 4A06, a recombinant human antibody that targets the CDCP1 ectodomain, was labeled with Zr-89 or Lu-177 and tested in tumor-bearing mice.Results:CDCP1 expression was observed in 90% of mCRPC biopsies, including small-cell neuroendocrine (SCNC) and adenocarcinomas with low FOLH1 (PSMA) levels. Fifteen of 17 evaluable mCRPC biopsies (85%) demonstrated membranous CDCP1 expression, and 4 of 17 (23%) had higher CDCP1 H-scores compared with PSMA. CDCP1 was expressed in 10 of 12 PDX samples. Bmax values of approximately 22,000, 6,200, and 2,800 fmol/mg were calculated for PC3, DU145, and C4–2B human prostate cancer cells, respectively. 89Zr-4A06 PET detected six human prostate cancer xenografts, including PSMA-low tumors. 177Lu-4A06 significantly suppressed growth of DU145 and C4–2B xenografts.Conclusions:The data provide the first evidence supporting CDCP1-directed RLT to treat mCRPC. Expanded studies are warranted to determine whether CDCP1 is a viable drug target for patients with mCPRC.
- Published
- 2023
10. Figures S1-S6, Tables S1 - S3 from Theranostic Targeting of CUB Domain Containing Protein 1 (CDCP1) in Pancreatic Cancer
- Author
-
Michael J. Evans, James A. Wells, Kimberly S. Kirkwood, Eric A. Collisson, Kevin K. Leung, Sean Carlin, Youngho Seo, Lydia H. Zhang, Shion A. Lim, Jie Zhuo, Alexander J. Martinko, Zhuo Chen, Yangjie Huang, Ning Zhao, Junnian Wei, Jeremy M. Sharib, Yung-Hua Wang, and Anna Moroz
- Abstract
Supplemental methods, figures, and tables
- Published
- 2023
11. Supplementary Data from Molecular Imaging of Prostate Cancer Targeting CD46 Using ImmunoPET
- Author
-
Jiang He, Robert R. Flavell, Bin Liu, Youngho Seo, Michael J. Evans, Rahul Aggarwal, Felix Y. Feng, Jonathan Chou, Emily A. Egusa, Renuka Sriram, Robert Dreicer, Daniel Gioeli, Sui Shen, Fujun Qin, Yangjie Huang, Yung-hua Wang, Ning Zhao, Tony L. Huynh, Mayuri Jayaraman, Walter Zhao, Denis R. Beckford-Vera, Yang Su, Jun Hua, Jun Li, and Sinan Wang
- Abstract
Supplemental
- Published
- 2023
12. In Vivo Profiling with 18F-YJH08 Reveals Diverse Tissue Patterns of Antagonist/Glucocorticoid Receptor Interactions
- Author
-
Hyunjung Kim, Yangjie Huang, Ning Zhao, Yung-hua Wang, Sasank Sakhamuri, Shalini Chopra, Nima Hooshdaran, Renuka Sriram, Rahul Aggarwal, and Michael J. Evans
- Subjects
Drug Discovery ,Pharmaceutical Science ,Molecular Medicine - Published
- 2022
13. In Vivo Measurement of Granzyme Proteolysis from Activated Immune Cells with PET
- Author
-
Lawrence Fong, Yung-hua Wang, Mark R. Looney, Conner Bardine, André Luiz Lourenço, David M. Wilson, Ning Zhao, David Y. Oh, Michael J. Evans, Yangjie Huang, Simon J. Cleary, and Charles S. Craik
- Subjects
Biodistribution ,medicine.diagnostic_test ,biology ,Chemistry ,General Chemical Engineering ,Proteolysis ,Bioengineering ,General Chemistry ,GZMB ,Cell biology ,Granzyme B ,Immune system ,Granzyme ,In vivo ,Knockout mouse ,Chemical Sciences ,medicine ,biology.protein ,QD1-999 ,Research Article - Abstract
The biology of human granzymes remains enigmatic in part due to our inability to probe their functions outside of in vitro assays or animal models with divergent granzyme species. We hypothesize that the biology of human granzymes could be better elaborated with a translational imaging technology to reveal the contexts in which granzymes are secreted and biochemically active in vivo. Here, we advance toward this goal by engineering a Granzyme targeting Restricted Interaction Peptide specific to family member B (GRIP B) to measure secreted granzyme B (GZMB) biochemistry with positron emission tomography. A proteolytic cleavage of 64Cu-labeled GRIP B liberates a radiolabeled form of Temporin L, which sequesters the radioisotope by binding to adjacent phospholipid bilayers. Thus, at extended time points postinjection (i.e., hours, not seconds), tissue biodistribution of the radioisotope in vivo reflects relative units of the GZMB activity. As a proof of concept, we show in three syngeneic mouse cancer models that 64Cu-GRIP B detects GZMB from T cells activated with immune checkpoint inhibitors (CPI). Remarkably, the radiotracer detects the proteolysis within tumors but also in lymphoid tissue, where immune cells are activated by a systemic CPI. Control experiments with an uncleavable analogue of 64Cu-GRIP B and tumor imaging studies in germline GZMB knockout mice were applied to show that 64Cu-GRIP B is specific for GZMB proteolysis. Furthermore, we explored a potential noncytotoxic function for GZMB by applying 64Cu-GRIP B to a model of pulmonary inflammation. In summary, we demonstrate that granzyme biochemistry can be assessed in vivo using an imaging modality that can be scaled vertically into human subjects., A protease-activated chemosensor for in vivo molecular imaging enables the detection of activated immune cells by specifically harnessing granzyme B catalysis.
- Published
- 2021
14. The Synthesis and Structural Requirements for Measuring Glucocorticoid Receptor Expression In Vivo with (±)-11C-YJH08 PET
- Author
-
Henry F. VanBrocklin, Yung-hua Wang, Yangjie Huang, Junnian Wei, Charles Truillet, Matthew F.L. Parker, Spencer C. Behr, Robert R. Flavell, Joseph E. Blecha, Christopher R. Drake, Rahul Aggarwal, Ning Zhao, Youngho Seo, Diego Garrido-Ruiz, Michael J. Evans, Matthew P. Jacobson, and David M. Wilson
- Subjects
dosimetry study ,Biodistribution ,Stereochemistry ,Clinical Sciences ,Blood–brain barrier ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Glucocorticoid ,Rare Diseases ,0302 clinical medicine ,Glucocorticoid receptor ,Protein Domains ,In vivo ,Receptors ,glucocorticoid receptor ,medicine ,Radioligand ,Animals ,Structure–activity relationship ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Carbon Radioisotopes ,carbon-11 ,030304 developmental biology ,0303 health sciences ,Synthetic ,Chemistry Techniques ,molecular imaging ,In vitro ,Nuclear Medicine & Medical Imaging ,PET ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,carbon‐11 ,Methyl iodide - Abstract
Non-invasive methods to study glucocorticoid receptor (GR) signaling are urgently needed to reveal the complexity of GR signaling in normal physiology and human disorders, as well as to identify selective GR modulators to treat diseases. Here, we report evidence supporting translational studies with (±)-[11C]-5-(4-fluorobenzyl)-10-methoxy-2,2,4-trimethyl-2,5-dihydro-1H-chromeno[3,4-f ]-quinoline (named as (±)-[11C]YJH08), a radioligand for positron emission tomography (PET) that engages the ligand binding domain on GR. Methods: (±)-[11C]YJH08 was synthesized by reacting the phenol precursor with [11C]methyl iodide. The biodistribution was studied in vivo with PET/CT and autoradiography. A library of analogues were synthesized and studied in vitro and in vivo to understand the (±)-[11C]YJH08 structure activity relationship. Rodent dosimetry studies were performed to estimate the human equivalent doses of (±)-[11C]YJH08. Results: (±)-[11C]YJH08 was synthesized by reaction of the phenolic precursor with [11C]methyl iodide giving a radiochemical yield of 51.7 ± 4.7% (decay corrected to starting [11C]methyl iodide). An analysis of the (±)-YJH08 structure-activity relationship with molecular dynamics simulations showed that (R)- and (S)-enantiomers occupy the ligand binding domain on GR with different binding modes and have indistinguishable patterns of biodistribution in vivo. Moreover, a focused chemical screen of analogues revealed that the aryl fluoride motif on YJH08 is essential for high affinity GR binding in vitro, high tissue uptake in vivo, and passage across the blood brain barrier. Lastly, we performed dosimetry studies in rodents, from which we showed estimated human equivalent doses of (±)-[11C]YJH08 to be commensurate with widely used carbon-11 and fluorine-18 tracers. Conclusion: In summary, these studies reveal the molecular determinants of a high affinity and selectivity ligand-receptor interaction and support the use of (±)-[11C]YJH08 PET to make the first measurements of GR expression in human subjects.
- Published
- 2020
15. Switchable assembly and function of antibody complexes in vivo using a small molecule
- Author
-
Alexander J. Martinko, Erin F. Simonds, Suchitra Prasad, Alberto Ponce, Colton J. Bracken, Junnian Wei, Yung-Hua Wang, Tiffany-Lynn Chow, Zhong Huang, Michael J. Evans, James A. Wells, and Zachary B. Hill
- Subjects
Small Molecule Libraries ,Immunoconjugates ,Multidisciplinary ,Antibody Specificity ,Humans ,Antibodies - Abstract
The antigen specificity and long serum half-life of monoclonal antibodies have made them a critical part of modern therapeutics. These properties have been coopted in a number of synthetic formats, such as antibody-drug conjugates, bispecific antibodies, or Fc-fusion proteins to generate novel biologic drug modalities. Historically, these new therapies have been generated by covalently linking multiple molecular moieties through chemical or genetic methods. This irreversible fusion of different components means that the function of the molecule is static, as determined by the structure. Here, we report the development of a technology for switchable assembly of functional antibody complexes using chemically induced dimerization domains. This approach enables control of the antibody's intended function in vivo by modulating the dose of a small molecule. We demonstrate this switchable assembly across three therapeutically relevant functionalities in vivo, including localization of a radionuclide-conjugated antibody to an antigen-positive tumor, extension of a cytokine's half-life, and activation of bispecific, T cell-engaging antibodies.
- Published
- 2022
16. Quantitative and Qualitative Improvement of Low-Count [68Ga]Citrate and [90Y]Microspheres PET Image Reconstructions Using Block Sequential Regularized Expectation Maximization Algorithm
- Author
-
Youngho Seo, Mohammad Mehdi Khalighi, Yung-Hua Wang, Spencer C. Behr, Kristen A. Wangerin, Rahul Aggarwal, Robert R. Flavell, Timothy Deller, Salma Jivan, Michael J. Evans, and Maureen P. Kohi
- Subjects
Male ,Cancer Research ,Physiology ,Image quality ,Image Processing ,Gallium ,Signal-To-Noise Ratio ,Castration-Resistant ,Phantoms ,Imaging ,030218 nuclear medicine & medical imaging ,Computer-Assisted ,0302 clinical medicine ,Positron Emission Tomography Computed Tomography ,Image noise ,Yttrium Radioisotopes ,Citrates ,Positron emission ,Image resolution ,Mathematics ,screening and diagnosis ,medicine.diagnostic_test ,[90Y]microspheres ,[68Ga]citrate ,Reconstruction algorithm ,Microspheres ,Low-count PET ,Detection ,Nuclear Medicine & Medical Imaging ,Oncology ,Positron emission tomography ,Biomedical Imaging ,[Y-90]microspheres ,Algorithms ,4.2 Evaluation of markers and technologies ,MRI ,Clinical Sciences ,Gallium Radioisotopes ,Bioengineering ,Imaging phantom ,03 medical and health sciences ,Regularized EM ,Expectation–maximization algorithm ,BSREM ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,business.industry ,[Ga-68]citrate ,Prostatic Neoplasms ,Pattern recognition ,PET ,PET/MRI ,Artificial intelligence ,Radiopharmaceuticals ,business - Abstract
PurposeThere are several important positron emission tomography (PET) imaging scenarios that require imaging with very low photon statistics, for which both quantitative accuracy and visual quality should not be neglected. For example, PET imaging with the low photon statistics is closely related to active efforts to significantly reduce radiation exposure from radiopharmaceuticals. We investigated two examples of low-count PET imaging: (a) imaging [90Y]microsphere radioembolization that suffers the very small positron emission fraction of Y-90's decay processes, and (b) cancer imaging with [68Ga]citrate with uptake time of 3-4 half-lives, necessary for visualizing tumors. In particular, we investigated a type of penalized likelihood reconstruction algorithm, block sequential regularized expectation maximization (BSREM), for improving both image quality and quantitative accuracy of these low-count PET imaging cases.ProceduresThe NEMA/IEC Body phantom filled with aqueous solution of Y-90 or Ga-68 was scanned to mimic the low-count scenarios of corresponding patient data acquisitions on a time-of-flight (TOF) PET/magnetic resonance imaging system. Contrast recovery, background variation, and signal-to-noise ratio were evaluated in different sets of count densities using both conventional TOF ordered subset expectation (TOF-OSEM) and TOF-BSREM algorithms. The regularization parameter, beta, in BSREM that controls the tradeoff between image noise and resolution was evaluated to find a value for improved confidence in image interpretation. Visual quality assessment of the images obtained from patients administered with [68Ga]citrate (n = 6) was performed. We also made preliminary visual image quality assessment for one patient with [90Y]microspheres. In Y-90 imaging, the effect of 511-keV energy window selection for minimizing the number of random events was also evaluated.ResultsQuantitatively, phantom images reconstructed with TOF-BSREM showed improved contrast recovery, background variation, and signal-to-noise ratio values over images reconstructed with TOF-OSEM. Both phantom and patient studies of delayed imaging of [68Ga]citrate show that TOF-BSREM with beta = 500 gives the best tradeoff between image noise and image resolution based on visual assessment by the readers. The NEMA-IQ phantom study with [90Y]microspheres shows that the narrow energy window (460-562keV) recovers activity concentrations in small spheres better than the regular energy window (425-650keV) with the beta value of 2000 using the TOF-BSREM algorithm. For the images obtained from patients with [68Ga]citrate using TOF-BSREM with beta = 500, the visual analogue scale (VAS) was improved by 17% and the Likert score was increased by 1 point on average, both in comparison to corresponding scores for images reconstructed using TOF-OSEM.ConclusionOur investigation shows that the TOF-BSREM algorithm improves the image quality and quantitative accuracy in low-count PET imaging scenarios. However, the beta value in this algorithm needed to be adjusted for each radiopharmaceutical and counting statistics at the time of scans.
- Published
- 2019
17. Tumor-conditional anti-CTLA4 uncouples antitumor efficacy from immunotherapy-related toxicity
- Author
-
Xiaoqing Lu, Pete Bousquet, Paul L. Richardson, Grace Lynch, Lawrence Fong, John Mankovich, Dave Banach, Wendy Ritacco, Donald M. Simons, Susan V. Westmoreland, Chien-Chun Steven Pai, Mingyi Chen, Anthony Chang, Michael J. Evans, Junnian Wei, Chanhyuk Park, Diana Bowley, Jiaxi Wang, Yung Hua Wang, Gillian Kingsbury, John C. Huang, Feng Dong, Christine Beam, Soumya Mitra, and Jane Seagal
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,Cancer immunotherapy ,T-Lymphocytes, Regulatory ,Medical and Health Sciences ,Mice ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Neoplasms ,Tumor Microenvironment ,Cytotoxic T cell ,CTLA-4 Antigen ,Cancer ,Mice, Knockout ,Antibody-dependent cell-mediated cytotoxicity ,Immunity, Cellular ,Tumor ,General Medicine ,Regulatory ,Immunological ,Oncology ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Immunotherapy ,Development of treatments and therapeutic interventions ,Research Article ,Knockout ,Immunology ,Antineoplastic Agents ,chemical and pharmacologic phenomena ,Cell Line ,Vaccine Related ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Immunologic Factors ,Animals ,Humans ,Tumor microenvironment ,business.industry ,Inflammatory and immune system ,Immunity ,Immune checkpoint ,Blockade ,HEK293 Cells ,030104 developmental biology ,Commentary ,Cancer research ,Immunization ,Cellular ,Digestive Diseases ,business ,CD8 ,Single-Chain Antibodies - Abstract
While immune checkpoint blockade leads to potent antitumor efficacy, it also leads to immune-related adverse events in cancer patients. These toxicities stem from systemic immune activation resulting in inflammation of multiple organs, including the gastrointestinal tract, lung, and endocrine organs. We developed a dual variable domain immunoglobulin of anti-CTLA4 antibody (anti-CTLA4 DVD, where CTLA4 is defined as cytotoxic T lymphocyte–associated antigen-4) possessing an outer tumor-specific antigen-binding site engineered to shield the inner anti-CTLA4–binding domain. Upon reaching the tumor, the outer domain was cleaved by membrane type-serine protease 1 (MT-SP1) present in the tumor microenvironment, leading to enhanced localization of CTLA4 blockade. Anti-CTLA4 DVD markedly reduced multiorgan immune toxicity by preserving tissue-resident Tregs in Rag 1(–/–) mice that received naive donor CD4(+) T cells from WT C57BL/6j mice. Moreover, anti-CTLA4 DVD induced potent antitumor effects by decreasing tumor-infiltrating Tregs and increasing the infiltration of antigen-specific CD8(+) T lymphocytes in TRAMP-C2–bearing C57BL/6j mice. Treg depletion was mediated through the antibody-dependent cellular cytotoxicity (ADCC) mechanism, as anti-CTLA4 without the FcγR-binding portion (anti-CTLA4 DANA) spared Tregs, preventing treatment-induced toxicities. In summary, our results demonstrate an approach to anti-CTLA4 blockade that depletes tumor-infiltrating, but not tissue-resident, Tregs, preserving antitumor effects while minimizing toxicity. Thus, our tumor-conditional anti-CTLA4 DVD provides an avenue for uncoupling antitumor efficacy from immunotherapy-induced toxicities.
- Published
- 2018
18. Ferrous iron-activatable drug conjugate achieves potent MAPK blockade in KRAS-driven tumors
- Author
-
Honglin Jiang, Ryan K. Muir, Ryan L. Gonciarz, Adam B. Olshen, Iwei Yeh, Byron C. Hann, Ning Zhao, Yung-hua Wang, Spencer C. Behr, James E. Korkola, Michael J. Evans, Eric A. Collisson, and Adam R. Renslo
- Subjects
Proto-Oncogene Proteins p21(ras) ,Cell Line, Tumor ,Iron ,Neoplasms ,Immunology ,Mutation ,Immunology and Allergy ,Animals ,Protein Kinase Inhibitors ,Signal Transduction - Abstract
KRAS mutations drive a quarter of cancer mortality, and most are undruggable. Several inhibitors of the MAPK pathway are FDA approved but poorly tolerated at the doses needed to adequately extinguish RAS/RAF/MAPK signaling in the tumor cell. We found that oncogenic KRAS signaling induced ferrous iron (Fe2+) accumulation early in and throughout mutant KRAS-mediated transformation. We converted an FDA-approved MEK inhibitor into a ferrous iron–activatable drug conjugate (FeADC) and achieved potent MAPK blockade in tumor cells while sparing normal tissues. This innovation allowed sustainable, effective treatment of tumor-bearing animals, with tumor-selective drug activation, producing superior systemic tolerability. Ferrous iron accumulation is an exploitable feature of KRAS transformation, and FeADCs hold promise for improving the treatment of KRAS-driven solid tumors.
- Published
- 2021
19. Molecular imaging of prostate cancer targeting CD46 using immunoPET
- Author
-
Yung-hua Wang, Robert Dreicer, Jonathan Chou, Denis R. Beckford-Vera, Daniel Gioeli, Jun Hua, Yang Su, Mayuri Jayaraman, Felix Y. Feng, Jiang He, Sui Shen, Jun Li, Robert R. Flavell, Emily A. Egusa, Yangjie Huang, Michael J. Evans, Youngho Seo, Bin Liu, Sinan Wang, Rahul Aggarwal, Fujun Qin, Ning Zhao, Walter Zhao, Renuka Sriram, and Tony Huynh
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Immunoconjugates ,Nude ,Apoptosis ,Mice, SCID ,urologic and male genital diseases ,Prostate cancer ,Mice ,0302 clinical medicine ,Prostate ,Mice, Inbred NOD ,Tumor Cells, Cultured ,Tissue Distribution ,Cancer ,Cultured ,Prostate Cancer ,Imaging agent ,Tumor Cells ,Molecular Imaging ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Adenocarcinoma ,Urologic Diseases ,Biodistribution ,Oncology and Carcinogenesis ,Mice, Nude ,SCID ,Article ,Membrane Cofactor Protein ,03 medical and health sciences ,DU145 ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,Cell Proliferation ,business.industry ,Prevention ,Prostatic Neoplasms ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Positron-Emission Tomography ,Cancer research ,Inbred NOD ,Zirconium ,Molecular imaging ,Radiopharmaceuticals ,business - Abstract
Purpose: We recently identified CD46 as a novel therapeutic target in prostate cancer. In this study, we developed a CD46-targeted PET radiopharmaceutical, [89Zr]DFO-YS5, and evaluated its performance for immunoPET imaging in murine prostate cancer models. Experimental Design: [89Zr]DFO-YS5 was prepared and its in vitro binding affinity for CD46 was measured. ImmunoPET imaging was conducted in male athymic nu/nu mice bearing DU145 [AR−, CD46+, prostate-specific membrane antigen–negative (PSMA−)] or 22Rv1 (AR+, CD46+, PSMA+) tumors, and in NOD/SCID gamma mice bearing patient-derived adenocarcinoma xenograft, LTL-331, and neuroendocrine prostate cancers, LTL-331R and LTL-545. Results: [89Zr]DFO-YS5 binds specifically to the CD46-positive human prostate cancer DU145 and 22Rv1 xenografts. In biodistribution studies, the tumor uptake of [89Zr]DFO-YS5 was 13.3 ± 3.9 and 11.2 ± 2.5 %ID/g, respectively, in DU145 and 22Rv1 xenografts, 4 days postinjection. Notably, [89Zr]DFO-YS5 demonstrated specific uptake in the PSMA- and AR-negative DU145 model. [89Zr]DFO-YS5 also showed uptake in the patient-derived LTL-331 and -331R models, with particularly high uptake in the LTL-545 neuroendocrine prostate cancer tumors (18.8 ± 5.3, 12.5 ± 1.8, and 32 ± 5.3 %ID/g in LTL-331, LTL-331R, and LTL-545, respectively, at 4 days postinjection). Conclusions: [89Zr]DFO-YS5 is an excellent PET imaging agent across a panel of prostate cancer models, including in both adenocarcinoma and neuroendocrine prostate cancer, both cell line- and patient-derived xenografts, and both PSMA-positive and -negative tumors. It demonstrates potential for clinical translation as an imaging agent, theranostic platform, and companion biomarker in prostate cancer.
- Published
- 2020
20. An Analysis of Isoclonal Antibody Formats Suggests a Role for Measuring PD-L1 with Low Molecular Weight PET Radiotracers
- Author
-
Michael J. Evans, Yichen Xu, Cheng-I Wang, Charles S. Craik, Robert R. Flavell, Junnian Wei, Charles Truillet, Chia Yin Lee, Davide Ruggero, Youngho Seo, Lawrence Fong, David Y. Oh, Henry F. VanBrocklin, and Yung-hua Wang
- Subjects
Cancer Research ,Physiology ,medicine.medical_treatment ,Endogeny ,Immune checkpoint inhibitor ,B7-H1 Antigen ,Mice ,Neoplasms ,Tissue Distribution ,Cancer ,Tumor ,biology ,Chemistry ,Precision medicine ,Predictive biomarker ,Nuclear Medicine & Medical Imaging ,Oncology ,Hepatocellular carcinoma ,Biomedical Imaging ,Immunotherapy ,Antibody ,Clone (B-cell biology) ,Biodistribution ,Clinical Sciences ,Bioengineering ,Article ,Antibodies ,Cell Line ,Rare Diseases ,PD-L1 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Animal ,medicine.disease ,Brain Disorders ,Molecular Weight ,Disease Models, Animal ,Kinetics ,Positron-Emission Tomography ,Disease Models ,Cancer research ,biology.protein ,Zirconium ,Radiopharmaceuticals ,Digestive Diseases ,Single-Chain Antibodies - Abstract
PURPOSE: The swell of new and diverse radiotracers to predict or monitor tumor response to cancer immunotherapies invites the opportunity for comparative studies to identify optimal platforms. To probe the significance of antibody format on image quality for PD-L1 imaging, we developed and studied the biodistribution of a library of antibodies based on the anti-PD-L1 IgG1 clone C4. PROCEDURE: A C4 minibody and scFv were cloned, expressed and characterized. The antibodies were functionalized with desferrioxamine and radiolabeled with Zr-89 to enable rigorous comparison to prior data collected using (89)Zr-labeled C4 IgG1. The biodistribution of the radiotracers was evaluated in C57Bl6/J or nu/nu mice bearing B16F10 or H1975 tumors, respectively, which are models that represent high and low tumor autonomous PD-L1 expression. RESULTS: The tumor uptake of the (89)Zr-C4 minibody was higher than (89)Zr-C4 scFv, and equivalent to previous data collected using (89)Zr-C4 IgG1. However, the peak tumor to normal tissue ratios were generally higher for (89)Zr-C4 scFv compared to (89)Zr-C4 minibody and (89)Zr-IgG1. Moreover, an exploratory study showed that the rapid clearance of (89)Zr-C4 scFv enabled detection of endogenous PD-L1 on a genetically engineered and orthotopic model of hepatocellular carcinoma. CONCLUSION: In summary, these data support the use of low molecular weight constructs for PD-L1 imaging, especially for tumor types that manifest in abdominal organs that are obstructed by the clearance of high molecular weight radioligands.
- Published
- 2020
21. Theranostic Targeting of CUB Domain Containing Protein 1 (CDCP1) in Pancreatic Cancer
- Author
-
Alexander J. Martinko, Lydia H. Zhang, Kevin Leung, Eric A. Collisson, Anna Moroz, Sean D. Carlin, Ning Zhao, Kimberly S. Kirkwood, Yung-hua Wang, Michael J. Evans, Shion A. Lim, Zhuo Chen, Jie Zhuo, Junnian Wei, James A. Wells, Jeremy Sharib, Yangjie Huang, and Youngho Seo
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Single Photon Emission Computed Tomography Computed Tomography ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Article ,03 medical and health sciences ,Mice ,Pancreatic Cancer ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Rare Diseases ,Antigens, Neoplasm ,Pancreatic cancer ,Positron Emission Tomography Computed Tomography ,medicine ,Radioligand ,Animals ,Humans ,Tissue Distribution ,Oncology & Carcinogenesis ,Antigens ,Precision Medicine ,Cytotoxicity ,Pancreas ,Cancer ,biology ,business.industry ,medicine.disease ,CUB domain ,Xenograft Model Antitumor Assays ,Pancreatic Neoplasms ,030104 developmental biology ,Immunological ,Oncology ,Ectodomain ,030220 oncology & carcinogenesis ,CDCP1 ,biology.protein ,Cancer research ,Neoplasm ,Antibody ,business ,Digestive Diseases ,Cell Adhesion Molecules - Abstract
Purpose: The recent emergence of radioligand therapies for cancer treatment has increased enthusiasm for developing new theranostic strategies coupling both imaging and cytotoxicity in the same entity. In this study, we evaluated whether CUB domain containing protein 1 (CDCP1), a single-pass transmembrane protein highly overexpressed in diverse human cancers, might be a target for cancer theranostics. Experimental Design: The ectodomain of CDCP1 was targeted using radiolabeled forms of 4A06, a potent and specific recombinant human antibody that we developed. Imaging and antitumor assessment studies were performed in animal models of pancreatic cancer, including two patient-derived xenograft models we developed for this study. For antitumor assessment studies, the endpoints were death due to tumor volume >3,000 mm3 or ≥20% loss in body weight. Specific tracer binding or antitumor effects were assessed with an unpaired, two-tailed Student t test and survival advantages were assessed with a log rank (Mantel–Cox) test. Differences at the 95% confidence level were interpreted to be significant. Results: 89Zr-4A06 detected a broad dynamic range of full length or cleaved CDCP1 expression on seven human pancreatic cancer tumors (n = 4/tumor). Treating mice with single or fractionated doses of 177Lu-4A06 significantly reduced pancreatic cancer tumor volume compared with mice receiving vehicle or unlabeled 4A06 (n = 8; P < 0.01). A single dose of 225Ac-4A06 also inhibited tumor growth, although the effect was less profound compared with 177Lu-4A06 (n = 8; P < 0.01). A significant survival advantage was imparted by 225Ac-4A06 (HR = 2.56; P < 0.05). Conclusions: These data establish that CDCP1 can be exploited for theranostics, a finding with widespread implications given its breadth of overexpression in cancer.
- Published
- 2020
22. Ferronostics: Measuring Tumoral Ferrous Iron with PET to Predict Sensitivity to Iron-Targeted Cancer Therapies
- Author
-
Davide Ruggero, Michael J. Evans, Ryan K Muir, Pavithra Viswanath, Youngho Seo, Yangjie Huang, Junnian Wei, Ying-Chu Chen, Adam R. Renslo, Nima Hooshdaran, Ning Zhao, and Yung-hua Wang
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Targeted therapy ,Basic Science Investigation ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Renal cell carcinoma ,Glioma ,Cell Line, Tumor ,Cytotoxic T cell ,Medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,business.industry ,Cancer ,Prodrug ,medicine.disease ,Molecular Imaging ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Female ,Radiopharmaceuticals ,business - Abstract
Background: Although cancer has been known for decades to harbor an insatiable appetite for iron, only recently has the chemistry emerged to exploit this altered state therapeutically, by targeting the expanded cytosolic ‘labile’ iron pool (LIP), of the cancer cell. The state of the art include therapies that react with the LIP to produce cytotoxic radical species (in some cases also releasing drug payloads), and molecules that exacerbate LIP-induced oxidative stress to trigger “ferroptosis”. Effectively implementing LIP targeted therapies in patients will require biomarkers to identify those tumors with the most elevated LIP, and thus most likely to succumb to LIP targeted interventions. Toward this goal, we tested herein whether tumor uptake of the novel LIP sensing radiotracer 18F-TRX aligns with tumor sensitivity to LIP targeted therapies. Methods:18F-TRX uptake was assessed in vivo among ten subcutaneous and orthotopic human xenograft models. Glioma and renal cell carcinoma were prioritized as these tumors have the highest relative expression levels of STEAP3, the oxidoreductase that reduces ferric iron to the ferrous oxidation state, in the Cancer Cell Line Encyclopedia. The antitumor effects of the LIP activated prodrug TRX-CBI, which releases the DNA alkylator cyclopropylbenzindoline (CBI), were compared in mice bearing U251 or PC3 xenografts, tumors with high and intermediate levels of 18F-TRX uptake, respectively. Results:18F-TRX showed a wide range of tumor accumulation. An antitumor assessment study showed that the growth of U251 xenografts, the model with the highest 18F-TRX uptake, was potently inhibited by TRX-CBI. Moreover, the antitumor effects against U251 were significantly greater than those observed for PC3 tumors, consistent with the relative 18F-TRX determined LIP levels in tumors prior to therapy. Lastly, a dosimetry study showed that the estimated effective human doses for adult males and females were comparable to those of other 18F-based imaging probes. Conclusion: We report the first evidence that tumor sensitivity to a LIP targeted therapy can be predicted with a molecular imaging tool. More generally, these data bring a new dimension to the nuclear theranostic model by showing a requirement for imaging to quantify in situ the concentration of a metastable bioanalyte toward predicting tumor drug sensitivity.
- Published
- 2020
23. A Novel Radioligand Reveals Tissue Specific Pharmacological Modulation of Glucocorticoid Receptor Expression with Positron Emission Tomography
- Author
-
Yangjie Huang, Charles Truillet, Michael J. Evans, Javier Villanueva-Meyer, Spencer C. Behr, David M. Wilson, Jason E. Gestwicki, Youngho Seo, Brian J. Feldman, Joseph E. Blecha, Hao Shao, Henry F. VanBrocklin, Rahul Aggarwal, Mohd Sayeed, Ning Zhao, Yung-hua Wang, and Junnian Wei
- Subjects
0301 basic medicine ,Agonist ,Male ,Fluorine Radioisotopes ,medicine.drug_class ,Knockout ,Adipose tissue ,Gene Expression ,Inbred C57BL ,01 natural sciences ,Biochemistry ,Dexamethasone ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Receptors, Glucocorticoid ,Glucocorticoid receptor ,Glucocorticoid ,In vivo ,Adipocyte ,Receptors ,Radioligand ,medicine ,Animals ,Glucocorticoids ,Mice, Knockout ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Articles ,General Medicine ,Biological Sciences ,0104 chemical sciences ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Nuclear receptor ,Positron-Emission Tomography ,Knockout mouse ,Chemical Sciences ,Quinolines ,Molecular Medicine ,Biomedical Imaging - Abstract
The complexity of glucocorticoid receptor (GR) signaling cannot be measured with direct tissue analysis in living subjects, which has stifled our understanding of GR’s role in human physiology or disease and impeded the development of selective GR modulators. Herein, we report 18F-5-(4-fluorobenzyl)-10-methoxy-2,2,4-trimethyl-2,5-dihydro-1H-chromeno[3,4-f]quinoline (18F-YJH08), a radioligand that enables noninvasive measurements of tissue autonomous GR expression levels in vivo with positron emission tomography (PET). YJH08 potently binds GR (Ki ∼ 0.4 nM) with ∼100-fold selectivity compared to nuclear hormone receptors in the same subfamily. 18F-YJH08 was prepared via Cu(OTf)2(py)4-mediated radiofluorination of an arylboronic acid pinacol ester with ∼12% decay corrected radiochemical yield from the starting 18F-fluoride ion. We applied treatment with the tissue-wide GR agonist dexamethasone and adrenalectomy and generated an adipocyte specific GR knockout mouse to show that 18F-YJH08 specifically binds GR in normal mouse tissues, including those for which aberrant GR expression is thought to drive severe diseases (e.g., brain, adipose tissue, kidneys). Remarkably, 18F-YJH08 PET also revealed that JG231, a potent and bioavailable HSP70 inhibitor, selectively degrades GR only in the adipose tissue of mice, a finding that foreshadows how GR targeted PET might be integrated into drug discovery to screen for selective GR modulation at the tissue level, beyond the historical screening that was performed at the transcriptional level. In summary, 18F-YJH08 enables a quantitative assessment of GR expression levels in real time among multiple tissues simultaneously, and this technology is a first step toward unraveling the daunting complexity of GR signaling and rationally engineering tissue specific therapeutic modulators in vivo.
- Published
- 2020
24. Full-field refractive index measurement using absolute-phase total internal reflection heterodyne interferometry
- Author
-
Jing-Heng Chen, Kun-Huang Chen, Yung-Hua Wang, and Chih-Hsiung Lin
- Subjects
Heterodyne ,Physics ,Total internal reflection ,Physics and Astronomy (miscellaneous) ,business.industry ,Absolute phase ,General Engineering ,Phase (waves) ,General Physics and Astronomy ,Interferometry ,Optics ,Amplitude ,Prism ,business ,Refractive index ,Computer Science::Databases - Abstract
In this paper, absolute-phase total internal reflection heterodyne interferometry is proposed for measuring a full-field refractive index. In this method, an optical configuration of prism-based total internal reflection is used. If an electro-optical modulator is driven by a sawtooth wave with an amplitude lower than the half-wave voltage, heterodyne interferometric signals can be generated in segment mode. After a camera has captured these discrete interferometric signals, the optimum signal segment can be analyzed; a specific time segment can be inserted so that the interferometric signals of every pixel can be fitted as a continuous sinusoidal wave; a least-squares sine-fitting algorithm can extract the initial phase. Finally, the absolute phase can be calculated by subtraction of the characteristic phase, and the full-field refractive index can be obtained. To prove the feasibility of the proposed method, the different combinations of specimens were measured, including air, water, glycerol, and castor oil. The experimental data correspond well with the theoretical values. This method features simple operation, low influence of environmental disturbance, rapid measurement, and high resolution.
- Published
- 2020
25. The Synthesis and Structural Requirements for Measuring Glucocorticoid Receptor Expression In Vivo with (±)
- Author
-
Yangjie, Huang, Ning, Zhao, Yung-Hua, Wang, Charles, Truillet, Junnian, Wei, Matthew F L, Parker, Joseph E, Blecha, Christopher R, Drake, Henry F, VanBrocklin, Diego, Garrido-Ruiz, Matthew P, Jacobson, Rahul, Aggarwal, Spencer C, Behr, Robert R, Flavell, David M, Wilson, Youngho, Seo, and Michael J, Evans
- Subjects
Mice ,Receptors, Glucocorticoid ,Gene Expression Regulation ,Protein Domains ,Positron-Emission Tomography ,Animals ,Tissue Distribution ,Carbon Radioisotopes ,Chemistry Techniques, Synthetic ,Basic Science Investigation - Abstract
Noninvasive methods to study glucocorticoid receptor (GR) signaling are urgently needed to elaborate the complexity of GR signaling in normal physiology and human disorders, as well as to identify selective GR modulators to treat diseases. Here, we report evidence supporting translational studies with (±)-(11)C-5-(4-fluorobenzyl)-10-methoxy-2,2,4-trimethyl-2,5-dihydro-1H-chromeno[3,4-f]-quinoline ((±)-(11)C-YJH08), a radioligand for PET that engages the ligand binding domain on GR. Methods: (±)-(11)C-YJH08 was synthesized by reacting the phenol precursor with (11)C-methyl iodide. The biodistribution was studied in vivo. Specific binding was tested in vivo with adrenalectomy and ligand competition. A library of analogs was synthesized and studied in vitro and in vivo to understand the (±)-(11)C-YJH08 structure–activity relationship. Rodent dosimetry studies were performed to estimate the human-equivalent doses of (±)-(11)C-YJH08. Results: (±)-(11)C-YJH08 was synthesized by reaction of the phenolic precursor with (11)C-methyl iodide, giving a radiochemical yield of 51.7% ± 4.7% (decay-corrected to starting (11)C-methyl iodide). Specific binding was observed in many tissues, including the brain. An analysis of the (±)-YJH08 structure–activity relationship showed that (R)- and (S)-enantiomers are equally avid for GR by occupying discrete binding modes. A focused chemical screen revealed that the aryl fluoride motif on YJH08 is essential for high-affinity GR binding in vitro, high tissue uptake in vivo, and efficient passage across the blood–brain barrier. Lastly, we performed dosimetry studies on rodents, from which we estimated the human-equivalent doses of (±)-(11)C-YJH08 to be commensurate with the widely used (11)C and (18)F tracers. Conclusion: These studies reveal the molecular determinants of a high-affinity and high-selectivity ligand–receptor interaction and support the use of (±)-(11)C-YJH08 PET to make the first measurements of GR expression in human subjects.
- Published
- 2020
26. Exploiting KRAS-Driven Ferroaddiction in Cancer Through Ferrous Iron-Activatable Drug Conjugates (FeADC)
- Author
-
Eric A. Collisson, Ryan K Muir, Adam R. Renslo, Yung-hua Wang, Spencer C. Behr, Byron Hann, Honglin Jiang, Iwei Yeh, Ning Zhao, Ryan L. Gonciarz, Michael J. Evans, and Adam B. Olshen
- Subjects
MAPK/ERK pathway ,Drug ,business.industry ,MEK inhibitor ,media_common.quotation_subject ,Cancer ,medicine.disease_cause ,medicine.disease ,Blockade ,Tolerability ,Cancer research ,Medicine ,Avidity ,KRAS ,business ,media_common - Abstract
KRAS mutations cause a quarter of cancer mortality and currently are not sensitive to any targeted, FDA-approved agents. Several inhibitors of the MAPK pathway are FDA approved but exhibit low clinical tolerability at doses needed to adequately extinguish KRAS signaling. We discovered an avidity for ferrous iron (Fe2+) induced by and dependent on oncogenic KRAS signaling. We leveraged anFDA-approved MEK inhibitor to produce a prototypical Ferrous Iron–Activatable Drug Conjugate (FeADC) and with this novel agent achieved MAPK blockade in tumor cells while sparing normal tissues. These improvements allowed sustainable, effective treatment of tumor bearing animals with superior tolerability. Iron-activated therapeutics are unknown outside of antiparasitic therapy, but may hold significant promise for the treatment of KRAS-driven solid tumors.
- Published
- 2020
27. Measuring glucocorticoid receptor expressionin vivowith PET
- Author
-
Khaled M. Jami, Matthew F.L. Parker, Renuka Sriram, Loc T. Huynh, Junnian Wei, Yung-hua Wang, Charles Truillet, David M. Wilson, Michael J. Evans, Yuqin S. Shen, and John Kurhanewicz
- Subjects
0301 basic medicine ,business.industry ,medicine.drug_class ,Adrenalectomy ,medicine.medical_treatment ,Antagonist ,Cancer ,Mifepristone ,medicine.disease ,3. Good health ,03 medical and health sciences ,Prostate cancer ,030104 developmental biology ,0302 clinical medicine ,Glucocorticoid receptor ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Corticosteroid ,business ,medicine.drug - Abstract
The glucocorticoid receptor (GR) is an emerging drug target for several common and deadly solid tumors like breast and prostate cancer, and clinical trials studying the antitumor effects of GR antagonists are beginning. Since GR expression can be variable in tumor cells, and virtually all normal mammalian tissues express some GR, we hypothesized that an imaging tool capable of detecting GR positive tumors and/or measuring GR occupancy by drug in tumor and normal tissues could improve the precision application of anti-GR therapies in the clinic. To this end, we developed a fluorine-18 labeled corticosteroid termed GR02 that potently binds the endogenous ligand binding pocket on full length GR. Binding of 18F-GR02 was suppressed in many normal tissues by co-treatment with mifepristone, a GR antagonist in human use, and was elevated in many normal tissues among mice lacking circulating corticosteroids due to adrenalectomy. 18F-GR02 also accumulated in GR positive subcutaneous and subrenal capsule prostate cancer models, and uptake in tumors was competed by mifepristone. Combined with a straightforward and high yielding radiosynthesis, these data establish the foundation for near-term clinical translation of 18F-GR02.
- Published
- 2018
28. Switchable assembly and function of antibody complexes in vivo using a small molecule.
- Author
-
Martinko, Alexander J., Simonds, Erin F., Prasad, Suchitra, Ponce, Alberto, Bracken, Colton J., Junnian Wei, Yung-Hua Wang, Chow, Tiffany-Lynn, Zhong Huang, Evans, Michael J., Wells, James A., and Hill, Zachary B.
- Subjects
SMALL molecules ,BISPECIFIC antibodies ,MONOCLONAL antibodies ,IMMUNOGLOBULINS ,ANTIBODY-drug conjugates ,COMMERCIAL products - Abstract
The antigen specificity and long serum half-life of monoclonal antibodies have made them a critical part of modern therapeutics. These properties have been coopted in a number of synthetic formats, such as antibody–drug conjugates, bispecific antibodies, or Fc-fusion proteins to generate novel biologic drug modalities. Historically, these new therapies have been generated by covalently linking multiple molecular moieties through chemical or genetic methods. This irreversible fusion of different components means that the function of the molecule is static, as determined by the structure. Here, we report the development of a technology for switchable assembly of functional antibody complexes using chemically induced dimerization domains. This approach enables control of the antibody’s intended function in vivo by modulating the dose of a small molecule. We demonstrate this switchable assembly across three therapeutically relevant functionalities in vivo, including localization of a radionuclide-conjugated antibody to an antigen-positive tumor, extension of a cytokine’s halflife, and activation of bispecific, T cell–engaging antibodies. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. In Vivo Profiling with 18F-YJH08 Reveals Diverse Tissue Patterns of Antagonist/Glucocorticoid Receptor Interactions.
- Author
-
Hyunjung Kim, Yangjie Huang, Ning Zhao, Yung-hua Wang, Sakhamuri, Sasank, Chopra, Shalini, Hooshdaran, Nima, Sriram, Renuka, Aggarwal, Rahul, and Evans, Michael J.
- Published
- 2022
- Full Text
- View/download PDF
30. Measuring Dynamic Changes in the Labile Iron Pool in Vivo with a Reactivity-Based Probe for Positron Emission Tomography
- Author
-
Anna Moroz, Ryan K Muir, Yangjie Huang, Ning Zhao, Adam R. Renslo, Davide Ruggero, Renuka Sriram, Michael J. Evans, Ying-Chu Chen, Junnian Wei, John Kurhanewicz, and Yung-hua Wang
- Subjects
General Chemical Engineering ,010402 general chemistry ,01 natural sciences ,Ferrous ,Prostate cancer ,In vivo ,medicine ,PTEN ,Reactivity (chemistry) ,QD1-999 ,Cancer ,medicine.diagnostic_test ,biology ,010405 organic chemistry ,Chemistry ,General Chemistry ,medicine.disease ,3. Good health ,0104 chemical sciences ,Biochemistry ,Positron emission tomography ,Chemical Sciences ,biology.protein ,Preclinical imaging - Abstract
[Image: see text] Redox cycling of iron powers various enzyme functions crucial for life, making the study of iron acquisition, storage, and disposition in the whole organism a worthy topic of inquiry. However, despite its important role in biology and disease, imaging iron in animals with oxidation-state specificity remains an outstanding problem in biology and medicine. Here we report a first-generation reactivity-based probe of labile ferrous iron suitable for positron emission tomography studies in live animals. The responses of this reagent to systemic changes in labile iron disposition were revealed using iron supplementation and sequestration treatments in mice, while the potential of this approach for in vivo imaging of cancer was demonstrated using genetically and pathologically diverse mouse models, including spontaneous tumors arising in a genetically engineered model of prostate cancer driven by loss of PTEN.
- Published
- 2019
31. Targeting iron metabolism in high-grade glioma with 68Ga-citrate PET/MR
- Author
-
Spencer C. Behr, Soonmee Cha, Julia K.L. Lee, Anna Moroz, Wendy Ma, Yan Li, Sarah J. Nelson, Michael J. Evans, Jeffrey C. Hsiao, Javier Villanueva-Meyer, Youngho Seo, Junnian Wei, Susan M. Chang, Kenneth T. Gao, David M. Wilson, and Yung-hua Wang
- Subjects
Male ,0301 basic medicine ,Pathology ,Magnetic Resonance Spectroscopy ,Gallium ,Brain cancer ,Ferric Compounds ,Central Nervous System Neoplasms ,Mice ,0302 clinical medicine ,Models ,Positron Emission Tomography Computed Tomography ,Enhancing Lesion ,Citrates ,Cancer ,chemistry.chemical_classification ,Transferrin ,Glioma ,General Medicine ,Middle Aged ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Models, Animal ,Diagnostic imaging ,Biomedical Imaging ,Female ,Research Article ,Biotechnology ,medicine.drug ,Adult ,medicine.medical_specialty ,Bevacizumab ,Iron ,Clinical Trials and Supportive Activities ,Bioengineering ,White matter ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,medicine ,Animals ,Humans ,High-Grade Glioma ,Animal ,business.industry ,Neurosciences ,Metabolism ,medicine.disease ,Brain Disorders ,030104 developmental biology ,chemistry ,Radiopharmaceuticals ,Neoplasm Grading ,Apoproteins ,business ,Neuroscience - Abstract
Noninvasive tools that target tumor cells could improve the management of glioma. Cancer generally has a high demand for Fe(III), an essential nutrient for a variety of biochemical processes. We tested whether 68Ga-citrate, an Fe(III) biomimetic that binds to apo-transferrin in blood, detects glioma in preclinical models and patients using hybrid PET/MRI. Mouse PET/CT studies showed that 68Ga-citrate accumulates in subcutaneous U87MG xenografts in a transferrin receptor-dependent fashion within 4 hours after injection. Seventeen patients with WHO grade III or IV glioma received 3.7-10.2 mCi 68Ga-citrate and were imaged with PET/MR 123-307 minutes after injection to establish that the radiotracer can localize to human tumors. Multiple contrast-enhancing lesions were PET avid, and tumor to adjacent normal white matter ratios were consistently greater than 10:1. Several contrast-enhancing lesions were not PET avid. One minimally enhancing lesion and another tumor with significantly reduced enhancement following bevacizumab therapy were PET avid. Advanced MR imaging analysis of one patient with contrast-enhancing glioblastoma showed that metabolic hallmarks of viable tumor spatially overlaid with 68Ga-citrate accumulation. These early data underscore that high-grade glioma may be detectable with a radiotracer that targets Fe(III) transport.
- Published
- 2018
32. A Preclinical Assessment Of 89Zr-atezolizumab Identifies A Requirement For Carrier Added Formulations Not Observed With 89Zr-C4
- Author
-
Chia Yin Lee, Natalia Sevillano, Charles S. Craik, Michael J. Evans, Anna Moroz, Lawrence Fong, Charles Truillet, Cheng-I Wang, Jeffrey C. Hsiao, and Yung-hua Wang
- Subjects
Drug Compounding ,Nude ,Biomedical Engineering ,Measure (physics) ,Pharmaceutical Science ,Mice, Nude ,Bioengineering ,Antineoplastic Agents ,Computational biology ,Drug Screening Assays ,Antibodies, Monoclonal, Humanized ,Protein expression ,Antibodies ,Article ,030218 nuclear medicine & medical imaging ,Cell Line ,03 medical and health sciences ,Medicinal and Biomolecular Chemistry ,Mice ,0302 clinical medicine ,Rare Diseases ,Atezolizumab ,Cell Line, Tumor ,Positron Emission Tomography Computed Tomography ,Monoclonal ,Animals ,Humans ,Tissue Distribution ,Humanized ,Cancer ,Pharmacology ,Drug Carriers ,Tumor ,Chemistry ,Organic Chemistry ,Antibodies, Monoclonal ,Antitumor ,Immune checkpoint ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Biochemistry and Cell Biology ,Zirconium ,Drug Screening Assays, Antitumor ,Radiopharmaceuticals ,Biotechnology - Abstract
The swell of experimental imaging technologies to non-invasively measure immune checkpoint protein expression presents the opportunity for rigorous comparative studies toward identifying a gold standard. (89)Zr-atezolizumab is currently in man, and early data show tumor targeting but also abundant uptake in several normal tissues. Therefore, we conducted a reverse translational study, both to understand if tumor to normal tissue ratios for (89)Zr-atezolizumab could be improved, and to make direct comparisons to (89)Zr-C4, a radiotracer that we showed can detect a large dynamic range of tumor-associated PD-L1 expression. PET/CT and biodistribution studies in tumor bearing immunocompetent and nu/nu mice revealed that high specific activity (89)Zr-atezolizumab (~2 μCi/μg) binds to PD-L1 on tumors, but also results in very high uptake in many normal mouse tissues, as expected. Unexpectedly, (89)Zr-atezolizumab uptake was generally higher in normal mouse tissues compared to (89)Zr-C4, and lower in H1975, a tumor model with modest PD-L1 expression. Also unexpectedly, reducing the specific activity at least 15 fold suppressed (89)Zr-atezo uptake in normal mouse tissues, but increased tumor uptake to levels observed with high specific activity (89)Zr-C4. In summary, these data reveal that low specific activity (89)Zr-atezo may be necessary for accurately measuring PD-L1 in the tumor microenvironment, assuming a threshold can be identified that preferentially suppresses binding in normal tissues without reducing binding to tumors with abundant expression. Alternatively, high specific activity approaches like (89)Zr-C4 PET may be simpler to implement clinically to measure the broad dynamic range of PD-L1 expression known to manifest among tumors.
- Published
- 2018
33. Imaging PD-L1 Expression with ImmunoPET
- Author
-
Anna Moroz, Khaled M. Jami, Chia Yin Lee, Hseuh Ling J. Oh, Emilie Jaumain, Cheng-I Wang, Yuqin S. Shen, Siok Ping Yeo, Victor Olivas, Albert J. Chang, Loc T. Huynh, Lawrence Fong, Michael J. Evans, Junnian Wei, Trever G. Bivona, Yung-hua Wang, Collin M. Blakely, Charles S. Craik, Matthew F.L. Parker, Charles Truillet, Benoit Jego, and Natalia Sevillano
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,Immunoconjugates ,medicine.medical_treatment ,Pharmaceutical Science ,Inbred C57BL ,Epitope ,B7-H1 Antigen ,Prostate cancer ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Carcinoma, Non-Small-Cell Lung ,Positron Emission Tomography Computed Tomography ,Non-Small-Cell Lung ,Lung ,Cancer ,Tumor ,biology ,Chemistry ,Lung Cancer ,Recombinant Proteins ,3. Good health ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Biomedical Imaging ,Antibody ,Development of treatments and therapeutic interventions ,Biotechnology ,Biomedical Engineering ,Bioengineering ,Article ,Cell Line ,03 medical and health sciences ,Medicinal and Biomolecular Chemistry ,Antigen ,Atezolizumab ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Lung cancer ,Pharmacology ,Radioisotopes ,Carcinoma ,Organic Chemistry ,medicine.disease ,Molecular biology ,Mice, Inbred C57BL ,030104 developmental biology ,Good Health and Well Being ,HEK293 Cells ,Cell culture ,Immunoglobulin G ,biology.protein ,Zirconium ,Biochemistry and Cell Biology - Abstract
High sensitivity imaging tools could provide a more holistic view of target antigen expression to improve the identification of patients who might benefit from cancer immunotherapy. We developed for immunoPET a novel recombinant human IgG1 (termed C4) that potently binds an extracellular epitope on human and mouse PD-L1 and radiolabeled the antibody with zirconium-89. Small animal PET/CT studies showed that 89Zr-C4 detected antigen levels on a patient derived xenograft (PDX) established from a non-small-cell lung cancer (NSCLC) patient before an 8-month response to anti-PD-1 and anti-CTLA4 therapy. Importantly, the concentration of antigen is beneath the detection limit of previously developed anti-PD-L1 radiotracers, including radiolabeled atezolizumab. We also show that 89Zr-C4 can specifically detect antigen in human NSCLC and prostate cancer models endogenously expressing a broad range of PD-L1. 89Zr-C4 detects mouse PD-L1 expression changes in immunocompetent mice, suggesting that endogenous PD-1/2 will not confound human imaging. Lastly, we found that 89Zr-C4 could detect acute changes in tumor expression of PD-L1 due to standard of care chemotherapies. In summary, we present evidence that low levels of PD-L1 in clinically relevant cancer models can be imaged with immunoPET using a novel recombinant human antibody.
- Published
- 2018
34. Ferronostics: Measuring Tumoral Ferrous Iron with PET to Predict Sensitivity to Iron-Targeted Cancer Therapies.
- Author
-
Ning Zhao, Yangjie Huang, Yung-hua Wang, Muir, Ryan K., Ying-Chu Chen, Junnian Wei, Hooshdaran, Nima, Viswanath, Pavithra, Youngho Seo, Ruggero, Davide, Renslo, Adam R., and Evans, Michael J.
- Published
- 2020
- Full Text
- View/download PDF
35. The synthesis and structural requirements for measuring glucocorticoid receptor expression in vivo with (±)-11C-YJH08 PET.
- Author
-
Yangjie Huang, Ning Zhao, Yung-hua Wang, Truillet, Charles, Junnian Wei, Parker, Matthew F. L., Blecha, Joseph E., Drake, Christopher R., VanBrocklin, Henry F., Garrido-Ruiz, Diego, Jacobson, Matthew P., Aggarwal, Rahul, Behr, Spencer C., Flavell, Robert R., Wilson, David M., Youngho Seo, and Evans, Michael J.
- Published
- 2020
- Full Text
- View/download PDF
36. Measuring glucocorticoid receptor expression
- Author
-
Charles, Truillet, Matthew F L, Parker, Loc T, Huynh, Junnian, Wei, Khaled M, Jami, Yung-Hua, Wang, Yuqin S, Shen, Renuka, Sriram, David M, Wilson, John, Kurhanewicz, and Michael J, Evans
- Subjects
positron emission tomography ,precision medicine ,glucocorticoid receptor ,pharmacodynamics ,cancer ,Research Paper - Abstract
The glucocorticoid receptor (GR) is an emerging drug target for several common and deadly solid tumors like breast and prostate cancer, and clinical trials studying the antitumor effects of GR antagonists are beginning. Since GR expression can be variable in tumor cells, and virtually all normal mammalian tissues express some GR, we hypothesized that an imaging tool capable of detecting GR positive tumors and/or measuring GR occupancy by drug in tumor and normal tissues could improve the precision application of anti-GR therapies in the clinic. To this end, we developed a fluorine-18 labeled corticosteroid termed GR02 that potently binds the endogenous ligand binding pocket on full length GR. Binding of 18F-GR02 was suppressed in many normal tissues by co-treatment with mifepristone, a GR antagonist in human use, and was elevated in many normal tissues among mice lacking circulating corticosteroids due to adrenalectomy. 18F-GR02 also accumulated in GR positive subcutaneous and subrenal capsule prostate cancer models, and uptake in tumors was competed by mifepristone. Combined with a straightforward and high yielding radiosynthesis, these data establish the foundation for near-term clinical translation of 18F-GR02.
- Published
- 2017
37. Imaging PD-L1 Expression with ImmunoPET.
- Author
-
Truillet, Charles, Oh, Hsueh Ling J., Siok Ping Yeo, Chia-Yin Lee, Huynh, Loc T., Junnian Wei, Parker, Matthew F. L., Blakely, Collin, Sevillano, Natalia, Yung-Hua Wang, Shen, Yuqin S., Olivas, Victor, Jami, Khaled M., Moroz, Anna, Jego, Benoit, Jaumain, Emilie, Lawrence Fong, Craik, Charles S., Chang, Albert J., and Bivona, Trever G.
- Published
- 2018
- Full Text
- View/download PDF
38. Targeting a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) for RAS-driven cancers.
- Author
-
Lim, Shion A., Jie Zhou, Martinko, Alexander J., Yung-Hua Wang, Filippova, Ekaterina V., Steri, Veronica, Donghui Wang, Remesh, Soumya G., Jia Liu, Hann, Byron, Kossiakoff, Anthony A., Evans, Michael J., Leung, Kevin K., Wells, James A., Zhou, Jie, Wang, Yung-Hua, Wang, Donghui, and Liu, Jia
- Subjects
- *
PANCREATIC tumors , *PROTEINS , *RESEARCH , *ANIMAL experimentation , *RESEARCH methodology , *METABOLISM , *EVALUATION research , *COMPARATIVE studies , *CELL adhesion molecules , *RESEARCH funding , *TUMORS , *TUMOR antigens , *CELL lines , *PERSONALITY tests , *ANTIGENS , *MICE - Abstract
Extracellular proteolysis is frequently dysregulated in disease and can generate proteoforms with unique neoepitopes not found in healthy tissue. Here, we demonstrate that Abs that selectively recognize a proteolytic neoepitope on CUB domain containing protein 1 (CDCP1) could enable more effective and safer treatments for solid tumors. CDCP1 is highly overexpressed in RAS-driven cancers, and its ectodomain is cleaved by extracellular proteases. Biochemical, biophysical, and structural characterization revealed that the 2 cleaved fragments of CDCP1 remain tightly associated with minimal proteolysis-induced conformational change. Using differential phage display, we generated recombinant Abs that are exquisitely selective to cleaved CDCP1 with no detectable binding to the uncleaved form. These Abs potently targeted cleaved CDCP1-expressing cancer cells as an Ab-drug conjugate, an Ab-radionuclide conjugate, and a bispecific T cell engager. In a syngeneic pancreatic tumor model, these cleaved-specific Abs showed tumor-specific localization and antitumor activity with superior safety profiles compared with a pan-CDCP1 approach. Targeting proteolytic neoepitopes could provide an orthogonal "AND" gate for improving the therapeutic index. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. Tumor-conditional anti-CTLA4 uncouples antitumor efficacy from immunotherapy-related toxicity.
- Author
-
Chien-Chun Steven Pai, Simons, Donald M., Xiaoqing Lu, Evans, Michael, Junnian Wei, Yung-hua Wang, Mingyi Chen, Huang, John, Chanhyuk Park, Chang, Anthony, Jiaxi Wang, Westmoreland, Susan, Beam, Christine, Banach, Dave, Bowley, Diana, Feng Dong, Seaga, Jane, Ritacco, Wendy, Richardson, Paul L., and Mitra, Soumya
- Subjects
- *
IMMUNOTHERAPY , *TUMOR treatment , *CANCER treatment , *LYMPHOCYTES , *IPILIMUMAB - Abstract
While immune checkpoint blockade leads to potent antitumor efficacy, it also leads to immune-related adverse events in cancer patients. These toxicities stem from systemic immune activation resulting in inflammation of multiple organs, including the gastrointestinal tract, lung, and endocrine organs. We developed a dual variable domain immunoglobulin of anti-CTLA4 antibody (anti-CTLA4 DVD, where CTLA4 is defined as cytotoxic T lymphocyte-associated antigen-4) possessing an outer tumor-specific antigen-binding site engineered to shield the inner anti-CTLA4-binding domain. Upon reaching the tumor, the outer domain was cleaved by membrane type-serine protease 1 (MT-SP1) present in the tumor microenvironment, leading to enhanced localization of CTLA4 blockade. Anti-CTLA4 DVD markedly reduced multiorgan immune toxicity by preserving tissue-resident Tregs in Rag 1-/- mice that received naive donor CD4+ T cells from WT C57BL/6j mice. Moreover, anti-CTLA4 DVD induced potent antitumor effects by decreasing tumor-infiltrating Tregs and increasing the infiltration of antigen-specific CD8+ T lymphocytes in TRAMP-C2-bearing C57BL/6j mice. Treg depletion was mediated through the antibody-dependent cellular cytotoxicity (ADCC) mechanism, as anti-CTLA4 without the FcγR-binding portion (anti-CTLA4 DANA) spared Tregs, preventing treatment-induced toxicities. In summary, our results demonstrate an approach to anti-CTLA4 blockade that depletes tumor-infiltrating, but not tissue-resident, Tregs, preserving antitumor effects while minimizing toxicity. Thus, our tumor-conditional anti-CTLA4 DVD provides an avenue for uncoupling antitumor efficacy from immunotherapy-induced toxicities. [ABSTRACT FROM AUTHOR]
- 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.