591 results on '"Jason S. Lewis"'
Search Results
2. Phase 1 study of intraventricular 131I-omburtamab targeting B7H3 (CD276)-expressing CNS malignancies
- Author
-
Kim Kramer, Neeta Pandit-Taskar, Brian H. Kushner, Pat Zanzonico, John L. Humm, Ursula Tomlinson, Maria Donzelli, Suzanne L. Wolden, Sophia Haque, Ira Dunkel, Mark M. Souweidane, Jeffrey P. Greenfield, Satish Tickoo, Jason S. Lewis, Serge K. Lyashchenko, Jorge A. Carrasquillo, Bae Chu, Christopher Horan, Steven M. Larson, Nai-Kong V. Cheung, and Shakeel Modak
- Subjects
CNS tumors ,CNS metastases ,Radioimmunotherapy ,B7H3 ,Omburtamab ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The prognosis for metastatic and recurrent tumors of the central nervous system (CNS) remains dismal, and the need for newer therapeutic targets and modalities is critical. The cell surface glycoprotein B7H3 is expressed on a range of solid tumors with a restricted expression on normal tissues. We hypothesized that compartmental radioimmunotherapy (cRIT) with the anti-B7H3 murine monoclonal antibody omburtamab injected intraventricularly could safely target CNS malignancies. Patients and methods We conducted a phase I trial of intraventricular 131I-omburtamab using a standard 3 + 3 design. Eligibility criteria included adequate cerebrospinal fluid (CSF) flow, no major organ toxicity, and for patients > dose level 6, availability of autologous stem cells. Patients initially received 74 MBq radioiodinated omburtamab to evaluate dosimetry and biodistribution followed by therapeutic 131I-omburtamab dose-escalated from 370 to 2960 MBq. Patients were monitored clinically and biochemically for toxicity graded using CTCAEv 3.0. Dosimetry was evaluated using serial CSF and blood sampling, and serial PET or gamma-camera scans. Patients could receive a second cycle in the absence of grade 3/4 non-hematologic toxicity or progressive disease. Results Thirty-eight patients received 100 radioiodinated omburtamab injections. Diagnoses included metastatic neuroblastoma (n = 16) and other B7H3-expressing solid tumors (n = 22). Thirty-five patients received at least 1 cycle of treatment with both dosimetry and therapy doses. Acute toxicities included
- Published
- 2022
- Full Text
- View/download PDF
3. Caveolin-1 temporal modulation enhances antibody drug efficacy in heterogeneous gastric cancer
- Author
-
Patrícia M. R. Pereira, Komal Mandleywala, Sébastien Monette, Melissa Lumish, Kathryn M. Tully, Sandeep Surendra Panikar, Mike Cornejo, Audrey Mauguen, Ashwin Ragupathi, Nai C. Keltee, Marissa Mattar, Yelena Y. Janjigian, and Jason S. Lewis
- Subjects
Science - Abstract
Clinical evidences have demonstrated limited efficacy of HER2-targeted therapies in patients with gastric cancer (GC). Here the authors show that survival benefit to anti-HER2 antibody Trastuzumab is reduced in GC patients with high levels of the caveolin-1 and that, in preclinical cancer models, antibody drug efficacy can be improved by modulating caveolin-1 levels with cholesterol-depleting drugs, statins.
- Published
- 2022
- Full Text
- View/download PDF
4. Chemical tools for epichaperome-mediated interactome dysfunctions of the central nervous system
- Author
-
Alexander Bolaender, Danuta Zatorska, Huazhong He, Suhasini Joshi, Sahil Sharma, Chander S. Digwal, Hardik J. Patel, Weilin Sun, Brandon S. Imber, Stefan O. Ochiana, Maulik R. Patel, Liza Shrestha, Smit. K. Shah, Shuo Wang, Rashad Karimov, Hui Tao, Pallav D. Patel, Ananda Rodilla Martin, Pengrong Yan, Palak Panchal, Justina Almodovar, Adriana Corben, Andreas Rimner, Stephen D. Ginsberg, Serge Lyashchenko, Eva Burnazi, Anson Ku, Teja Kalidindi, Sang Gyu Lee, Milan Grkovski, Bradley J. Beattie, Pat Zanzonico, Jason S. Lewis, Steve Larson, Anna Rodina, Nagavarakishore Pillarsetty, Viviane Tabar, Mark P. Dunphy, Tony Taldone, Fumiko Shimizu, and Gabriela Chiosis
- Subjects
Science - Abstract
Here, the authors show structural, biochemical, and functional insights into the discovery of epichaperome‐ directed chemical probes for use in central nervous system diseases. Probes emerging from this work have translated to human clinical studies in Alzheimer’s disease and cancer.
- Published
- 2021
- Full Text
- View/download PDF
5. Refined Chelator Spacer Moieties Ameliorate the Pharmacokinetics of PSMA-617
- Author
-
José Carlos dos Santos, Martin Schäfer, Ulrike Bauder-Wüst, Barbro Beijer, Matthias Eder, Karin Leotta, Christian Kleist, Jan-Philip Meyer, Thomas R. Dilling, Jason S. Lewis, Clemens Kratochwil, Klaus Kopka, Uwe Haberkorn, and Walter Mier
- Subjects
PSMA ,prostate cancer ,PET imaging ,endoradiotherapy ,chelator ,Chemistry ,QD1-999 - Abstract
Prostate-specific membrane antigen (PSMA) binding tracers are promising agents for the targeting of prostate tumors. To further optimize the clinically established radiopharmaceutical PSMA-617, novel PSMA ligands for prostate cancer endoradiotherapy were developed. A series of PSMA binding tracers that comprise a benzyl group at the chelator moiety were obtained by solid-phase synthesis. The compounds were labeled with 68Ga or 177Lu. Competitive cell-binding assays and internalization assays were performed using the cell line C4-2, a subline of the PSMA positive cell line LNCaP (human lymph node carcinoma of the prostate). Positron emission tomography (PET) imaging and biodistribution studies were conducted in a C4-2 tumor bearing BALB/c nu/nu mouse model. All 68Ga-labeled ligands were stable in human serum over 2 h; 177Lu-CA030 was stable over 72 h. The PSMA ligands revealed inhibition potencies [Ki] (equilibrium inhibition constants) between 4.8 and 33.8 nM. The percentage of internalization of the injected activity/106 cells of 68Ga-CA028, 68Ga-CA029, and 68Ga-CA030 was 41.2 ± 2.7, 44.3 ± 3.9, and 53.8 ± 5.4, respectively; for the comparator 68Ga-PSMA-617, 15.5 ± 3.1 was determined. Small animal PET imaging of the compounds showed a high tumor-to-background contrast. Organ distribution studies revealed high specific uptake in the tumor, that is, approximately 34.4 ± 9.8% of injected dose per gram (%ID/g) at 1 h post injection for 68Ga-CA028. At 1 h p.i., 68Ga-CA028 and 68Ga-CA030 demonstrated lower kidney uptake than 68Ga-PSMA-617, but at later time points, kidney time–activity curves converge. In line with the preclinical data, first diagnostic PET imaging using 68Ga-CA028 and 68Ga-CA030 revealed high-contrast detection of bone and lymph node lesions in patients with metastatic prostate cancer. The novel PSMA ligands, in particular CA028 and CA030, are promising agents for targeting PSMA-positive tumor lesions as shown in the preclinical evaluation and in a first patient, respectively. Thus, clinical translation of 68Ga-CA028 and 68Ga/177Lu-CA030 for diagnostics and endoradiotherapy of prostate cancer in larger cohorts of patients is warranted.
- Published
- 2022
- Full Text
- View/download PDF
6. Applications of nuclear-based imaging in gene and cell therapy: probe considerations
- Author
-
Alessia Volpe, Naga Vara Kishore Pillarsetty, Jason S. Lewis, and Vladimir Ponomarev
- Subjects
nuclear imaging ,cell therapy ,gene therapy ,radiotracer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Several types of gene- and cell-based therapeutics are now emerging in the cancer immunotherapy, transplantation, and regenerative medicine landscapes. Radionuclear-based imaging can be used as a molecular imaging tool for repetitive and non-invasive visualization as well as in vivo monitoring of therapy success. In this review, we discuss the principles of nuclear-based imaging and provide a comprehensive overview of its application in gene and cell therapy. This review aims to inform investigators in the biomedical field as well as clinicians on the state of the art of nuclear imaging, from probe design to available radiopharmaceuticals and advances of direct (probe-based) and indirect (transgene-based) strategies in both preclinical and clinical settings. Notably, as the nuclear-based imaging toolbox is continuously expanding, it will be increasingly incorporated into the clinical setting where the distribution, targeting, and persistence of a new generation of therapeutics can be imaged and ultimately guide therapeutic decisions.
- Published
- 2021
- Full Text
- View/download PDF
7. PET Imaging of Acidic Tumor Environment With 89Zr-labeled pHLIP Probes
- Author
-
David Bauer, Hannah Visca, Anuradha Weerakkody, Lukas M. Carter, Zachary Samuels, Spencer Kaminsky, Oleg A. Andreev, Yana K. Reshetnyak, and Jason S. Lewis
- Subjects
pH-low insertion peptides ,membrane-insertion behavior ,acidic tumor microenvironment ,zirconium-89 ,PET imaging ,human dosimetry estimates ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Acidosis of the tumor microenvironment is a hallmark of tumor progression and has emerged as an essential biomarker for cancer diagnosis, prognosis, and evaluation of treatment response. A tool for quantitatively visualizing the acidic tumor environment could significantly advance our understanding of the behavior of aggressive tumors, improving patient management and outcomes. 89Zr-labeled pH-low insertion peptides (pHLIP) are a class of radiopharmaceutical imaging probes for the in vivo analysis of acidic tumor microenvironments via positron emission tomography (PET). Their unique structure allows them to sense and target acidic cancer cells. In contrast to traditional molecular imaging agents, pHLIP’s mechanism of action is pH-dependent and does not rely on the presence of tumor-specific molecular markers. In this study, one promising acidity-imaging PET probe ([89Zr]Zr-DFO-Cys-Var3) was identified as a candidate for clinical translation.
- Published
- 2022
- Full Text
- View/download PDF
8. Fluorescence labeling of a NaV1.7-targeted peptide for near-infrared nerve visualization
- Author
-
Junior Gonzales, Giacomo Pirovano, Chun Yuen Chow, Paula Demetrio de Souza Franca, Lukas M. Carter, Julie K. Klint, Navjot Guru, Jason S. Lewis, Glenn F. King, and Thomas Reiner
- Subjects
Hs1a-FL ,Nerve imaging ,Near-infrared imaging ,Intraoperative ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Background Accidental peripheral nerve injury during surgical intervention results in a broad spectrum of potentially debilitating side effects. Tissue distortion and poor visibility can significantly increase the risk of nerve injury with long-lasting consequences for the patient. We developed and characterized Hs1a-FL, a fluorescent near-infrared molecule for nerve visualization in the operating theater with the aim of helping physicians to visualize nerves during surgery. Hs1a was derived from the venom of the Chinese bird spider, Haplopelma schmidti, and conjugated to Cy7.5 dye. Hs1a-FL was injected intravenously in mice, and harvested nerves were imaged microscopically and with epifluorescence. Results Hs1a-FL showed specific and stable binding to the sodium channel NaV1.7, present on the surface of human and mouse nerves. Hs1a-FL allowed epifluorescence visualization of sciatic mouse nerves with favorable nerve-to-muscle contrast. Conclusions Fluorescent NaV1.7-targeted tracers have the potential to be adopted clinically for the intraoperative visualization of peripheral nerves during surgery, providing guidance for the surgeon and potentially improving the standard of care.
- Published
- 2020
- Full Text
- View/download PDF
9. Caveolin-1 mediates cellular distribution of HER2 and affects trastuzumab binding and therapeutic efficacy
- Author
-
Patrícia M. R. Pereira, Sai Kiran Sharma, Lukas M. Carter, Kimberly J. Edwards, Jacob Pourat, Ashwin Ragupathi, Yelena Y. Janjigian, Jeremy C. Durack, and Jason S. Lewis
- Subjects
Science - Abstract
Trastuzumab binding to tumor cells depends on the availability of HER2 at the cell membrane. Here the authors show that caveolin-1 (CAV1) regulates HER2 density at the cell membranes and that CAV1 gene knockdown or protein depletion via the cholesterol modulator lovastatin, increases trastuzumab binding and anti-tumor activity.
- Published
- 2018
- Full Text
- View/download PDF
10. Leveraging PET to image folate receptor α therapy of an antibody-drug conjugate
- Author
-
Christian Brand, Ahmad Sadique, Jacob L. Houghton, Kishore Gangangari, Jose F. Ponte, Jason S. Lewis, Naga Vara Kishore Pillarsetty, Jason A. Konner, and Thomas Reiner
- Subjects
89Zr ,Companion diagnostic ,PET imaging ,Antibody-drug-conjugate ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Background The folate receptor α (FRα)-targeting antibody-drug conjugate (ADC), IMGN853, shows great antitumor activity against FRα-expressing tumors in vivo, but patient selection and consequently therapy outcome are based on immunohistochemistry. The aim of this study is to develop an antibody-derived immuno-PET imaging agent strategy for targeting FRα in ovarian cancer as a predictor of treatment success. Methods We developed [89Zr]Zr-DFO-M9346A, a humanized antibody-based radiotracer targeting tumor-associated FRα in the preclinical setting. [89Zr]Zr-DFO-M9346A’s binding ability was tested in an in vitro uptake assay using cell lines with varying FRα expression levels. The diagnostic potential of [89Zr]Zr-M9346A was evaluated in KB and OV90 subcutaneous xenografts. Following intravenous injection of [89Zr]Zr-DFO-M9346A (~90 μCi, 50 μg), PET imaging and biodistribution studies were performed. We determined the blood half-life of [89Zr]Zr-DFO-M9346A and compared it to the therapeutic, radioiodinated ADC [131I]-IMGN853. Finally, in vivo studies using IMG853 as a therapeutic, paired with [89Zr]Zr-DFO-M9346A as a companion diagnostic were performed using OV90 xenografts. Results DFO-M9346A was labeled with Zr-89 at 37 °C within 60 min and isolated in labeling yields of 85.7 ± 5.7%, radiochemical purities of 98.0 ± 0.7%, and specific activities of 3.08 ± 0.43 mCi/mg. We observed high specificity for binding FRα positive cells in vitro. For PET and biodistribution studies, [89Zr]Zr-M9346A displayed remarkable in vivo performance in terms of excellent tumor uptake for KB and OV xenografts (45.8 ± 29.0 %IA/g and 26.1 ± 7.2 %IA/g), with low non-target tissue uptake in other organs such as kidneys (4.5 ± 1.2 %IA/g and 4.3 ± 0.7 %IA/g). A direct comparison of the blood half life of [89Zr]Zr-M9346A and [131I]-IMGN853 corroborated the equivalency of the radiopharmaceutical and the ADC, paving the way for a companion PET imaging study. Conclusions We developed a new folate receptor-targeted 89Zr-labeled PET imaging agent with excellent pharmacokinetics in vivo. Good tumor uptake in subcutaneous KB and OV90 xenografts were obtained, and ADC therapy studies were performed with the precision predictor.
- Published
- 2018
- Full Text
- View/download PDF
11. Feed-forward alpha particle radiotherapy ablates androgen receptor-addicted prostate cancer
- Author
-
Michael R. McDevitt, Daniel L. J. Thorek, Takeshi Hashimoto, Tatsuo Gondo, Darren R. Veach, Sai Kiran Sharma, Teja Muralidhar Kalidindi, Diane S. Abou, Philip A. Watson, Bradley J. Beattie, Oskar Vilhemsson Timmermand, Sven-Erik Strand, Jason S. Lewis, Peter T. Scardino, Howard I. Scher, Hans Lilja, Steven M. Larson, and David Ulmert
- Subjects
Science - Abstract
Radionuclides that emit alpha particles (charged helium nuclei) are currently used clinically to treat cancers including prostate cancer. Here, the authors combine a humanized antibody to an alpha particle emitter, specifically to target a downstream effector of the androgen receptor and create a feed forward loop that increases the therapeutic efficacy.
- Published
- 2018
- Full Text
- View/download PDF
12. Pretargeting of internalizing trastuzumab and cetuximab with a 18F-tetrazine tracer in xenograft models
- Author
-
Outi Keinänen, Kimberly Fung, Jacob Pourat, Vilma Jallinoja, Delphine Vivier, NagaVara Kishore Pillarsetty, Anu J. Airaksinen, Jason S. Lewis, Brian M. Zeglis, and Mirkka Sarparanta
- Subjects
Fluorine-18 ,Inverse electron-demand Diels-Alder (IEDDA) reaction ,Pretargeting ,Tetrazine ,Trans-cyclooctene ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Background Pretargeting-based approaches are being investigated for radioimmunoimaging and therapy applications to reduce the effective radiation burden to the patient. To date, only a few studies have used short-lived radioisotopes for pretargeting of antibodies, and such examples with internalizing antibodies are even rarer. Herein, we have investigated pretargeting methodology using inverse electron-demand Diels-Alder (IEDDA) for tracing two clinically relevant, internalizing monoclonal antibodies, cetuximab and trastuzumab. Results Bioorthogonal reaction between tetrazine and trans-cyclooctene (TCO) was used for tracing cetuximab and trastuzumab in vivo with a fluorine-18 (t ½ = 109.8 min) labelled tracer. TCO-cetuximab or TCO-trastuzumab was administered 24, 48, or 72 h prior to the injection of tracer to A431 or BT-474 tumour-bearing mice, respectively. With cetuximab, the highest tumour-to-blood ratios were achieved when the lag time between antibody and tracer injections was 72 h. With trastuzumab, no difference was observed between different lag times. For both antibodies, the tumour could be clearly visualized in the PET images with the highest tumour uptake of 3.7 ± 0.1%ID/g for cetuximab and 1.5 ± 0.1%ID/g for trastuzumab as quantified by ex vivo biodistribution. In vivo IEDDA reaction was observed in the blood for both antibodies, but with trastuzumab, this was to a much lower degree than with cetuximab. Conclusions We could successfully visualize the tumours by using cetuximab and trastuzumab in pretargeted PET imaging despite the challenging circumstances where the antibody is internalized and there is still some unbound antibody circulating in the blood flow. This clearly demonstrates the potential of a pretargeted approach for targeting internalizing antigens and warrants development of pharmacokinetic optimization of the biorthogonal reactants to this end.
- Published
- 2017
- Full Text
- View/download PDF
13. Nanoreporter PET predicts the efficacy of anti-cancer nanotherapy
- Author
-
Carlos Pérez-Medina, Dalya Abdel-Atti, Jun Tang, Yiming Zhao, Zahi A. Fayad, Jason S. Lewis, Willem J. M. Mulder, and Thomas Reiner
- Subjects
Science - Abstract
Nanoparticle drug formulations are currently used as cancer treatment but the response in patients is highly variable. Here, the authors developed a Zirconium-89 nanoreporter able to predict using PET, therapeutic accumulation and efficacy of anti-cancer nanoparticle drug formulations when co-injected in a murine breast cancer model.
- Published
- 2016
- Full Text
- View/download PDF
14. Antibody-Targeted Imaging of Gastric Cancer
- Author
-
Komal Mandleywala, Shayla Shmuel, Patricia M. R. Pereira, and Jason S. Lewis
- Subjects
molecular imaging ,gastric cancer ,PET ,SPECT ,optical imaging ,Organic chemistry ,QD241-441 - Abstract
The specificity of antibodies for antigens overexpressed or uniquely expressed in tumor cells makes them ideal candidates in the development of bioconjugates for tumor imaging. Molecular imaging can aid clinicians in the diagnosis of gastric tumors and in selecting patients for therapies targeting receptors with a heterogeneous intratumoral or intertumoral expression. Antibodies labeled with an imaging radiometal can be used to detect primary tumors and metastases using whole-body positron emission tomography (PET) or single photon emission computed tomography (SPECT), both during diagnosis and monitoring disease response. Conjugated with fluorescent dyes, antibodies can image tumors by targeted optical imaging. This review provides an overview of the most recent advances in the use of antibodies labeled with radiometals or conjugated with fluorescent dyes for gastric cancer imaging.
- Published
- 2020
- Full Text
- View/download PDF
15. Manipulating the In Vivo Behaviour of 68Ga with Tris(Hydroxypyridinone) Chelators: Pretargeting and Blood Clearance
- Author
-
Cinzia Imberti, Pierre Adumeau, Julia E. Blower, Fahad Al Salemee, Julia Baguña Torres, Jason S. Lewis, Brian M. Zeglis, Samantha Y. A. Terry, and Philip J. Blower
- Subjects
metal chelation ,radionuclide imaging ,pretargeting ,gallium-68 ,hydroxypyridinones ,monoclonal antibodies ,bifunctional chelators ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Pretargeting is widely explored in immunoPET as a strategy to reduce radiation exposure of non-target organs and allow the use of short-lived radionuclides that would not otherwise be compatible with the slow pharmacokinetic profiles of antibodies. Here we investigate a pretargeting strategy based on gallium-68 and the chelator THPMe as a high-affinity pair capable of combining in vivo. After confirming the ability of THPMe to bind 68Ga in vivo at low concentrations, the bifunctional THPMe-NCS was conjugated to a humanised huA33 antibody targeting the A33 glycoprotein. Imaging experiments performed in nude mice bearing A33-positive SW1222 colorectal cancer xenografts compared pretargeting (100 μg of THPMe-NCS-huA33, followed after 24 h by 8−10 MBq of 68Ga3+) with both a directly labelled radioimmunoconjugate (89Zr-DFO-NCS-huA33, 88 μg, 7 MBq) and a 68Ga-only negative control (8−10 MBq of 68Ga3+). Imaging was performed 25 h after antibody administration (1 h after 68Ga3+ administration for negative control). No difference between pretargeting and the negative control was observed, suggesting that pretargeting via metal chelation is not feasible using this model. However, significant accumulation of “unchelated” 68Ga3+ in the tumour was found (12.9 %ID/g) even without prior administration of THPMe-NCS-huA33, though tumour-to-background contrast was impaired by residual activity in the blood. Therefore, the 68Ga-only experiment was repeated using THPMe (20 μg, 1 h after 68Ga3+ administration) to clear circulating 68Ga3+, producing a three-fold improvement of the tumour-to-blood activity concentration ratio. Although preliminary, these results highlight the potential of THPMe as a 68Ga clearing agent in imaging applications with gallium citrate.
- Published
- 2020
- Full Text
- View/download PDF
16. Intraoperative Imaging of Positron Emission Tomographic Radiotracers Using Cerenkov Luminescence Emissions
- Author
-
Jason P. Holland, Guillaume Normand, Alessandro Ruggiero, Jason S. Lewis, and Jan Grimm
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
Imaging the location and extent of cancer provides invaluable information before, during, and after surgery. The majority of “image-guided” methods that use, for example, positron emission tomography (PET) involve preoperative imaging and do not provide real-time information during surgery. It is now well established that the inherent optical emissions (Cerenkov radiation) from various β-emitting radionuclides can be visualized by Cerenkov luminescence imaging (CLI). Here we report the full characterization of CLI using the positron-emitting radiotracer 89 Zr-DFO-trastuzumab for target-specific, quantitative imaging of HER2/neu -positive tumors in vivo. We also provide the first demonstration of the feasibility of using CLI for true image-guided, intraoperative surgical resection of tumors. Analysis of optical CLIs provided accurate, quantitative information on radiotracer biodistribution and tissue uptake that correlated well with the concordant PET images. CLI, PET, and biodistribution studies revealed target-specific uptake of 89 Zr-DFO-trastuzumab in BT-474 ( HER2/neu positive) versus MDA-MB-468 ( HER2/neu negative) xenografts in the same mice. Competitive inhibition (blocking) studies followed by CLI also confirmed the in vivo immunoreactivity and specificity of 89 Zr-DFO-trastuzumab for HER2/neu . Overall, these results strongly support the continued development of CLI as a preclinical and possible clinical tool for use in molecular imaging and surgical procedures for accurately defining tumor margins.
- Published
- 2011
- Full Text
- View/download PDF
17. Unconventional Nuclides for Radiopharmaceuticals
- Author
-
Jason P. Holland, Matthew J. Williamson, and Jason S. Lewis
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
Rapid and widespread growth in the use of nuclear medicine for both diagnosis and therapy of disease has been the driving force behind burgeoning research interests in the design of novel radiopharmaceuticals. Until recently, the majority of clinical and basic science research has focused on the development of 11 C-, 13 N-, 15 O-, and 18 F-radiopharmaceuticals for use with positron emission tomography (PET) and 99m Tc-labeled agents for use with single-photon emission computed tomography (SPECT). With the increased availability of small, low-energy cyclotrons and improvements in both cyclotron targetry and purification chemistries, the use of “nonstandard” radionuclides is becoming more prevalent. This brief review describes the physical characteristics of 60 radionuclides, including β + , β −− , γ-ray, and α-particle emitters, which have the potential for use in the design and synthesis of the next generation of diagnostic and/or radiotherapeutic drugs. As the decay processes of many of the radionuclides described herein involve emission of high-energy γ-rays, relevant shielding and radiation safety issues are also considered. In particular, the properties and safety considerations associated with the increasingly prevalent PET nuclides 64 Cu, 68 Ga, 86 Y, 89 Zr, and 124 I are discussed.
- Published
- 2010
- Full Text
- View/download PDF
18. Landscape of prostate-specific membrane antigen heterogeneity and regulation in AR-positive and AR-negative metastatic prostate cancer
- Author
-
Martin K. Bakht, Yasutaka Yamada, Sheng-Yu Ku, Varadha Balaji Venkadakrishnan, Joshua A. Korsen, Teja M. Kalidindi, Kei Mizuno, Shin Hye Ahn, Ji-Heui Seo, Maria Mica Garcia, Francesca Khani, Olivier Elemento, Henry W. Long, Alain Chaglassian, Nagavarakishore Pillarsetty, Jason S. Lewis, Matthew Freedman, Anthony P. Belanger, Quang-De Nguyen, and Himisha Beltran
- Subjects
Cancer Research ,Oncology - Published
- 2023
19. Noninvasive Assessment of Human Epidermal Growth Factor Receptor 2 (HER2) in Esophagogastric Cancer Using89Zr-Trastuzumab PET: A Pilot Study
- Author
-
Melissa A. Lumish, Steven B. Maron, Viktoriya Paroder, Joanne F. Chou, Marinela Capanu, Steven Philemond, Joseph A. O’Donoghue, Heiko Schöder, Jason S. Lewis, Serge K. Lyashchenko, Neeta Pandit-Taskar, and Yelena Y. Janjigian
- Subjects
Radiology, Nuclear Medicine and imaging - Published
- 2022
20. Click Here for Better Chemistry
- Author
-
Brian M, Zeglis and Jason S, Lewis
- Subjects
General Medicine - Published
- 2022
21. Radiotheranostics in oncology: current challenges and emerging opportunities
- Author
-
Lisa Bodei, Ken Herrmann, Heiko Schöder, Andrew M. Scott, and Jason S. Lewis
- Subjects
Oncology ,Neoplasms ,Humans ,Precision Medicine ,Radiopharmaceuticals ,Medical Oncology - Abstract
Structural imaging remains an essential component of diagnosis, staging and response assessment in patients with cancer; however, as clinicians increasingly seek to noninvasively investigate tumour phenotypes and evaluate functional and molecular responses to therapy, theranostics - the combination of diagnostic imaging with targeted therapy - is becoming more widely implemented. The field of radiotheranostics, which is the focus of this Review, combines molecular imaging (primarily PET and SPECT) with targeted radionuclide therapy, which involves the use of small molecules, peptides and/or antibodies as carriers for therapeutic radionuclides, typically those emitting α-, β- or auger-radiation. The exponential, global expansion of radiotheranostics in oncology stems from its potential to target and eliminate tumour cells with minimal adverse effects, owing to a mechanism of action that differs distinctly from that of most other systemic therapies. Currently, an enormous opportunity exists to expand the number of patients who can benefit from this technology, to address the urgent needs of many thousands of patients across the world. In this Review, we describe the clinical experience with established radiotheranostics as well as novel areas of research and various barriers to progress.
- Published
- 2022
22. Click chemistry: a transformative technology in nuclear medicine
- Author
-
David Bauer, Samantha M. Sarrett, Jason S. Lewis, and Brian M. Zeglis
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
23. 89Zr-DFO-Isatuximab for CD38-Targeted ImmunoPET Imaging of Multiple Myeloma and Lymphomas
- Author
-
Natalia Herrero Alvarez, Alexa L. Michel, Tara D. Viray, Marius E. Mayerhoefer, and Jason S. Lewis
- Subjects
General Chemical Engineering ,General Chemistry - Published
- 2023
24. Figure S1 from EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer
- Author
-
Yelena Y. Janjigian, Nikolaus Schultz, Maurizio Scaltriti, Jorge A. Carrasquillo, David B. Solit, Jason S. Lewis, Barry S. Taylor, Elisa de Stanchina, Michael F. Berger, Wolfgang A. Weber, David H. Ilson, Christine A. Iacobuzio-Donahue, Heiko Schöder, Neeta Pandit-Taskar, Steven M. Larson, Richard B. Lanman, Rebecca J. Nagy, Todd Hembrough, Yuan Tian, Fabiola Cecchi, Besnik Qeriqi, Marissa Mattar, Marinela Capanu, Mario E. Lacouture, Karen Brown, Robert A. Lefkowitz, David P. Kelsen, Mark Schattner, Efsevia Vakiani, Joanne Soong, Gouri J. Nanjangud, Nancy Bouvier, Christopher J. Fong, Helen Won, Yaelle Tuvy, Geoffrey Y. Ku, Pau Castel, Jaclyn F. Hechtman, and Francisco Sanchez-Vega
- Abstract
Additional results for dual probe EGFR and ERBB2 FISH from Patients 9, 30 and 32.
- Published
- 2023
25. Supplementary Fig 3 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 3
- Published
- 2023
26. Supplementary Fig 2 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 2
- Published
- 2023
27. Supplementary Materials and Methods, Supplementary 1-7 and Supplementary Table 1. from ERK Inhibition Improves Anti–PD-L1 Immune Checkpoint Blockade in Preclinical Pancreatic Ductal Adenocarcinoma
- Author
-
Jason S. Lewis, Nagavarakishore Pillarsetty, Thomas R. Dilling, Maria Davydova, Ian L. Fox, Adam O. Michel, Mike Cornejo, Veronica L. Nagle, Kyeara N. Mack, and Kelly E. Henry
- Abstract
Supplementary Materials and Methods. Supplementary Figure 1. Fibroblast isolation from orthotopic PDAC tumor microenvironment. Supplementary Figure 2. Controls for immunofluorescence experiments to confirm no cross-reactivity or non-specific binding, and to identify regions to exclude via secondary entrapment. Supplementary Figure 3. Blocking study with [89Zr]Zr-DFO-anti-PD-L1. Supplementary Figure 4. Combination ERK inhibition and anti-PD-L1 does not improve survival in subcutaneous tumors. Supplementary Figure 5. Ex vivo analysis of endpoint tissues suggests immunosuppressive mechanism. Supplementary Figure 6. Immunohistochemical analysis of ex vivo tissues collected at endpoint indicate a potential immunosuppressive mechanism. Supplementary Figure 7. Immunohistochemical analysis of ex vivo tissues collected at single endpoint. Supplementary Table 1. Immunohistochemistry details for TME analysis.
- Published
- 2023
28. Data from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Amplification of and oncogenic mutations in ERBB2, the gene encoding the HER2 receptor tyrosine kinase, promote receptor hyperactivation and tumor growth. Here we demonstrate that HER2 ubiquitination and internalization, rather than its overexpression, are key mechanisms underlying endocytosis and consequent efficacy of the anti-HER2 antibody–drug conjugates (ADC) ado-trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) in lung cancer cell lines and patient-derived xenograft models. These data translated into a 51% response rate in a clinical trial of T-DM1 in 49 patients with ERBB2-amplified or -mutant lung cancers. We show that cotreatment with irreversible pan-HER inhibitors enhances receptor ubiquitination and consequent ADC internalization and efficacy. We also demonstrate that ADC switching to T-DXd, which harbors a different cytotoxic payload, achieves durable responses in a patient with lung cancer and corresponding xenograft model developing resistance to T-DM1. Our findings may help guide future clinical trials and expand the field of ADC as cancer therapy.Significance:T-DM1 is clinically effective in lung cancers with amplification of or mutations in ERBB2. This activity is enhanced by cotreatment with irreversible pan-HER inhibitors, or ADC switching to T-DXd. These results may help address unmet needs of patients with HER2-activated tumors and no approved targeted therapy.See related commentary by Rolfo and Russo, p. 643.This article is highlighted in the In This Issue feature, p. 627
- Published
- 2023
29. Data from ERK Inhibition Improves Anti–PD-L1 Immune Checkpoint Blockade in Preclinical Pancreatic Ductal Adenocarcinoma
- Author
-
Jason S. Lewis, Nagavarakishore Pillarsetty, Thomas R. Dilling, Maria Davydova, Ian L. Fox, Adam O. Michel, Mike Cornejo, Veronica L. Nagle, Kyeara N. Mack, and Kelly E. Henry
- Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) do not benefit from immune checkpoint blockade (ICB) along the PD-1/PD-L1 axis. Variable PD-L1 expression in PDAC indicates a potential access issue of PD-L1–targeted therapy. To monitor target engagement of PD-L1–targeted therapy, we generated a PD-L1–targeted PET tracer labeled with zirconium-89 (89Zr). As the MAPK signaling pathway (MEK and ERK) is known to modulate PD-L1 expression in other tumor types, we used [89Zr]Zr-DFO-anti–PD-L1 as a tool to noninvasively assess whether manipulation of the MAPK signaling cascade could be leveraged to modulate PD-L1 expression and thereby immunotherapeutic outcomes in PDAC. In this study, we observed that the inhibition of MEK or ERK is sufficient to increase PD-L1 expression, which we hypothesized could be leveraged for anti–PD-L1 immune checkpoint therapy. We found that the combination of ERK inhibition and anti–PD-L1 therapy corresponded with a significant improvement of overall survival in a syngeneic mouse model of PDAC. Furthermore, IHC analysis indicates that the survival benefit may be CD8+ T-cell mediated. The therapeutic and molecular imaging tool kit developed could be exploited to better structure clinical trials and address the therapeutic gaps in challenging malignancies such as PDAC.
- Published
- 2023
30. Supplementary Fig 5 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 5
- Published
- 2023
31. Supplementary Tables 1-16, Figures 1-15 from Imaging Androgen Receptor Signaling with a Radiotracer Targeting Free Prostate-Specific Antigen
- Author
-
Charles L. Sawyers, Jason S. Lewis, Hans Lilja, Steven M. Larson, Peter T. Scardino, Per-Anders Abrahamsson, Kim Pettersson, John Wongvipat, Samuel L. Rice, Jason P. Holland, Michael J. Evans, and David Ulmert
- Abstract
PDF file - 1.7MB
- Published
- 2023
32. Supporting Information from Establishment of the In Vivo Efficacy of Pretargeted Radioimmunotherapy Utilizing Inverse Electron Demand Diels-Alder Click Chemistry
- Author
-
Brian M. Zeglis, Jason S. Lewis, Pat B. Zanzonico, Wolfgang W. Scholz, Sean Carlin, Kristen M. Cunanan, Dalya Abdel-Atti, Rosemery Membreno, and Jacob L. Houghton
- Abstract
This document contains all supporting materials and methods as well as supplemental figures and tables.
- Published
- 2023
33. Press Conference from Imaging Androgen Receptor Signaling with a Radiotracer Targeting Free Prostate-Specific Antigen
- Author
-
Charles L. Sawyers, Jason S. Lewis, Hans Lilja, Steven M. Larson, Peter T. Scardino, Per-Anders Abrahamsson, Kim Pettersson, John Wongvipat, Samuel L. Rice, Jason P. Holland, Michael J. Evans, and David Ulmert
- Abstract
mp3 file (37.5 MB). "Personalized Medicine: Late-breaking Science from AACR Journals," Saturday, March 31, 2012. Hosted by Lewis C. Cantley, Co-Editor-in-Chief of Cancer Discovery, this press conference held during the AACR Annual Meeting covers the following topics:Lack of Oxygen Influenced Tumor Behavior and Patient Outcome in Intermediate-risk Prostate CancerNovel Technology Allows for Noninvasive Imaging of Prostate Cancer (Ulmert et al., April issue of Cancer Discovery) EGFR Mutation Unique to Glioblastoma May Explain Lack of Response to Traditional EGFR Inhibitors
- Published
- 2023
34. Supplementary Table 1 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Table 1
- Published
- 2023
35. Supplementary Fig 6 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 6
- Published
- 2023
36. Supplementary Data from Noninvasive Imaging of CD4+ T Cells in Humanized Mice
- Author
-
Jason S. Lewis, Ingo K. Mellinghoff, Andrea Schietinger, Nagavarakishore Pillarsetty, Carl Campos, Maya S. Graham, Kelly E. Henry, Charli Ann J. Hertz, and Veronica L. Nagle
- Abstract
Supplementary Data from Noninvasive Imaging of CD4+ T Cells in Humanized Mice
- Published
- 2023
37. Supplementary Fig 4 from HER2-Mediated Internalization of Cytotoxic Agents in ERBB2 Amplified or Mutant Lung Cancers
- Author
-
Maurizio Scaltriti, Charles M. Rudin, Neal Rosen, Elisa de Stanchina, Gary A. Ulaner, Junji Tsurutani, Ronglai Shen, John T. Poirier, Mark G. Kris, Maria E. Arcila, Pedram Razavi, Jorge S. Reis-Filho, Vicky Makker, Alan L. Ho, Darren J. Buonocore, Jason S. Lewis, David M. Hyman, Fabiola Cecchi, Anuja Bhalkikar, Wei-Li Liao, Sheeno Thyparambil, Helena A. Yu, David R. Jones, James M. Isbell, Michael Offin, Tony Ng, Paul R. Barber, Michael F. Berger, David B. Solit, Nancy U. Lin, Rachel A. Freedman, Irmina Diala, Alshad S. Lalani, Clare J. Wilhem, Gregory Weitsman, Besnik Qeriqi, Megan Little, Inna Khodos, Marissa Mattar, Chongrui Xu, Mackenzie L. Myers, Hai-Yan Tu, Sophie Shifman, Yanyan Cai, Laura Baldino, Emiliano Cocco, Sandra Misale, Flavia Michelini, and Bob T. Li
- Abstract
Supplementary Fig 4
- Published
- 2023
38. Supplementary Figure Legends from EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer
- Author
-
Yelena Y. Janjigian, Nikolaus Schultz, Maurizio Scaltriti, Jorge A. Carrasquillo, David B. Solit, Jason S. Lewis, Barry S. Taylor, Elisa de Stanchina, Michael F. Berger, Wolfgang A. Weber, David H. Ilson, Christine A. Iacobuzio-Donahue, Heiko Schöder, Neeta Pandit-Taskar, Steven M. Larson, Richard B. Lanman, Rebecca J. Nagy, Todd Hembrough, Yuan Tian, Fabiola Cecchi, Besnik Qeriqi, Marissa Mattar, Marinela Capanu, Mario E. Lacouture, Karen Brown, Robert A. Lefkowitz, David P. Kelsen, Mark Schattner, Efsevia Vakiani, Joanne Soong, Gouri J. Nanjangud, Nancy Bouvier, Christopher J. Fong, Helen Won, Yaelle Tuvy, Geoffrey Y. Ku, Pau Castel, Jaclyn F. Hechtman, and Francisco Sanchez-Vega
- Abstract
Supplementary Figure Legends
- Published
- 2023
39. Data from Imaging of Cancer γ-Secretase Activity Using an Inhibitor-Based PET Probe
- Author
-
Yue-Ming Li, Naga Vara Kishore Pillarsetty, Jason S. Lewis, Lukas M. Carter, Blesida Punzalan, Kelly E. Henry, Georgia Frost, George P. Liao, Thomas Li, Xianzhong Wu, Veronica L. Nagle, Teja Kalidindi, and Pengju Nie
- Abstract
Purpose:Abnormal Notch signaling promotes cancer cell growth and tumor progression in various cancers. Targeting γ-secretase, a pivotal regulator in the Notch pathway, has yielded numerous γ-secretase inhibitors (GSIs) for clinical investigation in the last 2 decades. However, GSIs have demonstrated minimal success in clinical trials in part due to the lack of specific and precise tools to assess γ-secretase activity and its inhibition in vivo.Experimental Design:We designed an imaging probe based on GSI Semagacestat structure and synthesized the radioiodine-labeled analogues [131I]- or [124I]-PN67 from corresponding trimethyl-tin precursors. Both membrane- and cell-based ligand-binding assays were performed using [131I]-PN67 to determine the binding affinity and specificity for γ-secretase in vitro. Moreover, we evaluated [124I]-PN67 by PET imaging in mammary tumor and glioblastoma mouse models.Results:The probe was synthesized through iodo-destannylation using chloramine-T as an oxidant with a high labeling yield and efficiency. In vitro binding results demonstrate the high specificity of this probe and its ability for target replacement study by clinical GSIs. PET imaging studies demonstrated a significant (P < 0.05) increased in the uptake of [124I]-PN67 in tumors versus blocking or sham control groups across multiple mouse models, including 4T1 allograft, MMTV-PyMT breast cancer, and U87 glioblastoma allograft. Ex vivo biodistribution and autoradiography corroborate these results, indicating γ-secretase specific tumor accumulation of [124I]-PN67.Conclusions:[124I]-PN67 is a novel PET imaging agent that enables assessment of γ-secretase activity and target engagement of clinical GSIs.
- Published
- 2023
40. Supplementary Data from Retooling a Blood-Based Biomarker: Phase I Assessment of the High-Affinity CA19-9 Antibody HuMab-5B1 for Immuno-PET Imaging of Pancreatic Cancer
- Author
-
Wolfgang A. Weber, Jason S. Lewis, Paul W. Maffuid, Wolfgang Scholz, Rebecca Teng, Shutian Ruan, Serge K. Lyashchenko, Jorge A. Carrasquillo, Neeta Pandit-Taskar, Joseph A. O'Donoghue, Eileen M. O'Reilly, and Christian Lohrmann
- Abstract
Supplementary Data
- Published
- 2023
41. Data from iNOS Regulates the Therapeutic Response of Pancreatic Cancer Cells to Radiotherapy
- Author
-
Jason S. Lewis, Taha Merghoub, Christine A. Iacobuzio-Donahue, Abu-Akeel Mohsen, Lolkje Abma, Mike Cornejo, Luis Felipe Campesato, Freddy E. Escorcia, Lukas M. Carter, Komal Mandleywala, Kimberly J. Edwards, and Patricia M.R. Pereira
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly resistant to radiotherapy, chemotherapy, or a combination of these modalities, and surgery remains the only curative intervention for localized disease. Although cancer-associated fibroblasts (CAF) are abundant in PDAC tumors, the effects of radiotherapy on CAFs and the response of PDAC cells to radiotherapy are unknown. Using patient samples and orthotopic PDAC biological models, we showed that radiotherapy increased inducible nitric oxide synthase (iNOS) in the tumor tissues. Mechanistic in vitro studies showed that, although undetectable in radiotherapy-activated tumor cells, iNOS expression and nitric oxide (NO) secretion were significantly increased in CAFs secretome following radiotherapy. Culture of PDAC cells with conditioned media from radiotherapy-activated CAFs increased iNOS/NO signaling in tumor cells through NF-κB, which, in turn, elevated the release of inflammatory cytokines by the tumor cells. Increased NO after radiotherapy in PDAC contributed to an acidic microenvironment that was detectable using the radiolabeled pH (low) insertion peptide (pHLIP). In murine orthotopic PDAC models, pancreatic tumor growth was delayed when iNOS inhibition was combined with radiotherapy. These data show the important role that iNOS/NO signaling plays in the effectiveness of radiotherapy to treat PDAC tumors.Significance:A radiolabeled pH-targeted peptide can be used as a PET imaging tool to assess therapy response within PDAC and blocking iNOS/NO signaling may improve radiotherapy outcomes.
- Published
- 2023
42. Supplementary Figures 1-5 from Phase I Trial of Well-Differentiated Neuroendocrine Tumors (NETs) with Radiolabeled Somatostatin Antagonist 177Lu-Satoreotide Tetraxetan
- Author
-
Wolfgang A. Weber, Lisa Bodei, Christian Lohrmann, Mithat Gönen, Nitya Raj, Jason S. Lewis, Serge K. Lyashchenko, Kevin D. Staton, Simone Krebs, Joseph A. O'Donoghue, Neeta Pandit-Taskar, and Diane Reidy-Lagunes
- Abstract
Supplementary Figures 1-5
- Published
- 2023
43. Supplementary Figure from Radioimmunotherapy Targeting Delta-like Ligand 3 in Small Cell Lung Cancer Exhibits Antitumor Efficacy with Low Toxicity
- Author
-
Jason S. Lewis, Charles M. Rudin, John T. Poirier, Nagavarakishore Pillarsetty, Triparna Sen, William D. Travis, Alessandra Piersigilli, Avelyn Mae Delos Reyes, Joshua A. Korsen, Komal Mandleywala, Zachary V. Samuels, Sai Kiran Sharma, Lukas M. Carter, Salomon Tendler, and Kathryn M. Tully
- Abstract
Supplementary Figure from Radioimmunotherapy Targeting Delta-like Ligand 3 in Small Cell Lung Cancer Exhibits Antitumor Efficacy with Low Toxicity
- Published
- 2023
44. Supplementary Data from iNOS Regulates the Therapeutic Response of Pancreatic Cancer Cells to Radiotherapy
- Author
-
Jason S. Lewis, Taha Merghoub, Christine A. Iacobuzio-Donahue, Abu-Akeel Mohsen, Lolkje Abma, Mike Cornejo, Luis Felipe Campesato, Freddy E. Escorcia, Lukas M. Carter, Komal Mandleywala, Kimberly J. Edwards, and Patricia M.R. Pereira
- Abstract
Supplementary Methods Supplementary Figure 1. iNOS staining in non-tumor and PDAC tumors obtained before treatment and after RT. Supplementary Figure 2. Western blot of iNOS in the total lysates of FC1245 orthotopic PDAC tumors obtained 2 days post-RT therapy. Supplementary Figure 3. Immunohistochemistry of trichrome in FC1245 orthotopic PDAC tumors obtained at 2 days post-RT therapy. Supplementary Figure 4. CAFs isolation and expansion from FC1245 orthotopic PDAC tumors. Supplementary Figure 5. Timeline for FC1245 culture with CM from CAFs or co-cultured with CAFs. Supplementary Figure 6. Cell growth of FC1245 cultured in the presence or absence of conditioned media from irradiated CAFs or co-cultured with irradiated CAFs. Supplementary Figure 7. NO2- levels in supernatants of human pancreatic cancer cell lines in the presence and absence of supernatants from human CAFs and treated with iNOS inhibitor 1400W. Supplementary Figure 8. IL-6, LIF, RANTES, and IL-13 concentrations in the supernatants of FC1245 PDAC cells, FC1245 incubated with conditioned medium from non-RT CAFs, and FC1245 incubated with conditioned medium from RT CAFs. Supplementary Figure 9. Gene expression heatmap of non-RT and RT fibroblasts. Supplementary Figure 10. Confocal images and quantification (mean {plus minus} S.E.M, n = 3) of immunofluorescence staining of α-SMA (green) in FC1245 orthotopic PDAC tumors at 14 days after cells' implantation. Supplementary Figure 11. FC1245 migration images and quantification using the wound healing assay. Supplementary Figure 12. Liver and pancreatic tumors at 6 days after surgery. Supplementary Figure 13. Liver, stomach, spleen, and pancreatic tumors at 14 days after surgery. Supplementary Figure 14. FC1245 cell growth and NO2- levels in supernatants of FC1245 with or without DETA-NONOate, 1400W or with the combination of DETA-NONOate/1400W. Supplementary Figure 15. FC1245 cell growth and NO2- levels in supernatants of FC1245 transfected with iNOS CRISPR activation plasmid or control CRISPR activation plasmid. Supplementary Figure 16. Cell growth of FC1245 cultured in the presence or absence of irradiated and non-irradiated CAFs. Supplementary Figure 17. Survival of mice implanted with FC1245 orthotopic PDAC tumors and treated with RT. Supplementary Figure 18. Tumor weight in PDAC tumors from RT, 1400W, and 1400W/RT treated mice on day 14 post-therapy. Supplementary Figure 19. Glucose uptake and lactate production of FC1245 PDAC cells treated with or without DETA-NO, 1400W, and the combination of DETA-NO/1400W. Supplementary Figure 20. Biodistribution data at 2 h after injection of 18F-FDG in mice bearing FC1245 orthotopic tumors. Supplementary Figure 21. Biodistribution data at 24 h after injection of 67Ga-labeled pHLIP in mice bearing FC1245 orthotopic tumors. Supplementary Table 1. RT administrations, time-points, and human PDAC sample collection time-points. Supplementary Table 2. Quantification of iNOS+/α-SMA+ in PDAC RT-treated samples obtained from patient #3. Supplementary Table 3. Quantification of iNOS+/cytokeratin+ in PDAC RT-treated samples obtained from patient #3. Supplementary Table 4. Quantification of iNOS+/α-SMA+ in PDAC CT samples obtained from FC1245 orthotopic tumors. Supplementary Table 5. Quantification of iNOS+/α-SMA+ in PDAC RT-treated samples obtained from FC1245 orthotopic tumors. Supplementary Table 6. Quantification of iNOS+/cytokeratin+ in PDAC CT samples obtained from FC1245 orthotopic tumors. Supplementary Table 7. Quantification of iNOS+/cytokeratin+ in PDAC RT-treated samples obtained from FC1245 orthotopic tumors. Supplementary Table 8. Quantification of GLUT1+/α-SMA+ in PDAC CT samples obtained from FC1245 orthotopic tumors. Supplementary Table 9. Quantification of GLUT1+/α-SMA+ in PDAC RT-treated samples obtained from FC1245 orthotopic tumors. Supplementary Table 10. Quantification of GLUT1+/cytokeratin+ in PDAC CT samples obtained from FC1245 orthotopic tumors. Supplementary Table 11. Quantification of GLUT1+/cytokeratin+ in PDAC RT-treated samples obtained from FC1245 orthotopic tumors.
- Published
- 2023
45. Supplementary Methods from Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation
- Author
-
Charles L. Sawyers, Jason S. Lewis, Donald J. Tindall, William Polkinghorn, George G. Klee, Kathryn E. Carnazza, John Wongvipat, Neel Shah, Man Xia Lee, Michael G. Doran, Brian J. Davis, Michael J. Evans, and Daniel E. Spratt
- Abstract
Supplementary Methods Additional methodology of experimental methods.
- Published
- 2023
46. Data from Retooling a Blood-Based Biomarker: Phase I Assessment of the High-Affinity CA19-9 Antibody HuMab-5B1 for Immuno-PET Imaging of Pancreatic Cancer
- Author
-
Wolfgang A. Weber, Jason S. Lewis, Paul W. Maffuid, Wolfgang Scholz, Rebecca Teng, Shutian Ruan, Serge K. Lyashchenko, Jorge A. Carrasquillo, Neeta Pandit-Taskar, Joseph A. O'Donoghue, Eileen M. O'Reilly, and Christian Lohrmann
- Abstract
Purpose:In patients with cancer who have an abnormal biomarker finding, the source of the biomarker in the bloodstream must be located for confirmation of diagnosis, staging, and therapy planning. We evaluated if immuno-PET with the radiolabeled high-affinity antibody HuMab-5B1 (MVT-2163), binding to the cancer antigen CA19-9, can identify the source of elevated biomarkers in patients with pancreatic cancer.Patients and Methods:In this phase I dose-escalating study, 12 patients with CA19-9–positive metastatic malignancies were injected with MVT-2163. Within 7 days, all patients underwent a total of four whole-body PET/CT scans. A diagnostic CT scan was performed prior to injection of MVT-2163 to correlate findings on MVT-2163 PET/CT.Results:Immuno-PET with MVT-2163 was safe and visualized known primary tumors and metastases with high contrast. In addition, radiotracer uptake was not only observed in metastases known from conventional CT, but also seen in subcentimeter lymph nodes located in typical metastatic sites of pancreatic cancer, which were not abnormal on routine clinical imaging studies. A significant fraction of the patients demonstrated very high and, over time, increased uptake of MVT-2163 in tumor tissue, suggesting that HuMab-5B1 labeled with beta-emitting radioisotopes may have the potential to deliver therapeutic doses of radiation to cancer cells.Conclusions:Our study shows that the tumor antigen CA19-9 secreted to the circulation can be used for sensitive detection of primary tumors and metastatic disease by immuno-PET. This significantly broadens the number of molecular targets that can be used for PET imaging and offers new opportunities for noninvasive characterization of tumors in patients.
- Published
- 2023
47. Figure S1 from Imaging of Cancer γ-Secretase Activity Using an Inhibitor-Based PET Probe
- Author
-
Yue-Ming Li, Naga Vara Kishore Pillarsetty, Jason S. Lewis, Lukas M. Carter, Blesida Punzalan, Kelly E. Henry, Georgia Frost, George P. Liao, Thomas Li, Xianzhong Wu, Veronica L. Nagle, Teja Kalidindi, and Pengju Nie
- Abstract
Supplementary Figure 1. The radiolabeling product of [124I]PN67 was confirmed by co-injecting cold compound 2 (100uM in 100ul DMSO/H2O/MeCN) with the radioactive fraction (50uCi in 100ul H2O/MeCN) onto HPLC. (A) UV 235/254nm wavelength channels were used to detect absorption of cold PN67. Retention time (RT): 8.17min; (B) The radioactivity was detected by a bioscan flow-count detector. Retention time (RT): 8.28min. Radiochemical purity (>99%) was calculated based on integration of area under curve. The bioscan peak is slightly delayed compared with UV signal because of instrument setting.
- Published
- 2023
48. Supplementary Figure 2 from Androgen Receptor Upregulation Mediates Radioresistance after Ionizing Radiation
- Author
-
Charles L. Sawyers, Jason S. Lewis, Donald J. Tindall, William Polkinghorn, George G. Klee, Kathryn E. Carnazza, John Wongvipat, Neel Shah, Man Xia Lee, Michael G. Doran, Brian J. Davis, Michael J. Evans, and Daniel E. Spratt
- Abstract
Supplementary Figure 2. A) In vitro comparison of LNCaP and LNCaP-AR and DNA damage post-radiotherapy. B) In vivo correlation of baseline AR-output using bioluminescence to tumor progression.
- Published
- 2023
49. Supplementary Data from Noninvasive Interrogation of DLL3 Expression in Metastatic Small Cell Lung Cancer
- Author
-
Jason S. Lewis, John T. Poirier, Scott J. Dylla, Charles M. Rudin, Brian M. Zeglis, David Liu, Vikram Sisodiya, Viola Allaj, Eric B. Johansen, Kristen M. Cunanan, Joseph Sandoval, Sheila Bheddah, Kumiko Isse, Omar Hamdy, Eric E. Gardner, Alexander J. Bankovich, Alessandra Piersigilli, Sean D. Carlin, Dalya Abdel-Atti, Jacob Pourat, and Sai Kiran Sharma
- Abstract
Supplemental data detailing the methods and results for the synthesis and in vitro characterization of the radioimmunoconjugates, longitudinal PET imaging and ex vivo analyses in various models described within the main text.
- Published
- 2023
50. Table S1 from Imaging of Cancer γ-Secretase Activity Using an Inhibitor-Based PET Probe
- Author
-
Yue-Ming Li, Naga Vara Kishore Pillarsetty, Jason S. Lewis, Lukas M. Carter, Blesida Punzalan, Kelly E. Henry, Georgia Frost, George P. Liao, Thomas Li, Xianzhong Wu, Veronica L. Nagle, Teja Kalidindi, and Pengju Nie
- Abstract
Supplementary Table 1. Biodistribution data of [124I]PN67 versus time/h, administrated by i.v. tail-vein injection in BALB/c mice bearing 4T1-Luc tumors. Data is represented as average %ID/g {plus minus} SD with n=5 animals/group.
- Published
- 2023
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.