1
|
Aguirre N, Veach DR, Cercek A, Cheal SM, Larson SM, Nash GM, Cheung NKV. Radioimmunotherapy for peritoneal carcinomatosis: Preclinical proof of concept to clinical translation. Cell Rep Med 2025; 6:102040. [PMID: 40154493 PMCID: PMC12047513 DOI: 10.1016/j.xcrm.2025.102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 01/09/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Peritoneal carcinomatosis (PC), characterized by the dissemination of metastatic tumor cells throughout the peritoneal cavity from several gastrointestinal and gynecological malignancies, has significantly compromised patient survival. The standard of care is cytoreductive surgery with or without intraperitoneal chemotherapy. However, surgical resection often leaves behind microscopic or clinically occult disease due to the complex anatomy of the peritoneum, where intraperitoneal chemotherapy and systemic chemotherapy have shown limited success. To improve the therapeutic outcome, targeted therapy using radionuclides such as alpha, beta, and Auger emitters delivered by antibodies is actively being investigated. While preclinical murine models of PC have shown the potential of radioimmunotherapy (RIT) using various radioisotopes across a wide spectrum of antigen targets and tumor diagnoses with acceptable toxicities, successful clinical trials are lacking. Here, we retrospectively summarize preclinical and clinical PC studies, consider their translational potential, and examine paths to development that maximize the clinical benefit of RIT in this context.
Collapse
Affiliation(s)
- Nicole Aguirre
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Darren R Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Cercek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah M Cheal
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Garrett M Nash
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nai-Kong V Cheung
- Pediatric Oncology, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2
|
Calistri S, Ottaviano G, Ubaldini A. Radiopharmaceuticals for Pancreatic Cancer: A Review of Current Approaches and Future Directions. Pharmaceuticals (Basel) 2024; 17:1314. [PMID: 39458955 PMCID: PMC11510189 DOI: 10.3390/ph17101314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The poor prognosis of pancreatic cancer requires novel treatment options. This review examines the evolution of radiopharmaceuticals in the treatment of pancreatic cancer. Established strategies such as peptide receptor radionuclide therapy (PRRT) offer targeted and effective treatment, compared to conventional treatments. However, there are currently no radiopharmaceuticals approved for the treatment of pancreatic cancer in Europe, which requires further research and novel approaches. New radiopharmaceuticals including radiolabeled antibodies, peptides, and nanotechnological approaches are promising in addressing the challenges of pancreatic cancer therapy. These new agents may offer more specific targeting and potentially improve efficacy compared to traditional therapies. Further research is needed to optimize efficacy, address limitations, and explore the overall potential of these new strategies in the treatment of this aggressive and harmful pathology.
Collapse
Affiliation(s)
- Sara Calistri
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, C.R. Bologna, Via Martiri di Monte Sole 4, 40129 Bologna, Italy; (G.O.); (A.U.)
| | - Giuseppe Ottaviano
- ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, C.R. Bologna, Via Martiri di Monte Sole 4, 40129 Bologna, Italy; (G.O.); (A.U.)
| | - Alberto Ubaldini
- ENEA, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, C.R. Bologna, Via Martiri di Monte Sole 4, 40129 Bologna, Italy; (G.O.); (A.U.)
| |
Collapse
|
3
|
Nguyen TMC, Hoang LDC, Nguyen TKG, Nguyen TN, Nguyen QC, Nguyen TB, Dang HHQ, Bui VC, Pham TM, Nguyen TT. Safety assessment, radioiodination and preclinical evaluation of antinuclear antibody as novel medication for prostate cancer in mouse xenograft model. Sci Rep 2023; 13:18753. [PMID: 37907691 PMCID: PMC10618443 DOI: 10.1038/s41598-023-45984-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023] Open
Abstract
This study aims to provide in vitro and in vivo data to support the utilization of antinuclear antibodies (ANAs) as novel tools for the diagnosis and treatment of prostate cancers. The hematological, biochemical, and histological toxicities of ANAs were assessed at the doses of 5 and 50 μg per mouse. Radiolabeling study was then conducted with ANA and 131I using the chloramine T method, and the biodistribution and treatment efficacy were subsequently investigated in a PC3 xenograft model. No changes in clinical behavior or signs of intoxication, necrosis, or malignancy were observed in ANA-treated mice. 131I-ANA was obtained in very high yield and radiochemical purity, at 94.97 ± 0.98% and 98.56 ± 0.29%, respectively. They achieved immunoreactivity fraction of 0.841 ± 0.17% with PC-3 cells. Levels of radiolabeled ANAs were 1.15-10.14 times higher in tumor tissues than in other examined organs at 24 h post-injection. The tumor growth inhibition rates were 28.33 ± 5.01% in PC3 xenografts mice treated with 131I-ANAs compared with controls and a nearly twofold improvement in median survival was observed. These results demonstrate that radioimmunotherapy of radiolabeled natural ANAs may be an effective treatment for prostate tumors.
Collapse
Affiliation(s)
- Thu Minh Chau Nguyen
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
- Hanoi Medical University, Hanoi, Vietnam
| | | | - Thi Khanh Giang Nguyen
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Thi Ngoc Nguyen
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Quang Chien Nguyen
- Department of Hematology and Blood Transfusion, Vietnam Military Medical University, Military Hospital 103, Hanoi, Vietnam
| | - Thanh Binh Nguyen
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Ho Hong Quang Dang
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Van Cuong Bui
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Thanh Minh Pham
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam
| | - Thi Thu Nguyen
- Center for Research and Production of Radioisotopes, Nuclear Research Institute, 01 Nguyen Tu Luc Street, Dalat, Lam-Dong, Vietnam.
| |
Collapse
|
4
|
Yoshii Y, Matsumoto H, Igarashi C, Tachibana T, Hihara F, Shinada M, Waki A, Yoshida S, Naito K, Ito K, Higashi T, Kurihara H, Ueno M. Process to Remove the Size Variants Contained in the Antibody-Chelator Complex PCTA-NCAB001 for Radiolabeling with Copper-64. Pharmaceuticals (Basel) 2023; 16:1341. [PMID: 37895812 PMCID: PMC10610008 DOI: 10.3390/ph16101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Understanding the physicochemical properties of antibody-drug conjugates is critical to assess their quality at manufacturing and monitor them during subsequent storage. For radiometal-antibody complexes, it is important to control the properties of the antibody-chelator conjugate to maintain the quality of the final product. We have been developing 64Cu-labeled anti-epidermal growth factor receptor antibody NCAB001 (64Cu-NCAB001) for the early diagnosis and therapy of pancreatic cancer with positron-emission tomography. Here, we characterized the larger size variants contained in the antibody-chelator conjugate PCTA-NCAB001 by multi-angle light scattering coupled with size-exclusion chromatography. Secondly, we developed a chromatographic method to remove these size variants. Lastly, we demonstrated the stability of PCTA-NCAB001 after the removal of size variants. Dimer and oligomers were identified in PCTA-NCAB001. These larger size variants, together with some smaller size variants, could be removed by hydrophobic interaction chromatography. The PCTA-NCAB001 product, after the removal of these size variants, could be stored at 4 °C for six months. The methods developed here can be applied to assure the quality of PCTA-NCAB001 and other antibody-drug conjugates to facilitate the development of antibody-radiometal conjugates for positron-emission tomography and radioimmunotherapy of malignant cancers.
Collapse
Affiliation(s)
- Yukie Yoshii
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Hiroki Matsumoto
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Chika Igarashi
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Tomoko Tachibana
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Biology, Graduate School of Science, Toho University, Chiba 274-8510, Japan
| | - Fukiko Hihara
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Mitsuhiro Shinada
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Chemistry, Graduate School of Science, Toho University, Chiba 274-8510, Japan
| | - Atsuo Waki
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Sei Yoshida
- Department of Research, NanoCarrier Co., Ltd., Tokyo 104-0031, Japan; (S.Y.); (K.N.)
| | - Kenichiro Naito
- Department of Research, NanoCarrier Co., Ltd., Tokyo 104-0031, Japan; (S.Y.); (K.N.)
| | - Kimiteru Ito
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Tatsuya Higashi
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| |
Collapse
|
5
|
Duan H, Li L, He S. Advances and Prospects in the Treatment of Pancreatic Cancer. Int J Nanomedicine 2023; 18:3973-3988. [PMID: 37489138 PMCID: PMC10363367 DOI: 10.2147/ijn.s413496] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
Pancreatic cancer is a highly malignant and incurable disease, characterized by its aggressive nature and high fatality rate. The most common type is pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis and high mortality rate. Current treatments for pancreatic cancer mainly encompass surgery, chemotherapy, radiotherapy, targeted therapy, and combination regimens. However, despite efforts to improve prognosis, and the 5-year survival rate for pancreatic cancer remains very low. Therefore, it's urgent to explore novel therapeutic approaches. With the rapid development of therapeutic strategies in recent years, new ideas have been provided for treating pancreatic cancer. This review expositions the advancements in nano drug delivery system, molecular targeted drugs, and photo-thermal treatment combined with nanotechnology for pancreatic cancer. It comprehensively analyzes the prospects of combined drug delivery strategies for treating pancreatic cancer, aiming at a deeper understanding of the existing drugs and therapeutic approaches, promoting the development of new therapeutic drugs, and attempting to enhance the therapeutic effect for patients with this disease.
Collapse
Affiliation(s)
- Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People’s Republic of China
| | - Li Li
- Department of Hepatobiliary Pancreatic Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, People’s Republic of China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, People’s Republic of China
| |
Collapse
|
6
|
Boyle AJ, Cai Z, O'Brien S, Crick J, Angers S, Reilly RM. Relative Biological Effectiveness (RBE) of [ 64Cu]Cu and [ 177Lu]Lu-NOTA-panitumumab F (ab') 2 radioimmunotherapeutic agents vs. γ-radiation for decreasing the clonogenic survival in vitro of human pancreatic ductal adenocarcinoma (PDAC) cells. Nucl Med Biol 2023; 122-123:108367. [PMID: 37506639 DOI: 10.1016/j.nucmedbio.2023.108367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
INTRODUCTION Our objective was to compare [64Cu]Cu-NOTA-panitumumab F(ab')2 and [177Lu]Lu-NOTA-panitumumab F(ab')2 radioimmunotherapy (RIT) agents for decreasing the clonogenic survival fraction (SF) in vitro of EGFR-positive human pancreatic ductal adenocarcinoma (PDAC) cell lines and estimate the relative biological effectiveness (RBE) vs. γ-radiation (XRT). METHODS EGFR-positive PDAC cell lines (AsPC-1, PANC-1, MIAPaCa-2, Capan-1) and EGFR-knockout PANC-1 EGFR KO cells were treated in vitro for 18 h with (0-19.65 MBq; 72 nmols/L) of [64Cu]Cu-NOTA-panitumumab F(ab')2 or [177Lu]Lu-NOTA-panitumumab F(ab')2 or XRT (0-8 Gy) followed by clonogenic assay. The SF was determined after culturing single treated cells for 14 d. Cell fractionation studies were performed for cells incubated with 1 MBq (72 nmols/L) of [64Cu]Cu-NOTA-panitumumab F(ab')2 or [177Lu]Lu-NOTA-panitumumab F(ab')2 for 1, 4, or 24 h to estimate the time-integrated activity (Ã) on the cell surface, cytoplasm, nucleus and medium. Radiation absorbed doses in the nucleus were calculated by multiplying à by S-factors calculated by Monte Carlo N Particle (MCNP) modeling using monolayer cell culture geometry. The SF of PDAC cells was plotted vs. dose and fitted to a linear quadratic model to estimate the dose required to decrease the SF to 0.1 (D10). The D10 for RIT agents were compared to XRT to estimate the RBE. DNA double-strand breaks (DSBs) caused by [64Cu]Cu-NOTA-panitumumab F(ab')2 or [177Lu]Lu-NOTA-panitumumab F(ab')2 continuous exposure for 5 h or 20 h were probed by immunofluorescence for γ-H2AX. Relative EGFR expression of PDAC cells was assessed by flow cytometry (scored + to +++) and cell doubling times for untreated cells were determined. RESULTS The D10 for [64Cu]Cu-NOTA-panitumumab F(ab')2 ranged from 9.1 Gy (PANC-1) to 39.9 Gy (Capan-1). The D10 for [177Lu]Lu-NOTA-panitumumab F(ab')2 ranged from 11.7 Gy (AsPC-1) to 170.8 Gy (Capan-1). The D10 for XRT ranged from 2.5 Gy (Capan-1) to 6.7 Gy (PANC-1 EGFR KO). D10 values were not correlated with EGFR expression over a relatively narrow range (++ to +++) or with cell doubling times. Based on D10 values, PANC-1 EGFR KO cells were 1.6-fold less sensitive than PANC-1 cells to [64Cu]Cu-NOTA-panitumumab F(ab')2 and 1.9-fold less sensitive to [177Lu]Lu-NOTA-panitumumab F(ab')2. The RBE for [64Cu]Cu-NOTA-panitumumab F(ab')2 ranged from 0.06 for Capan-1 cells to 0.45 for PANC-1 cells. The RBE for [177Lu]Lu-NOTA-panitumumab F(ab')2 ranged from 0.015 for Capan-1 cells to 0.28 for AsPC-1 cells. DNA DSBs were detected in PDAC cells exposed to [64Cu]Cu-NOTA-panitumumab F(ab')2 or [177Lu]Lu-NOTA-panitumumab F(ab')2 but were not correlated with the SF of the cells. CONCLUSIONS We conclude that at the same dose delivered to the cell nucleus [64Cu]Cu-NOTA-panitumumab F(ab')2 and [177Lu]Lu-NOTA-panitumumab F(ab')2 were less radiobiologically effective than XRT for decreasing the SF of human PDAC cells, but [64Cu]Cu-NOTA-panitumumab F(ab')2 was more cytotoxic than [177Lu]Lu-NOTA-panitumumab F(ab')2 except for AsPC-1 cells which were more sensitive to [177Lu]Lu-NOTA-panitumumab F(ab')2. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE This study demonstrates that higher radiation doses may be required for RIT than XRT to achieve radiobiologically equivalent effects when used to treat PDAC.
Collapse
Affiliation(s)
- Amanda J Boyle
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Siobhan O'Brien
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jennifer Crick
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON M5S 3M2, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON M5T 1W7, Canada.
| |
Collapse
|
7
|
Métivier C, Le Saëc P, Gaschet J, Chauvet C, Marionneau-Lambot S, Hofgaard PO, Bogen B, Pineau J, Le Bris N, Tripier R, Alliot C, Haddad F, Chérel M, Chouin N, Faivre-Chauvet A, Rbah-Vidal L. Preclinical Evaluation of a 64Cu-Based Theranostic Approach in a Murine Model of Multiple Myeloma. Pharmaceutics 2023; 15:1817. [PMID: 37514004 PMCID: PMC10385603 DOI: 10.3390/pharmaceutics15071817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Although the concept of theranostics is neither new nor exclusive to nuclear medicine, it is a particularly promising approach for the future of nuclear oncology. This approach is based on the use of molecules targeting specific biomarkers in the tumour or its microenvironment, associated with optimal radionuclides which, depending on their emission properties, allow the combination of diagnosis by molecular imaging and targeted radionuclide therapy (TRT). Copper-64 has suitable decay properties (both β+ and β- decays) for PET imaging and potentially for TRT, making it both an imaging and therapy agent. We developed and evaluated a theranostic approach using a copper-64 radiolabelled anti-CD138 antibody, [64Cu]Cu-TE1PA-9E7.4 in a MOPC315.BM mouse model of multiple myeloma. PET imaging using [64Cu]Cu-TE1PA-9E7.4 allows for high-resolution PET images. Dosimetric estimation from ex vivo biodistribution data revealed acceptable delivered doses to healthy organs and tissues, and a very encouraging tumour absorbed dose for TRT applications. Therapeutic efficacy resulting in delayed tumour growth and increased survival without inducing major or irreversible toxicity has been observed with 2 doses of 35 MBq administered at a 2-week interval. Repeated injections of [64Cu]Cu-TE1PA-9E7.4 are safe and can be effective for TRT application in this syngeneic preclinical model of MM.
Collapse
Affiliation(s)
- Cassandra Métivier
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Patricia Le Saëc
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Joëlle Gaschet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | - Catherine Chauvet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| | | | - Peter O Hofgaard
- Department of Immunology, Oslo University Hospital, 04024 Oslo, Norway
| | - Bjarne Bogen
- Department of Immunology, Oslo University Hospital, 04024 Oslo, Norway
| | - Julie Pineau
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Nathalie Le Bris
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Raphaël Tripier
- Univ. Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France
| | - Cyrille Alliot
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
- GIP ARRONAX, 44800 Saint-Herblain, France
| | - Férid Haddad
- IMT Atlantique, Nantes Université, Subatech, 44307 Nantes, France
- GIP ARRONAX, 44800 Saint-Herblain, France
| | - Michel Chérel
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
- Nuclear Medicine Department, ICO-René Gauducheau Cancer Center, 44800 Saint-Herblain, France
| | - Nicolas Chouin
- Nantes Université, Inserm, CNRS, Université d'Angers, Oniris, CRCI2NA, 44007 Nantes, France
| | - Alain Faivre-Chauvet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, CHU Nantes, 44007 Nantes, France
| | - Latifa Rbah-Vidal
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44007 Nantes, France
| |
Collapse
|
8
|
Hull A, Li Y, Bartholomeusz D, Hsieh W, Tieu W, Pukala TL, Staudacher AH, Bezak E. Preliminary Development and Testing of C595 Radioimmunoconjugates for Targeting MUC1 Cancer Epitopes in Pancreatic Ductal Adenocarcinoma. Cells 2022; 11:cells11192983. [PMID: 36230945 PMCID: PMC9563759 DOI: 10.3390/cells11192983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Mucin 1 is a transmembrane glycoprotein which overexpresses cancer-specific epitopes (MUC1-CE) on pancreatic ductal adenocarcinoma (PDAC) cells. As PDAC is a low survival and highly aggressive malignancy, developing radioimmunoconjugates capable of targeting MUC1-CE could lead to improvements in PDAC outcomes. The aim of this study was to develop and perform preliminary testing of diagnostic and therapeutic radioimmunoconjugates for PDAC using an anti-MUC1 antibody, C595. Firstly, p-SCN-Bn-DOTA was conjugated to the C595 antibody to form a DOTA-C595 immunoconjugate. The stability and binding affinity of the DOTA-C595 conjugate was evaluated using mass spectrometry and ELISA. DOTA-C595 was radiolabelled to Copper-64, Lutetium-177, Gallium-68 and Technetium-99m to form novel radioimmunoconjugates. Cell binding assays were performed in PANC-1 (strong MUC1-CE expression) and AsPC-1 (weak MUC1-CE expression) cell lines using 64Cu-DOTA-C595 and 177Lu-DOTA-C595. An optimal molar ratio of 4:1 DOTA groups per C595 molecule was obtained from the conjugation process. DOTA-C595 labelled to Copper-64, Lutetium-177, and Technetium-99m with high efficiency, although the Gallium-68 labelling was low. 177Lu-DOTA-C595 demonstrated high cellular binding to the PANC-1 cell lines which was significantly greater than AsPC-1 binding at concentrations exceeding 100 nM (p < 0.05). 64Cu-DOTA-C595 showed similar binding to the PANC-1 and AsPC-1 cells with no significant differences observed between cell lines (p > 0.05). The high cellular binding of 177Lu-DOTA-C595 to MUC1-CE positive cell lines suggests promise as a therapeutic radioimmunoconjugate against PDAC while further work is required to harness the potential of 64Cu-DOTA-C595 as a diagnostic radioimmunoconjugate.
Collapse
Affiliation(s)
- Ashleigh Hull
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
- Correspondence:
| | - Yanrui Li
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Dylan Bartholomeusz
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - William Hsieh
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
| | - William Tieu
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Tara L. Pukala
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Alexander H. Staudacher
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Eva Bezak
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
9
|
Preclinical Safety Evaluation of Intraperitoneally Administered Cu-Conjugated Anti-EGFR Antibody NCAB001 for the Early Diagnosis of Pancreatic Cancer Using PET. Pharmaceutics 2022; 14:pharmaceutics14091928. [PMID: 36145676 PMCID: PMC9504034 DOI: 10.3390/pharmaceutics14091928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Detecting tumor lesions <1 cm in size using current imaging methods remains a clinical challenge, especially in pancreatic cancer. Previously, we developed a method to identify pancreatic tumor lesions ≥3 mm using positron emission tomography (PET) with an intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor (EGFR) antibody (64Cu-NCAB001 ipPET). Here, we conducted an extended single-dose toxicity study of 64Cu-NCAB001 ipPET in mice based on approach 1 of the current ICH M3 [R2] guideline, as our new drug formulation contains 45 μg of the antibody. We used NCAB001 labeled with stable copper isotope instead of 64Cu. The total content of size variants was approximately 6.0% throughout the study. The relative binding potency of Cu-NCAB001 to recombinant human EGFR was comparable to that of cetuximab. The general and neurological toxicities of Cu-NCAB001 ipPET at 62.5 or 625 μg/kg were assessed in mice. The no-observed-adverse-effect level of Cu-NCAB001 was 625 μg/kg, a dose approximately 1000-fold higher at the μg/kg level than the dose of 64Cu-NCAB001 in our formulation (45 µg). The size variants did not affect the safety of the formulation. Therefore, clinical studies on the efficacy of 64Cu-NCAB001 ipPET for early detection of pancreatic cancer using PET imaging can be safely conducted.
Collapse
|
10
|
Wei GX, Du Y, Zhou YW, Li LJ, Qiu M. Peritoneal carcinomatosis with intraperitoneal immunotherapy: current treatment options and perspectives. Expert Rev Gastroenterol Hepatol 2022; 16:851-861. [PMID: 36107723 DOI: 10.1080/17474124.2022.2125866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Peritoneal carcinomatosis (PC) is an advanced malignancy that is not sensitive to systemic conventional chemotherapy. Treatment options for PC are usually palliative rather than curative. Cytoreductive surgery and hyperthermic intraperitoneal (IP) chemotherapy are associated with limited efficacy in patients with PC. However, the peritoneum can produce effective immunity by inducing T-lymphocyte recruitment and proliferation, and the unique immune environment of the peritoneum provides the rationale for IP immunotherapy in PC. AREAS COVERED The authors retrieved relevant documents of IP immunotherapy for PC from PubMed and Medline. This review elaborates on the knowledge of the peritoneal immune microenvironment and IP immunotherapy for PC covering immune stimulators, radioimmunotherapy, catumaxomab, cancer vaccines, chimeric antigen receptor (CAR)-T cells, and immune checkpoint inhibitors. EXPERT OPINION The prognosis of PC is poor. However, the peritoneal cavity is a unique immune compartment with abundant immune cells which can produce effective immunity. IP immunotherapy may be a promising strategy in patients with PC.
Collapse
Affiliation(s)
- Gui-Xia Wei
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yang Du
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Lin-Juan Li
- Thoracic Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Pu N, Yin H, Chen Q, Zhang J, Wu W, Lou W. Current status and future perspectives of clinical research in pancreatic cancer: Establishment of evidence by science. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2022; 29:741-757. [PMID: 34514722 DOI: 10.1002/jhbp.1045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 11/09/2022]
Abstract
Pancreatic cancer is one of the most aggressive diseases in the world due to a lack of early detection, leading to an overall 5-year survival of only 10%. In recent years, clinical trials targeted pancreatic cancer in efforts to improve survival. These studies introduce new technologies, concepts, and evidence which have instilled new optimism for improving prognosis. This review summarizes the current status of the recent (5-year) clinical trials and describes contemporary research on pancreatic cancer, including surgical technology, diagnostic skills, traditional chemoradiotherapy, neoadjuvant chemotherapy, immunotherapy, targeted therapy, and precision medicine. Then, the future trend and direction of clinical trials on pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanlin Yin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jicheng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Wheeler TT, Cao P, Ghouri MD, Ji T, Nie G, Zhao Y. Nanotechnological strategies for prostate cancer imaging and diagnosis. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
13
|
Hihara F, Matsumoto H, Yoshimoto M, Masuko T, Endo Y, Igarashi C, Tachibana T, Shinada M, Zhang MR, Kurosawa G, Sugyo A, Tsuji AB, Higashi T, Kurihara H, Ueno M, Yoshii Y. In Vitro Tumor Cell-Binding Assay to Select High-Binding Antibody and Predict Therapy Response for Personalized 64Cu-Intraperitoneal Radioimmunotherapy against Peritoneal Dissemination of Pancreatic Cancer: A Feasibility Study. Int J Mol Sci 2022; 23:5807. [PMID: 35628616 PMCID: PMC9146758 DOI: 10.3390/ijms23105807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
Peritoneal dissemination of pancreatic cancer has a poor prognosis. We have reported that intraperitoneal radioimmunotherapy using a 64Cu-labeled antibody (64Cu-ipRIT) is a promising adjuvant therapy option to prevent this complication. To achieve personalized 64Cu-ipRIT, we developed a new in vitro tumor cell-binding assay (64Cu-TuBA) system with a panel containing nine candidate 64Cu-labeled antibodies targeting seven antigens (EGFR, HER2, HER3, TfR, EpCAM, LAT1, and CD98), which are reportedly overexpressed in patients with pancreatic cancer. We investigated the feasibility of 64Cu-TuBA to select the highest-binding antibody for individual cancer cell lines and predict the treatment response in vivo for 64Cu-ipRIT. 64Cu-TuBA was performed using six human pancreatic cancer cell lines. For three cell lines, an in vivo treatment study was performed with 64Cu-ipRIT using high-, middle-, or low-binding antibodies in each peritoneal dissemination mouse model. The high-binding antibodies significantly prolonged survival in each mouse model, while low-and middle-binding antibodies were ineffective. There was a correlation between in vitro cell binding and in vivo therapeutic efficacy. Our findings suggest that 64Cu-TuBA can be used for patient selection to enable personalized 64Cu-ipRIT. Tumor cells isolated from surgically resected tumor tissues would be suitable for analysis with the 64Cu-TuBA system in future clinical studies.
Collapse
Affiliation(s)
- Fukiko Hihara
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Hiroki Matsumoto
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, National Cancer Center Hospital East, Chiba 277-8577, Japan;
| | - Takashi Masuko
- School of Pharmacy, Kindai University, Osaka 577-8502, Japan; (T.M.); (Y.E.)
| | - Yuichi Endo
- School of Pharmacy, Kindai University, Osaka 577-8502, Japan; (T.M.); (Y.E.)
| | - Chika Igarashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Tomoko Tachibana
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Mitsuhiro Shinada
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Faculty of Science, Toho University, Chiba 274-8510, Japan
| | - Ming-Rong Zhang
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Gene Kurosawa
- International Center for Cell and Gene Therapy, Fujita Health University, Aichi 470-1192, Japan;
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Atsushi B. Tsuji
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Tatsuya Higashi
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| | - Yukie Yoshii
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan;
(F.H.); (H.M.); (C.I.); (T.T.); (M.S.); (M.-R.Z.); (A.S.); (A.B.T.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Kanagawa 241-8515, Japan;
| |
Collapse
|
14
|
Characterization and Stabilization of a New 64Cu-Labeled Anti-EGFR Antibody NCAB001 for the Early Detection of Pancreatic Cancer with Positron Emission Tomography. Pharmaceutics 2021; 14:pharmaceutics14010067. [PMID: 35056963 PMCID: PMC8779674 DOI: 10.3390/pharmaceutics14010067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/24/2021] [Accepted: 12/25/2021] [Indexed: 12/20/2022] Open
Abstract
Early diagnosis of pancreatic cancer using current imaging modalities remains challenging. We have developed a new approach to identify tumor lesions ≥ 3 mm in the pancreas by positron emission tomography (PET) with a new intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor (EGFR) antibody (encoded as NCAB001), called 64Cu-NCAB001 ipPET. Generally, in clinical research, a radiometal-antibody complex must be prepared immediately before use at the imaging site. To make 64Cu-NCAB001 ipPET available to daily clinical practices in a sustainable way, the NCAB001-chelator conjugate and 64Cu-NCAB001 must be characterized and stabilized. NCAB001 was manufactured under cGMP conditions. NCAB001 was conjugated with a bifunctional chelator (p-SCN-Bn-PCTA), and the antibody-chelator conjugate (PCTA-NCAB001) was characterized by LC/MS and ELISA. Thereafter, to effectively manufacture 64Cu-NCAB001, we developed a new formulation to stabilize PCTA-NCAB001 and 64Cu-NCAB001. An average of three PCTA chelators were conjugated per molecule of NCAB001. The relative binding potency of PCTA-NCAB001 was comparable to cetuximab. The formulation consisting of acetate buffer, glycine, and polysorbate-80 stabilized PCTA-NCAB001 for a year-long storage. Additionally, this formulation enabled the stabilization of 64Cu-NCAB001 for up to 24 h after radiolabeling with a sufficient radioactivity concentration for clinical use. These results may accelerate the future use of 64Cu-NCAB001 ipPET in clinical settings for the early diagnosis and treatment of pancreatic cancer.
Collapse
|
15
|
Abstract
Pancreatic cancer is a kind of digestive tract malignant tumor with a poor prognosis. Radical surgery is the preferred alternative choice for patients with pancreatic cancer, but most patients have no chance of radical surgery when they are diagnosed. At present, a number of studies have been carried out on immunotherapies for pancreatic cancer, mainly including immune checkpoint inhibitors, tumor vaccines, and adoptive cell therapy, which are expected to become a new strategy for the treatment of pancreatic cancer, and ultimately achieve the purpose of improving the overall prognosis of patients with pancreatic cancer. In this paper, we summarize the current status of pancreatic cancer immunotherapy and analyze the future trend of immunotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Cheng-Yi Sun
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
16
|
Igarashi C, Yoshii Y, Tashima H, Iwao Y, Sakurai K, Hihara F, Tachibana T, Yoshida E, Wakizaka H, Akamatsu G, Yamaya T, Yoshimoto M, Matsumoto H, Zhang MR, Nagatsu K, Sugyo A, Tsuji AB, Higashi T. Usefulness of PET-guided surgery with 64Cu-labeled cetuximab for resection of intrapancreatic residual tumors in a xenograft mouse model of resectable pancreatic cancer. Nucl Med Commun 2021; 42:1112-1121. [PMID: 34100794 DOI: 10.1097/mnm.0000000000001442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In pancreatic cancer surgery, accurate identification and resection of intrapancreatic residual tumors are quite difficult. We have developed a novel open-typed PET system (called 'OpenPET'), which enables high-resolution PET-guided surgery in real time, and demonstrated that OpenPET-guided surgery with intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor antibody cetuximab is useful to detect and resect primary pancreatic cancer. Here, we investigated applicability of OpenPET-guided surgery for unexpected residual intrapancreatic tumors and examined its survival benefit over conventional surgery. METHODS A mouse model with large (>1 cm) resectable pancreatic cancer of xPA-1-DC cells expressing red fluorescent protein was used. OpenPET-guided surgery was conducted 24 h after intraperitoneal administration of 64Cu-labeled cetuximab (7.4 MBq/mouse). For comparison, similar surgical procedures were conducted, and conventional tumor resection was attempted using only the naked eye (control). Survival rate after OpenPET-guided surgery was compared to that after control operations. RESULTS Intraoperative OpenPET guidance enabled detection and resection of small residual tumors. Ten residual tumor specimens (3-10 mm in diameter) were intraoperatively isolated with OpenPET guidance (n = 7 mice). All isolated specimens showed tumor RFP signals. No resection of tumor tissue was performed in control group because the tumor could not be clearly detected with the naked eye alone. Mice after OpenPET-guided surgery showed significantly longer survival rates than those in control group. CONCLUSIONS OpenPET-guided surgery with 64Cu-labeled-cetuximab enabled intraoperative identification and resection of intrapancreatic small residual tumors. This technology could be useful to prevent tumor residuals during surgery and improve pancreatic cancer survival.
Collapse
Affiliation(s)
| | - Yukie Yoshii
- Department of Molecular Imaging and Theranostics
| | - Hideaki Tashima
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Yuma Iwao
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | | | | | | | - Eiji Yoshida
- Department of Molecular Imaging and Theranostics
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | | | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics
| | | | | |
Collapse
|
17
|
Evaluation of 64Cu-Labeled New Anti-EGFR Antibody NCAB001 with Intraperitoneal Injection for Early PET Diagnosis of Pancreatic Cancer in Orthotopic Tumor-Xenografted Mice and Nonhuman Primates. Pharmaceuticals (Basel) 2021; 14:ph14100950. [PMID: 34681174 PMCID: PMC8540406 DOI: 10.3390/ph14100950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023] Open
Abstract
Objectives: To improve the prognosis of pancreatic cancer, new imaging methods to identify tumor lesions at a size of <1 cm are urgently needed. To approach this clinical issue, we developed a new method to detect small tumor lesions in the pancreas (≥3 mm) by positron emission tomography (PET) using an intraperitoneally (ip)-administered 64Cu-labeled new anti-epidermal growth factor receptor (EGFR) antibody (encoded as NCAB001), called 64Cu-NCAB001 ipPET. Methods: NCAB001 was manufactured under cGMP conditions and labeled with 64Cu. The radiochemical and biological properties of 64Cu-NCAB001 were evaluated. Tumor uptake of an ip-administered 64Cu-NCAB001 in mice with orthotopic pancreatic tumor xPA1-DC xenografts was also evaluated. Pharmacokinetics and radiation dosimetry were examined using PET images acquired after the ip administration of 64Cu-NCAB001 into cynomolgus monkeys with pharmacologic safety monitoring. Results: Radio-chromatography, cell-binding assays, and biodistribution of 64Cu-NCAB001 in mice were identical to those of our previous data with clinically available cetuximab. Small tumor lesions in the pancreas (≥3 mm) of mice could be identified by 64Cu-NCAB001 ipPET. The ip administration of 64Cu-NCAB001 into monkeys was safely conducted using ultrasound imaging. PET images in monkeys showed that ip-administered 64Cu-NCAB001 was distributed throughout the intraperitoneal cavity for up to 6 h and cleared thereafter. Most of the radioactivity was distributed in the liver and the large intestine. The radioactivity around the pancreas became negligible 24 h after administration. The estimated human effective dose was 0.0174 mSv/MBq. Conclusion: Our data support the initiation of clinical trials of 64Cu-NCAB001 ipPET to transfer this promising tool for the early diagnosis of pancreatic cancers.
Collapse
|
18
|
Chomet M, van Dongen GAMS, Vugts DJ. State of the Art in Radiolabeling of Antibodies with Common and Uncommon Radiometals for Preclinical and Clinical Immuno-PET. Bioconjug Chem 2021; 32:1315-1330. [PMID: 33974403 PMCID: PMC8299458 DOI: 10.1021/acs.bioconjchem.1c00136] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Inert
and stable radiolabeling of monoclonal antibodies (mAb),
antibody fragments, or antibody mimetics with radiometals is a prerequisite
for immuno-PET. While radiolabeling is preferably fast, mild, efficient,
and reproducible, especially when applied for human use in a current
Good Manufacturing Practice compliant way, it is crucial that the
obtained radioimmunoconjugate is stable and shows preserved immunoreactivity
and in vivo behavior. Radiometals and chelators have
extensively been evaluated to come to the most ideal radiometal–chelator
pair for each type of antibody derivative. Although PET imaging of
antibodies is a relatively recent tool, applications with 89Zr, 64Cu, and 68Ga have greatly increased in
recent years, especially in the clinical setting, while other less
common radionuclides such as 52Mn, 86Y, 66Ga, and 44Sc, but also 18F as in [18F]AlF are emerging promising candidates for the radiolabeling
of antibodies. This review presents a state of the art overview of
the practical aspects of radiolabeling of antibodies, ranging from
fast kinetic affibodies and nanobodies to slow kinetic intact mAbs.
Herein, we focus on the most common approach which consists of first
modification of the antibody with a chelator, and after eventual storage
of the premodified molecule, radiolabeling as a second step. Other
approaches are possible but have been excluded from this review. The
review includes recent and representative examples from the literature
highlighting which radiometal–chelator–antibody combinations
are the most successful for in vivo application.
Collapse
Affiliation(s)
- Marion Chomet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Guus A M S van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| | - Danielle J Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Radiology & Nuclear Medicine, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
19
|
Mallya K, Gautam SK, Aithal A, Batra SK, Jain M. Modeling pancreatic cancer in mice for experimental therapeutics. Biochim Biophys Acta Rev Cancer 2021; 1876:188554. [PMID: 33945847 DOI: 10.1016/j.bbcan.2021.188554] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy.
Collapse
Affiliation(s)
- Kavita Mallya
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
20
|
Intraperitoneal Glucose Transport to Micrometastasis: A Multimodal In Vivo Imaging Investigation in a Mouse Lymphoma Model. Int J Mol Sci 2021; 22:ijms22094431. [PMID: 33922728 PMCID: PMC8123046 DOI: 10.3390/ijms22094431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Bc-DLFL.1 is a novel spontaneous, high-grade transplantable mouse B-cell lymphoma model for selective serosal propagation. These cells attach to the omentum and mesentery and show dissemination in mesenteric lymph nodes. We aimed to investigate its early stage spread at one day post-intraperitoneal inoculation of lymphoma cells (n = 18 mice), and its advanced stage at seven days post-inoculation with in vivo [18F]FDG-PET and [18F]PET/MRI, and ex vivo by autoradiography and Cherenkov luminescence imaging (CLI). Of the early stage group, nine animals received intraperitoneal injections, and nine received intravenous [18F]FDG injections. The advanced stage group (n = 3) received intravenous FDG injections. In the early stage, using autoradiography we observed a marked accumulation in the mesentery after intraperitoneal FDG injection. Using other imaging methods and autoradiography, following the intravenous injection of FDG no accumulations were detected. At the advanced stage, tracer accumulation was clearly detected in mesenteric lymph nodes and in the peritoneum after intravenous administration using PET. We confirmed the results with immunohistochemistry. Our results in this model highlight the importance of local FDG administration during diagnostic imaging to precisely assess early peritoneal manifestations of other malignancies (colon, stomach, ovary). These findings also support the importance of applying topical therapies, in addition to systemic treatments in peritoneal cancer spread.
Collapse
|
21
|
White JM, Escorcia FE, Viola NT. Perspectives on metals-based radioimmunotherapy (RIT): moving forward. Theranostics 2021; 11:6293-6314. [PMID: 33995659 PMCID: PMC8120204 DOI: 10.7150/thno.57177] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Radioimmunotherapy (RIT) is FDA-approved for the clinical management of liquid malignancies, however, its use for solid malignancies remains a challenge. The putative benefit of RIT lies in selective targeting of antigens expressed on the tumor surface using monoclonal antibodies, to systemically deliver cytotoxic radionuclides. The past several decades yielded dramatic improvements in the quality, quantity, recent commercial availability of alpha-, beta- and Auger Electron-emitting therapeutic radiometals. Investigators have created new or improved existing bifunctional chelators. These bifunctional chelators bind radiometals and can be coupled to antigen-specific antibodies. In this review, we discuss approaches to develop radiometal-based RITs, including the selection of radiometals, chelators and antibody platforms (i.e. full-length, F(ab')2, Fab, minibodies, diabodies, scFv-Fc and nanobodies). We cite examples of the performance of RIT in the clinic, describe challenges to its implementation, and offer insights to address gaps toward translation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/metabolism
- Antineoplastic Agents, Immunological/therapeutic use
- Chelating Agents/administration & dosage
- Chelating Agents/metabolism
- Click Chemistry
- Clinical Trials as Topic
- Dose Fractionation, Radiation
- Drug Delivery Systems
- Forecasting
- Humans
- Immunoglobulin Fab Fragments/administration & dosage
- Immunoglobulin Fab Fragments/therapeutic use
- Lymphoma, Non-Hodgkin/radiotherapy
- Mice
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasms, Experimental/diagnostic imaging
- Neoplasms, Experimental/radiotherapy
- Organ Specificity
- Precision Medicine
- Radiation Tolerance
- Radioimmunotherapy/methods
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/therapeutic use
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Single-Chain Antibodies/administration & dosage
- Single-Chain Antibodies/therapeutic use
- Single-Domain Antibodies/administration & dosage
- Single-Domain Antibodies/therapeutic use
- Yttrium Radioisotopes/administration & dosage
- Yttrium Radioisotopes/therapeutic use
Collapse
Affiliation(s)
- Jordan M. White
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20814
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48201
| |
Collapse
|
22
|
Nguyen TT, Ho AS, Nguyen TKG, Nguyen TN, Bui VC, Nguyen TB, Dang HHQ, Nguyen DK, Nguyen TN, Nguyen LT. Efficacy of nimotuzumab (hR3) conjugated with 131I or 90Y in laryngeal carcinoma xenograft mouse model. Int J Radiat Biol 2021; 97:704-713. [PMID: 33617414 DOI: 10.1080/09553002.2021.1889703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 01/06/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE The humanized monoclonal antibody hR3, both alone and in combination with other chemotherapeutic agents and radiotherapy, can be used to treat head and neck cancers. Substantial progress has been made in the development of targeted radioimmunotherapy using iodine-131 (131I) and yttrium-90 (90Y) radioisotopes in recent years. In the present study, we examined the efficacy of hR3 conjugated with 131I or 90Y to inhibit tumor growth in a laryngeal carcinoma xenograft tumor model. METHODS hR3 was labeled with 131I or 90Y to generate the conjugates 131I-hR3 or 90Y-hR3. The conjugates were incubated with HEp-2 laryngeal carcinoma cells to evaluate binding capacity. The efficacy of the labeled hR3 conjugates to treat laryngeal cancer was also evaluated in nude mice inoculated with HEp-2 tumors. RESULTS The purified radioimmunoconjugates with specific activities of 187-191 MBq/mg had radiochemical purity >98% and >80% immunoreactivity with HEp-2 cells. Mice with HEp-2 xenografts treated with 131I-hR3 or 90Y-hR3 showed reduced tumor volume and improved survival rates compared to the untreated control group and the group treated with unlabeled hR3. At equivalent doses, radioimmunotherapeutic hR3 labeled with 90Y had increased tumor inhibition activity compared to hR3 labeled with 131I. CONCLUSIONS 131I-hR3 and 90Y-hR3 are promising targeted radiopharmaceuticals for treatment of head and neck cancers, especially laryngeal cancers.
Collapse
Affiliation(s)
| | - Anh-Son Ho
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | | | | | | | | | | | | | | | - Linh-Toan Nguyen
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| |
Collapse
|
23
|
Martin S, Maus S, Stemler T, Rosar F, Khreish F, Holland JP, Ezziddin S, Bartholomä MD. Proof-of-Concept Study of the NOTI Chelating Platform: Preclinical Evaluation of 64Cu-Labeled Mono- and Trimeric c(RGDfK) Conjugates. Mol Imaging Biol 2021; 23:95-108. [PMID: 32856224 PMCID: PMC7782405 DOI: 10.1007/s11307-020-01530-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/14/2020] [Accepted: 08/09/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE We recently developed a chelating platform based on the macrocycle 1,4,7-triazacyclononane with up to three five-membered azaheterocyclic arms for the preparation of 68Ga- and 64Cu-based radiopharmaceuticals. Based on this platform, the chelator scaffold NOTI-TVA with three additional carboxylic acid groups for bioconjugation was synthesized and characterized. The primary aims of this proof-of-concept study were (1) to evaluate if trimeric radiotracers on the basis of the NOTI-TVA 6 scaffold can be developed, (2) to determine if the additional substituents for bioconjugation at the non-coordinating NH atoms of the imidazole residues of the building block NOTI influence the metal binding properties, and (3) what influence multiple targeting vectors have on the biological performance of the radiotracer. The cyclic RGDfK peptide that specifically binds to the αvß3 integrin receptor was selected as the biological model system. PROCEDURES Two different synthetic routes for the preparation of NOTI-TVA 6 were explored. Three c(RGDfK) peptide residues were conjugated to the NOTI-TVA 6 building block by standard peptide chemistry providing the trimeric bioconjugate NOTI-TVA-c(RGDfK)3 9. Labeling of 9 with [64Cu]CuCl2 was performed manually at pH 8.2 at ambient temperature. Binding affinities of Cu-8, the Cu2+ complex of the previously described monomer NODIA-Me-c(RGDfK) 8, and the trimer Cu-9 to integrin αvß3 were determined in competitive cell binding experiments in the U-87MG cell line. The pharmacokinetics of both 64Cu-labeled conjugates [64Cu]Cu-8 and [64Cu]Cu-9 were determined by small-animal PET imaging and ex vivo biodistribution studies in mice bearing U-87MG xenografts. RESULTS Depending on the synthetic route, NOTI-TVA 6 was obtained with an overall yield up to 58 %. The bioconjugate 9 was prepared in 41 % yield. Both conjugates [64Cu]Cu-8 and [64Cu]Cu-9 were radiolabeled quantitatively at ambient temperature in high molar activities of Am ~ 20 MBq nmol-1 in less than 5 min. Competitive inhibitory constants IC50 of c(RDGfK) 7, Cu-8, and Cu-9 were determined to be 159.5 ± 1.3 nM, 256.1 ± 2.1 nM, and 99.5 ± 1.1 nM, respectively. In small-animal experiments, both radiotracers specifically delineated αvß3 integrin-positive U-87MG tumors with low uptake in non-target organs and rapid blood clearance. The trimer [64Cu]Cu-9 showed a ~ 2.5-fold higher tumor uptake compared with the monomer [64Cu]Cu-8. CONCLUSIONS Functionalization of NOTI at the non-coordinating NH atoms of the imidazole residues for bioconjugation was straightforward and allowed the preparation of a homotrimeric RGD conjugate. After optimization of the synthesis, required building blocks to make NOTI-TVA 6 are now available on multi-gram scale. Modifications at the imidazole groups had no measurable impact on metal binding properties in vitro and in vivo suggesting that the NOTI scaffold is a promising candidate for the development of 64Cu-labeled multimeric/multifunctional radiotracers.
Collapse
Affiliation(s)
- Sebastian Martin
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue de Bugnon 25A, CH-1011, Lausanne, Switzerland
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Tobias Stemler
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Florian Rosar
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Fadi Khreish
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Jason P Holland
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Mark D Bartholomä
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany.
- Department of Nuclear Medicine, University of Freiburg - Medical Center, Hugstetterstrasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
24
|
Zhou H, Zhang Q, Cheng Y, Xiang L, Shen G, Wu X, Cai H, Li D, Zhu H, Zhang R, Li L, Cheng Z. 64Cu-labeled melanin nanoparticles for PET/CT and radionuclide therapy of tumor. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102248. [PMID: 32574686 DOI: 10.1016/j.nano.2020.102248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Melanin is a group of natural pigments found in living organism. It can be used for positron emission tomography (PET) imaging due to its inherent chelating ability to radioactive cupric ion. This study was to prepare 64Cu-labeled PEGylated melanin nanoparticles (64Cu-PEG-MNPs), and to further take advantage of the enhanced permeability and retention (EPR) effect of radiolabeled nanoparticles to realize the integration of tumor diagnosis and treatment. We successfully synthesized PEG-MNPs. Saline and serum stability experiments demonstrated good stability. PET/CT showed high tumor aggregation. Moreover, 64Cu-PEG-MNPs resulted in a therapeutic effect on the A431 tumor-bearing mice in the treatment group. The pathological results further confirmed that the therapeutic doses of 64Cu-PEG-MNPs cause pathological changes of tumor tissues while showing minimal toxicity to normal tissues. Our data successfully demonstrate the good imaging performance of 64Cu-PEG-MNPs on A431 tumors and further proved its therapeutic effect, highlighting a great potential in targeted radionuclide therapy.
Collapse
Affiliation(s)
- Huijun Zhou
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Qing Zhang
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA; Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yan Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lili Xiang
- Department of Gastrointestinal Surgery, West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guohua Shen
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Daifeng Li
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA; Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hua Zhu
- Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ruiping Zhang
- The Affiliated Shanxi Bethune Hospital of Shanxi Medical University; The Affiliated Cancer Hospital of Shanxi Medical University, Taiyuan, China.
| | - Lin Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Immuno-OpenPET: a novel approach for early diagnosis and image-guided surgery for small resectable pancreatic cancer. Sci Rep 2020; 10:4143. [PMID: 32157106 PMCID: PMC7064510 DOI: 10.1038/s41598-020-61056-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) has a poor prognosis owing to difficulties in the diagnosis of resectable PC at early stages. Several clinical studies have indicated that the detection and surgery of small resectable PC (<1 cm) can significantly improve survival; however, imaging diagnosis and accurate resection of small PC remain challenging. Here, we report the feasibility of "immuno-OpenPET" as a novel approach enabling not only early diagnosis but also image-guided surgery, using a small (<1 cm) resectable PC orthotopic xenograft mouse model. For immuno-OpenPET, we utilized our original OpenPET system, which enables high-resolution positron emission tomography (PET) imaging with depth-of-interaction detectors, as well as real-time image-guided surgery, by arranging the detectors to create an open space for surgery and accelerating the image reconstruction process by graphics processing units. For immuno-OpenPET, 64Cu-labeled anti-epidermal growth factor receptor antibody cetuximab was intraperitoneally administered into mice. It clearly identified PC tumors ≥3 mm. In contrast, neither OpenPET with intravenous-administered 64Cu-cetuximab nor intraperitoneal/intravenous-administered 18F-FDG (a traditional PET probe) could detect PC in this model. Immuno-OpenPET-guided surgery accurately resected small PC in mice and achieved significantly prolonged survival. This technology could provide a novel diagnostic and therapeutic strategy for small resectable PC to improve patient survival.
Collapse
|
26
|
Hull A, Li Y, Bartholomeusz D, Hsieh W, Allen B, Bezak E. Radioimmunotherapy of Pancreatic Ductal Adenocarcinoma: A Review of the Current Status of Literature. Cancers (Basel) 2020; 12:E481. [PMID: 32092952 PMCID: PMC7072553 DOI: 10.3390/cancers12020481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/11/2020] [Accepted: 02/15/2020] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has long been associated with low survival rates. A lack of accurate diagnostic tests and limited treatment options contribute to the poor prognosis of PDAC. Radioimmunotherapy using α- or β-emitting radionuclides has been identified as a potential treatment for PDAC. By harnessing the cytotoxicity of α or β particles, radioimmunotherapy may overcome the anatomic and physiological factors which traditionally make PDAC resistant to most conventional treatments. Appropriate selection of target receptors and the development of selective and cytotoxic radioimmunoconjugates are needed to achieve the desired results of radioimmunotherapy. The aim of this review is to examine the growing preclinical and clinical trial evidence regarding the application of α and β radioimmunotherapy for the treatment of PDAC. A systematic search of MEDLINE® and Scopus databases was performed to identify 34 relevant studies conducted on α or β radioimmunotherapy of PDAC. Preclinical results demonstrated α and β radioimmunotherapy provided effective tumour control. Clinical studies were limited to investigating β radioimmunotherapy only. Phase I and II trials observed disease control rates of 11.2%-57.9%, with synergistic effects noted for combination therapies. Further developments and optimisation of treatment regimens are needed to improve the clinical relevance of α and β radioimmunotherapy in PDAC.
Collapse
Affiliation(s)
- Ashleigh Hull
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (Y.L.); (W.H.); (E.B.)
| | - Yanrui Li
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (Y.L.); (W.H.); (E.B.)
| | - Dylan Bartholomeusz
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia;
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - William Hsieh
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (Y.L.); (W.H.); (E.B.)
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia;
| | - Barry Allen
- Faculty of Medicine, Western Sydney University, Liverpool, NSW 2170, Australia;
| | - Eva Bezak
- Cancer Research Institute and School of Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (Y.L.); (W.H.); (E.B.)
- Department of Physics, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
27
|
Boyle AJ, Cao PJ, Cai Z, Chan C, Hedley DW, Reilly RM. Radioimmunotherapy of human pancreatic cancer xenografts in NOD-scid mice with [ 64Cu]Cu-NOTA-panitumumab F(ab') 2 alone or combined with radiosensitizing gemcitabine and the PARP inhibitor, rucaparib. Nucl Med Biol 2020; 84-85:46-54. [PMID: 32062317 DOI: 10.1016/j.nucmedbio.2020.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/20/2020] [Accepted: 02/02/2020] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Our objective was to determine the feasibility of extending our previously reported PET imaging study of pancreatic cancer (PnCa) with [64Cu]Cu-NOTA-panitumumab F(ab')2 to radioimmunotherapy (RIT) by exploiting the β-particle and Auger electron emissions of 64Cu (PET theranostic concept). To enhance the effectiveness of [64Cu]Cu-NOTA-panitumumab F(ab')2, we further combined RIT with radiosensitizing gemcitabine (GEM) and the poly(ADP)ribose polymerase inhibitor (PARPi), rucaparib. METHODS Normal tissue toxicity was assessed in non-tumor-bearing NOD-scid mice injected i.v. with [64Cu]Cu-NOTA-panitumumab F(ab')2 (1.85-9.25 MBq; 10 μg) or [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 (12.95 MBq). Body weight was monitored, and hematopoietic (CBC), liver (ALT) and kidney [creatinine (SCr)] toxicity were assessed. RIT studies were performed in NOD-scid mice with s.c. OCIP23 human PnCa patient-derived xenografts (PDX) administered [64Cu]Cu-NOTA-panitumumab F(ab')2 (3.7 MBq; 10 μg), unlabeled panitumumab F(ab')2 (10 μg) or normal saline every two weeks. Subsequent studies evaluated RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) administered alone or combined with GEM and the PARPi, rucaparib administered on a 14-day treatment cycle for up to 6 cycles in NOD-scid mice with s.c. PANC-1 human PnCa xenografts. The radiation absorbed dose in PANC-1 tumors and normal organs in mice after a single i.v. injection of [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) was estimated based on previously reported biodistribution studies of [64Cu]Cu-NOTA-panitumumab F(ab')2. RESULTS No normal tissue toxicity was observed in non-tumor-bearing NOD-scid mice administered up to 3.7 MBq (10 μg) of [64Cu]Cu-NOTA-panitumumab F(ab')2 but slightly increased ALT was noted at 9.25 MBq. Administration of [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 (12.95 MBq; 10 μg) caused some hematopoietic toxicity but no increase in ALT or SCr or decreased body weight. A slight tumor growth delay and increased survival was noted in NOD-scid mice with s.c. OCIP23 PDX treated with [64Cu]Cu-NOTA-panitumumab F(ab')2 (3.7 MBq; 10 μg) or unlabeled panitumumab F(ab')2 (10 μg) compared to normal saline treated mice. RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.95 MBq; 10 μg) combined with GEM + PARPi for up to 6 cycles was most effective for the treatment of PANC-1 tumors. Tumor doubling time increased to 13.3 ± 0.9 days vs. 7.8 ± 3.7 days for RIT alone and 9.3 ± 2.2 days for normal saline treatment. Median survival was significantly longer (P < 0.05) than in mice treated with normal saline (35 days) for RIT + GEM + PARPi (71 days), GEM + PARPi (44 days) and RIT + GEM (43 days) but not for RIT alone (25 days). RIT + GEM + PARPi provided a longer median survival than RIT (P < 0.01), GEM + PARPi (P = 0.01) but not RIT + GEM (P = 0.23). Nonetheless, PANC-1 tumors grew exponentially in all treatment groups. The absorbed dose in PANC-1 tumors after a single i.v. injection of [64Cu]Cu-NOTA-panitumumab F(ab')2 (12.85 MBq; 10 μg) was 0.8 Gy, while the dose in normal organs ranged from 0.6-1.2 Gy. CONCLUSIONS We conclude that RIT with [64Cu]Cu-NOTA-panitumumab F(ab')2 did not cause significant normal tissue toxicity but was not effective when administered alone for treatment of PnCa xenografts in NOD-scid mice. Combining RIT with GEM and the PARPi, rucaparib enhanced its effectiveness but tumors continued to grow exponentially. Our results suggest that 64Cu is not feasible for RIT of PnCa due to low tumor absorbed doses. 177Lu which has a higher abundance of moderate energy β-particle emissions may be more effective than 64Cu. The hematopoietic toxicity of [64Cu]Cu-NOTA-anti-mouse EGFR Ab30 F(ab')2 may be mediated by binding to mouse EGFR expressed on some hematopoietic stem cells. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Direct extension of PET with 64Cu(Cu)-NOTA-panitumumab F(ab')2 to RIT exploiting the β-particle and Auger electron emissions of 64Cu is not feasible. Theranostic approaches that combine PET with RIT employing 177Lu may be more promising and should be explored.
Collapse
Affiliation(s)
- Amanda J Boyle
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Ping-Jiang Cao
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zhongli Cai
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - Conrad Chan
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| | - David W Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Raymond M Reilly
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada; Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, Joint Department of Medical Imaging, University Health Network, Toronto, ON, Canada.
| |
Collapse
|