1
|
Ondrák Fialová K, Ondrák L, Vlk M, Kozempel J, Nováková K, Nový Z, Hajduová K, Hajdúch M, Petřík M, Pruszynski M, Bruchertseifer F, Morgenstern A. In vitro and in vivo evaluation of anti-HER2 antibody conjugates labelled with 225Ac. EJNMMI Radiopharm Chem 2025; 10:16. [PMID: 40183827 PMCID: PMC11971111 DOI: 10.1186/s41181-025-00337-8] [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: 01/06/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Overexpression of human epidermal growth factor receptor type 2 (HER2) occurs in multiple carcinomas. For example, up to 20% of breast cancer cases are classified as HER2 positive (HER2+). Treatment of this condition typically involves immunotherapy using monoclonal antibodies, such as trastuzumab or pertuzumab. The precise targeting of monoclonal antibodies to HER2+ tumour lesions can be used as well in radioimmunotherapy to deliver medical radionuclides exactly to the afflicted area and therefore minimize radiation exposure of healthy tissues. In this study, DOTA conjugates of monoclonal antibodies trastuzumab and pertuzumab were prepared and tested in vitro. One of these, 225Ac-DOTA-pertuzumab, was also the subject of an ex vivo biodistribution study with normal as well as HER2+ and HER2- tumour-xenografted mice. This radioconjugate has not been previously described. RESULTS Three DOTA-conjugates of HER2 targeting monoclonal antibodies, one of trastuzumab and two of pertuzumab, were prepared and radiolabelled with 225Ac in different molar ratios. This procedure led to an optimisation of the preparation and radiolabelling process. The radioconjugates were shown to be highly stable in vitro in both fetal bovine serum and phosphate buffered saline under room temperature and decreased temperature for 10 days. In vitro cell studies with HER2-overexpressing cell-line (SKOV-3) and low HER2-expressing cell line (MDA-MB-231) proved that radioconjugates of both antibodies have high binding specificity and affinity towards HER2 receptors. These findings were confirmed for a novel radioconjugate 225Ac-DOTA-pertuzumab in an ex vivo biodistribution study, where uptake in HER2+ tumour was 50 ± 14% ID/g and HER2- tumour showed uptake comparable with healthy tissues (max. 5.0 ± 1.7% ID/g). The high uptake observed in the spleen can be attributed to the elimination of the antibody, as well as the use of an immunedeficient mouse strain (SCID). CONCLUSIONS During this study, the optimization of preparation and radiolabelling of HER2 targeting antibodies with 225Ac was accomplished. Furthermore, the radioconjugate 225Ac-DOTA-pertuzumab was prepared and evaluated for the first time. The radioconjugates of both tested antibodies demonstrated excellent qualities in terms of stability and HER2 receptor affinity. Initial ex vivo studies indicated that especially the radioconjugate 225Ac-DOTA-pertuzumab is a very promising candidate for further more detailed in vivo studies.
Collapse
Affiliation(s)
- Kateřina Ondrák Fialová
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 87/7, 115 19, Prague, Czech Republic.
| | - Lukáš Ondrák
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 87/7, 115 19, Prague, Czech Republic
| | - Martin Vlk
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 87/7, 115 19, Prague, Czech Republic
| | - Ján Kozempel
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 87/7, 115 19, Prague, Czech Republic
| | - Kateřina Nováková
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo naměstí 542/2, 16000, Prague, Czech Republic
| | - Zbyněk Nový
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00, Olomouc, Czech Republic
| | - Katarína Hajduová
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00, Olomouc, Czech Republic
| | - Miloš Petřík
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, 779 00, Olomouc, Czech Republic
| | - Marek Pruszynski
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195, Warsaw, Poland
- NOMATEN Centre of Excellence, National Centre for Nuclear Research, Andrzeja Soltana 7, 05-400, Otwock, Poland
| | | | | |
Collapse
|
2
|
Houvast RD, van Duijvenvoorde M, Thijse K, de Steur WO, de Geus-Oei LF, Crobach ASLP, Burggraaf J, Vahrmeijer AL, Kuppen PJK. Selecting Targets for Molecular Imaging of Gastric Cancer: An Immunohistochemical Evaluation. Mol Diagn Ther 2025; 29:213-227. [PMID: 39541080 PMCID: PMC11860997 DOI: 10.1007/s40291-024-00755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Tumor-targeted positron emission tomography (PET) and fluorescence-guided surgery (FGS) could address current challenges in pre- and intraoperative imaging of gastric cancer. Adequate selection of molecular imaging targets remains crucial for successful tumor visualization. This study evaluated the potential of integrin αvβ6, carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), epidermal growth factor receptor (EGFR), epithelial cell adhesion molecule (EpCAM) and human epidermal growth factor receptor-2 (HER2) for molecular imaging of primary gastric cancer, as well as lymph node and distant metastases. METHODS Expression of αvβ6, CEACAM5, EGFR, EpCAM and HER2 was determined using immunohistochemistry in human tissue specimens of primary gastric adenocarcinoma, healthy surrounding stomach, esophageal and duodenal tissue, tumor-positive and tumor-negative lymph nodes, and distant metastases, followed by quantification using the total immunostaining score (TIS). RESULTS Positive biomarker expression in primary gastric tumors was observed in 86% for αvβ6, 72% for CEACAM5, 77% for EGFR, 93% for EpCAM and 71% for HER2. Tumor expression of CEACAM5, EGFR and EpCAM was higher compared to healthy stomach tissue expression, while this was not the case for αvβ6 and HER2. Tumor-positive lymph nodes could be distinguished from tumor-negative lymph nodes, with accuracy ranging from 82 to 93% between biomarkers. CEACAM5, EGFR and EpCAM were abundantly expressed on distant metastases, with expression in 88-95% of tissue specimens. CONCLUSION Our findings show that CEACAM5, EGFR and EpCAM are promising targets for molecular imaging of primary gastric cancer, as well as visualization of both lymph node and distant metastases. Further clinical evaluation of PET and FGS tracers targeting these antigens is warranted.
Collapse
Affiliation(s)
- Ruben D Houvast
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | | | - Kira Thijse
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Wobbe O de Steur
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiation Science & Technology, Delft University of Technology, Delft, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - A Stijn L P Crobach
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus Burggraaf
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
3
|
Ding J, Qin S, Hou X, Zhang J, Yang M, Ma S, Zhu H, Feng Y, Yu F. Recent advances in emerging radiopharmaceuticals and the challenges in radiochemistry and analytical chemistry. Trends Analyt Chem 2025; 182:118053. [DOI: 10.1016/j.trac.2024.118053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Simó C, Shmuel S, Vanover A, Pereira PMR. [ 64Cu]Cu-NOTA-Trastuzumab and [ 89Zr]Zr-DFO-Trastuzumab in Xenografts with Varied HER2 Expression. Mol Pharm 2024; 21:6311-6322. [PMID: 39471823 PMCID: PMC11611601 DOI: 10.1021/acs.molpharmaceut.4c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
Positron emission tomography (PET) has potential as a complementary technique to biomarker analysis, especially for human epidermal growth factor receptor 2 (HER2)-expressing tumors characterized by high heterogeneity. In this study, zirconium-89 (89Zr) and copper-64 (64Cu) labeled trastuzumab were employed to monitor varying levels of tumoral HER2 expression. Additionally, we studied the use of the cholesterol-depleting lovastatin as a pharmacological approach to enhance cell-surface HER2 expression in tumors with moderate to low HER2 levels, aiming to increase antibody accumulation in these tumor types. Both 89Zr- and 64Cu-labeled trastuzumab effectively monitor HER2 expression levels in xenografts exhibiting varying HER2 expression. No significant difference in tumor uptake was observed between 89Zr- or 64Cu-labeled trastuzumab, and tumor uptake for both radioimmunoconjugates positively correlated with HER2 protein levels. These findings underscore the potential of PET to monitor HER2 protein levels across heterogeneous tumors. Furthermore, our results suggest that further optimization of statin dosing and timing could offer a promising strategy to enhance trastuzumab accumulation in HER2-high, HER2-moderate, and HER2-low tumors.
Collapse
Affiliation(s)
- Cristina Simó
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shayla Shmuel
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Alex Vanover
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| | - Patrícia M. R. Pereira
- Department of Radiology,
Mallinckrodt Institute of Radiology, Washington
University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
5
|
Li N, Chen S, Cai X. Harnessing molecular probes for imaging of human epidermal growth factor receptor (HER) family. Bioorg Med Chem 2024; 113:117931. [PMID: 39362074 DOI: 10.1016/j.bmc.2024.117931] [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: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
The human epidermal growth factor receptor (HER) family plays a critical role in the development, migration, and invasion of various cancers. Currently, the FDA has approved numerous targeting therapies for the HER family consist of small molecule drugs, monoclonal antibodies and antibody-drug conjugates. To facilitate precision therapy using currently approved targeted agents, early detection and quantification of each HER receptor are essential for assessment, treatment, and prognostic purposes. This study provides a comprehensive review of the latest advancements in detection and quantification of HER receptors, including traditional biopsies, liquid biopsies, and non-invasive detection methods. Although traditional histological methods, such as immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), have yielded valuable insights, advancements in real-time and non-invasive detection technologies necessitate improved methods for the dynamic evaluation of HER status. This article also reviews several emerging real-time techniques for detecting and quantifying HER status in circulating tumor cells (CTCs) extracted from blood samples, as well as in vivo assessments using positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging. This review emphasizes the importance of continuous innovation in the application of HER receptor imaging technologies, with the goal of enhancing treatment outcomes and prognoses for cancer patients.
Collapse
Affiliation(s)
- Na Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Shengxi Chen
- Biodesign Center for BioEnergetics, Arizona State University, Tempe 85287, USA.
| | - Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China.
| |
Collapse
|
6
|
Cox KE, Turner MA, Lwin TM, Amirfakhri S, Kelly KJ, Hosseini M, Ghosh P, Obonyo M, Hoffman RM, Yazaki PJ, Bouvet M. Targeting Patient-Derived Orthotopic Gastric Cancers with a Fluorescent Humanized Anti-CEA Antibody. Ann Surg Oncol 2024; 31:6291-6299. [PMID: 38888861 PMCID: PMC11300635 DOI: 10.1245/s10434-024-15570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Gastric cancer poses a major diagnostic and therapeutic challenge as surgical resection provides the only opportunity for a cure. Specific labeling of gastric cancer could distinguish resectable and nonresectable disease and facilitate an R0 resection, which could improve survival. METHODS Two patient-derived gastric cancer lines, KG8 and KG10, were established from surgical specimens of two patients who underwent gastrectomy for gastric adenocarcinoma. Harvested tumor fragments were implanted into the greater curvature of the stomach to establish patient-derived orthotopic xenograft (PDOX) models. M5A (humanized anti-CEA antibody) or IgG control antibodies were conjugated with the near-infrared dye IRDye800CW. Mice received 50 µg of M5A-IR800 or 50 µg of IgG-IR800 intravenously and were imaged after 72 hr. Fluorescence imaging was performed by using the LI-COR Pearl Imaging System. A tumor-to-background ratio (TBR) was calculated by dividing the mean fluorescence intensity of the tumor versus adjacent stomach tissue. RESULTS M5A-IR800 administration resulted in bright labeling of both KG8 and K10 tumors. In the KG8 PDOX models, the TBR for M5A-IR800 was 5.85 (SE ± 1.64) compared with IgG-IR800 at 0.70 (SE ± 0.17). The K10 PDOX models had a TBR of 3.71 (SE ± 0.73) for M5A-IR800 compared with 0.66 (SE ± 0.12) for IgG-IR800. CONCLUSIONS Humanized anti-CEA (M5A) antibodies conjugated to fluorescent dyes provide bright and specific labeling of gastric cancer PDOX models. This tumor-specific fluorescent antibody is a promising potential clinical tool to detect the extent of disease for the determination of resectability as well as to visualize tumor margins during gastric cancer resection.
Collapse
Affiliation(s)
- Kristin E Cox
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Michael A Turner
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Thinzar M Lwin
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Siamak Amirfakhri
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| | - Kaitlyn J Kelly
- Department of Surgical Oncology, University of Wisconsin, Madison, WI, USA
| | - Mojgan Hosseini
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert M Hoffman
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- AntiCancer Inc, San Diego, CA, USA
| | - Paul J Yazaki
- Department of Immunology & Theranostics, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, CA, USA.
- VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
7
|
Badier L, Quelven I. Zirconium 89 and Copper 64 for ImmunoPET: From Antibody Bioconjugation and Radiolabeling to Molecular Imaging. Pharmaceutics 2024; 16:882. [PMID: 39065579 PMCID: PMC11279968 DOI: 10.3390/pharmaceutics16070882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Immunotherapy has transformed cancer treatment. Nevertheless, given the heterogeneity of clinical efficacy, the multiplicity of treatment options available and the possibility of serious adverse effects, selecting the most effective treatment has become the greatest challenge. Molecular imaging offers an attractive way for this purpose. ImmunoPET provides specific imaging with positron emission tomography (PET) using monoclonal antibodies (mAb) or its fragments as vector. By combining the high targeting specificity of mAb and the sensitivity of PET technique, immunoPET could noninvasively and dynamically reveal tumor antigens expression and provide theranostic tools of several types of malignancies. Because of their slow kinetics, mAbs require radioelements defined by a consistent half-life. Zirconium 89 (89Zr) and Copper 64 (64Cu) are radiometals with half-lives suitable for mAb labeling. Radiolabeling with a radiometal requires the prior use of a bifunctional chelate agent (BFCA) to functionalize mAb for radiometal chelation, in a second step. There are a number of BFCA available and much research is focused on antibody functionalization techniques or on developing the optimum chelating agent depending the selected radiometal. In this manuscript, we present a critical account of radiochemical techniques with radionuclides 89Zr and 64Cu and their applications in preclinical and clinical immuno-PET imaging.
Collapse
Affiliation(s)
| | - Isabelle Quelven
- Toulouse NeuroImaging Center (ToNIC), INSERM/UPS UMR 1214, University Hospital of Toulouse-Purpan, CEDEX 3, 31024 Toulouse, France;
| |
Collapse
|
8
|
Ruby L, Jayaprakasam VS, Fernandes MC, Paroder V. Advances in the Imaging of Esophageal and Gastroesophageal Junction Malignancies. Hematol Oncol Clin North Am 2024; 38:711-730. [PMID: 38575457 DOI: 10.1016/j.hoc.2024.02.003] [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] [Indexed: 04/06/2024]
Abstract
Accurate imaging is key for the diagnosis and treatment of esophageal and gastroesophageal junction cancers . Current imaging modalities, such as computed tomography (CT) and 18F-FDG (2-deoxy-2-[18F]fluoro-D-glucose) positron emission tomography (PET)/CT, have limitations in accurately staging these cancers. MRI shows promise for T staging and residual disease assessment. Novel PET tracers, like FAPI, FLT, and hypoxia markers, offer potential improvements in diagnostic accuracy. 18F-FDG PET/MRI combines metabolic and anatomic information, enhancing disease evaluation. Radiomics and artificial intelligence hold promise for early detection, treatment planning, and response assessment. Theranostic nanoparticles and personalized medicine approaches offer new avenues for cancer therapy.
Collapse
Affiliation(s)
- Lisa Ruby
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Vetri Sudar Jayaprakasam
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Maria Clara Fernandes
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
9
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Cytryn SL, Pandit-Taskar N, Lumish MA, Maron SB, Gu P, Ku GY, Chou JF, Capanu M, Antoine A, Loegel D, Feder L, Philemond S, Lyashchenko SK, Lewis JS, Paroder V, Srivastava A, Tang LH, Schoder H, Janjigian YY. 18F-BMS-986229 PET to Assess Programmed-Death Ligand 1 Status in Gastroesophageal Cancer. J Nucl Med 2024; 65:722-727. [PMID: 38514081 PMCID: PMC11064823 DOI: 10.2967/jnumed.123.267186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Anti-programmed death 1 (PD-1) inhibitors are the standard of care for advanced gastroesophageal cancer. Although recommendations and approval by regulatory agencies are often based on programmed death ligand 1 (PD-L1) expression, pathologic assessments of PD-L1 status have several limitations. Single-site biopsies do not adequately capture disease heterogeneity within individual tumor lesions or among several lesions within the same patient, the PD-L1 combined positive score is a dynamic biomarker subject to evolution throughout a patient's disease course, and repeated biopsies are invasive and not always feasible. Methods: This was a prospective pilot study of the PD-L1-targeting radiotracer, 18F-BMS-986229, with PET imaging (PD-L1 PET) in patients with gastroesophageal cancer. Patients were administered the 18F-BMS-986229 radiotracer intravenously at a dose of 370 MBq over 1-2 min and underwent whole-body PET/CT imaging 60 min later. The primary objective of this study was to evaluate the safety and feasibility of 18F-BMS-986229. The trial is registered with ClinicalTrials.gov (NCT04161781). Results: Between February 3, 2020, and February 2, 2022, 10 patients with gastroesophageal adenocarcinoma underwent PD-L1 PET. There were no adverse events associated with the 18F-BMS-986229 tracer, and imaging did not result in treatment delays; the primary endpoint was achieved. Radiographic evaluation of PD-L1 expression was concordant with pathologic assessment in 88% of biopsied lesions, and 18F-BMS-986229 uptake on PET imaging correlated with pathologic evaluation by the combined positive score (Spearman rank correlation coefficient, 0.64). Seventy-one percent of patients with 18F-BMS-986229 accumulation on PET imaging also had lesions without 18F-BMS-986229 uptake, highlighting the intrapatient heterogeneity of PD-L1 expression. Patients treated with frontline programmed death 1 inhibitors who had 18F-BMS-986229 accumulation in any lesions on PET imaging had longer progression-free survival than patients without tracer accumulation in any lesions (median progression-free survival, 28.4 vs. 9.9 mo), though the small sample size prevents any definitive conclusions. Conclusion: PD-L1 PET imaging was safe, feasible, and concordant with pathologic evaluation and offers a potential noninvasive tool to assess PD-L1 expression.
Collapse
Affiliation(s)
- Samuel L Cytryn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Melissa A Lumish
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven B Maron
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Ping Gu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Geoffrey Y Ku
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Ariel Antoine
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Diane Loegel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Lara Feder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven Philemond
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Amitabh Srivastava
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Laura H Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schoder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
11
|
Stokke C, Gnesin S, Tran-Gia J, Cicone F, Holm S, Cremonesi M, Blakkisrud J, Wendler T, Gillings N, Herrmann K, Mottaghy FM, Gear J. EANM guidance document: dosimetry for first-in-human studies and early phase clinical trials. Eur J Nucl Med Mol Imaging 2024; 51:1268-1286. [PMID: 38366197 PMCID: PMC10957710 DOI: 10.1007/s00259-024-06640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/04/2024] [Indexed: 02/18/2024]
Abstract
The numbers of diagnostic and therapeutic nuclear medicine agents under investigation are rapidly increasing. Both novel emitters and novel carrier molecules require careful selection of measurement procedures. This document provides guidance relevant to dosimetry for first-in human and early phase clinical trials of such novel agents. The guideline includes a short introduction to different emitters and carrier molecules, followed by recommendations on the methods for activity measurement, pharmacokinetic analyses, as well as absorbed dose calculations and uncertainty analyses. The optimal use of preclinical information and studies involving diagnostic analogues is discussed. Good practice reporting is emphasised, and relevant dosimetry parameters and method descriptions to be included are listed. Three examples of first-in-human dosimetry studies, both for diagnostic tracers and radionuclide therapies, are given.
Collapse
Affiliation(s)
- Caroline Stokke
- Department of Diagnostic Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
- Department of Physics, University of Oslo, Oslo, Norway.
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Johannes Tran-Gia
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Francesco Cicone
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Søren Holm
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Marta Cremonesi
- Department of Medical Imaging and Radiation Sciences, European Institute of Oncology, IRCCS, Milan, Italy
| | - Johan Blakkisrud
- Department of Diagnostic Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Thomas Wendler
- Computer-Aided Medical Procedures and Augmented Reality, Technische Universität München, Munich, Germany
- Clinical Computational Medical Imaging Research, Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Augsburg, Augsburg, Germany
| | - Nic Gillings
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
- National Center for Tumor Diseases (NCT), NCT West, Heidelberg, Germany
| | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Jonathan Gear
- Joint Department of Physics, Royal Marsden NHSFT & Institute of Cancer Research, Sutton, UK
| |
Collapse
|
12
|
Yeh R, O'Donoghue JA, Jayaprakasam VS, Mauguen A, Min R, Park S, Brockway JP, Bromberg JF, Zhi WI, Robson ME, Sanford R, Modi S, Agnew BJ, Lyashchenko SK, Lewis JS, Ulaner GA, Zeglis BM. First-in-Human Evaluation of Site-Specifically Labeled 89Zr-Pertuzumab in Patients with HER2-Positive Breast Cancer. J Nucl Med 2024; 65:386-393. [PMID: 38272704 PMCID: PMC10924157 DOI: 10.2967/jnumed.123.266392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
Radioimmunoconjugates targeting human epidermal growth factor receptor 2 (HER2) have shown potential to noninvasively visualize HER2-positive tumors. However, the stochastic approach that has been traditionally used to radiolabel these antibodies yields poorly defined and heterogeneous products with suboptimal in vivo performance. Here, we describe a first-in-human PET study on patients with HER2-positive breast cancer evaluating the safety, biodistribution, and dosimetry of 89Zr-site-specific (ss)-pertuzumab PET, a site-specifically labeled radioimmunoconjugate designed to circumvent the limitations of random stochastic lysine labeling. Methods: Six patients with HER2-positive metastatic breast cancer were enrolled in a prospective clinical trial. Pertuzumab was site-specifically modified with desferrioxamine (DFO) via a novel chemoenzymatic strategy and subsequently labeled with 89Zr. Patients were administered 74 MBq of 89Zr-ss-pertuzumab in 20 mg of total antibody intravenously and underwent PET/CT at 1 d, 3-4 d, and 5-8 d after injection. PET imaging, whole-body probe counts, and blood draws were performed to assess the pharmacokinetics, biodistribution, and dosimetry. Results: 89Zr-ss-pertuzumab PET/CT was used to assess HER2 status and heterogeneity to guide biopsy and decide the next line of treatment at progression. The radioimmunoconjugate was able to detect known sites of malignancy, suggesting that these tumor lesions were HER2-positive. The optimal imaging time point was 5-8 d after administration, and no toxicities were observed. Dosimetry estimates from OLINDA showed that the organs receiving the highest doses (mean ± SD) were kidney (1.8 ± 0.5 mGy/MBq), liver (1.7 ± 0.3 mGy/MBq), and heart wall (1.2 ± 0.1 mGy/MBq). The average effective dose for 89Zr-ss-pertuzumab was 0.54 ± 0.03 mSv/MBq, which was comparable to both stochastically lysine-labeled 89Zr-DFO-pertuzumab and 89Zr-DFO-trastuzumab. One patient underwent PET/CT with both 89Zr-ss-pertuzumab and 89Zr-DFO-pertuzumab 1 mo apart, with 89Zr-ss-pertuzumab demonstrating improved lesion detection and higher tracer avidity. Conclusion: This study demonstrated the safety, dosimetry, and potential clinical applications of 89Zr-ss-pertuzumab PET/CT. 89Zr-ss-pertuzumab may detect more lesions than 89Zr-DFO-pertuzumab. Potential clinical applications include real-time evaluation of HER2 status to guide biopsy and assist in treatment decisions.
Collapse
Affiliation(s)
- Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vetri Sudar Jayaprakasam
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Audrey Mauguen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryan Min
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sue Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Julia P Brockway
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jacqueline F Bromberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - W Iris Zhi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rachel Sanford
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Brian J Agnew
- Biosciences Division, Thermo Fisher Scientific, Eugene, Oregon
| | - Serge K Lyashchenko
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gary A Ulaner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, California
- Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, California; and
| | - Brian M Zeglis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
- Department of Chemistry, Hunter College, New York, New York
| |
Collapse
|
13
|
Mohr P, van Sluis J, Lub-de Hooge MN, Lammertsma AA, Brouwers AH, Tsoumpas C. Advances and challenges in immunoPET methodology. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1360710. [PMID: 39355220 PMCID: PMC11440922 DOI: 10.3389/fnume.2024.1360710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 02/05/2024] [Indexed: 10/03/2024]
Abstract
Immuno-positron emission tomography (immunoPET) enables imaging of specific targets that play a role in targeted therapy and immunotherapy, such as antigens on cell membranes, targets in the disease microenvironment, or immune cells. The most common immunoPET applications use a monoclonal antibody labeled with a relatively long-lived positron emitter such as 89Zr (T 1/2 = 78.4 h), but smaller antibody-based constructs labeled with various other positron emitting radionuclides are also being investigated. This molecular imaging technique can thus guide the development of new drugs and may have a pivotal role in selecting patients for a particular therapy. In early phase immunoPET trials, multiple imaging time points are used to examine the time-dependent biodistribution and to determine the optimal imaging time point, which may be several days after tracer injection due to the slow kinetics of larger molecules. Once this has been established, usually only one static scan is performed and semi-quantitative values are reported. However, total PET uptake of a tracer is the sum of specific and nonspecific uptake. In addition, uptake may be affected by other factors such as perfusion, pre-/co-administration of the unlabeled molecule, and the treatment schedule. This article reviews imaging methodologies used in immunoPET studies and is divided into two parts. The first part summarizes the vast majority of clinical immunoPET studies applying semi-quantitative methodologies. The second part focuses on a handful of studies applying pharmacokinetic models and includes preclinical and simulation studies. Finally, the potential and challenges of immunoPET quantification methodologies are discussed within the context of the recent technological advancements provided by long axial field of view PET/CT scanners.
Collapse
Affiliation(s)
- Philipp Mohr
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joyce van Sluis
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Lammertsma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Charalampos Tsoumpas
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
14
|
Schomäcker K, Dietlein F, Muñoz Vázquez S, Braun F, Fischer T, Krapf P, Drzezga A, Dietlein M. From Bench to Bedside: Patient-Oriented Radiopharmaceutical Development in Nuclear Medicine Based on the Example of [ 89Zr]Zr-PSMA-DFO. Molecules 2023; 29:185. [PMID: 38202768 PMCID: PMC10780766 DOI: 10.3390/molecules29010185] [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: 11/21/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The interdisciplinary possibilities inherent in nuclear medicine offer an opportunity for the patient-centered development of radioactive pharmaceuticals based on specific research questions. This approach provides radiopharmaceutical manufacturers with a robust scientific foundation on which to navigate the regulatory requirements for drug approval laid down by the law. A vivid illustration of this interdisciplinary cooperation has been the development of a Zr-89-labeled PSMA ligand where reliable results have been obtained across various domains, including chemistry, radiochemistry, biochemistry, and preclinical research. This comprehensive process extended to feasibility studies conducted with carefully selected patients from a single nuclear medicine clinic. The approach demonstrates how far close collaboration between different disciplines within nuclear medicine can further the move towards patient-oriented radiopharmaceutical treatments while simultaneously meeting regulatory demands. With such a strategy, innovative radiopharmaceutical solutions can be brought to the market more swiftly and efficiently, in line with the needs of patients.
Collapse
Affiliation(s)
- Klaus Schomäcker
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
| | - Felix Dietlein
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
- Computational Health Informatics Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sergio Muñoz Vázquez
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
| | - Feodor Braun
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
| | - Thomas Fischer
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
| | - Philipp Krapf
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (F.D.); (S.M.V.); (F.B.); (T.F.); (P.K.); (A.D.); (M.D.)
| |
Collapse
|
15
|
Linguanti F, Abenavoli EM, Calabretta R, Berti V, Lopci E. ImmunoPET Targeting Receptor Tyrosine Kinase: Clinical Applications. Cancers (Basel) 2023; 15:5886. [PMID: 38136430 PMCID: PMC10741542 DOI: 10.3390/cancers15245886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Receptor tyrosine kinases, or RTKs, are one large family of cell surface receptors involved in signal transduction, which represent an integral part of the signaling pathways. They play a crucial role in most important cellular processes, starting with the cell cycle, proliferation and differentiation, as well as cell migration, metabolism and survival. The introduction of ImmunoPET evaluating the expression of RTKs by specific monoclonal antibodies (mAbs) or antibody fragments is regarded as a promising tool for imaging treatment efficacy and developing anticancer therapeutics. Our review focuses mainly on the current clinical research regarding ImmunoPET targeting RTKs, with particular interest in the epidermal growth factor family, or HER family, and vascular endothelial-derived growth factor/receptor.
Collapse
Affiliation(s)
- Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (F.L.); (V.B.)
- Nuclear Medicine Department, Ospedale San Donato, 52100 Arezzo, Italy
| | | | - Raffaella Calabretta
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Valentina Berti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (F.L.); (V.B.)
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS—Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| |
Collapse
|
16
|
Watabe T, Kabayama K, Naka S, Yamamoto R, Kaneda K, Serada S, Ooe K, Toyoshima A, Wang Y, Haba H, Kurimoto K, Kobayashi T, Shimosegawa E, Tomiyama N, Fukase K, Naka T. Immuno-PET and Targeted α-Therapy Using Anti-Glypican-1 Antibody Labeled with 89Zr or 211At: A Theranostic Approach for Pancreatic Ductal Adenocarcinoma. J Nucl Med 2023; 64:1949-1955. [PMID: 37827841 PMCID: PMC10690121 DOI: 10.2967/jnumed.123.266313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/06/2023] [Indexed: 10/14/2023] Open
Abstract
Glypican-1 (GPC1) is overexpressed in several solid cancers and is associated with tumor progression, whereas its expression is low in normal tissues. This study aimed to evaluate the potential of an anti-GPC1 monoclonal antibody (GPC1 mAb) labeled with 89Zr or 211At as a theranostic target in pancreatic ductal adenocarcinoma. Methods: GPC1 mAb clone 01a033 was labeled with 89Zr or 211At with a deferoxamine or decaborane linker, respectively. The internalization ability of GPC1 mAb was evaluated by fluorescence conjugation using a confocal microscope. PANC-1 xenograft mice (n = 6) were intravenously administered [89Zr]GPC1 mAb (0.91 ± 0.10 MBq), and PET/CT scanning was performed for 7 d. Uptake specificity was confirmed through a comparative study using GPC1-positive (BxPC-3) and GPC1-negative (BxPC-3 GPC1-knockout) xenografts (each n = 3) and a blocking study. DNA double-strand breaks were evaluated using the γH2AX antibody. The antitumor effect was evaluated by administering [211At]GPC1 mAb (∼100 kBq) to PANC-1 xenograft mice (n = 10). Results: GPC1 mAb clone 01a033 showed increased internalization ratios over time. One day after administration, a high accumulation of [89Zr]GPC1 mAb was observed in the PANC-1 xenograft (SUVmax, 3.85 ± 0.10), which gradually decreased until day 7 (SUVmax, 2.16 ± 0.30). The uptake in the BxPC-3 xenograft was significantly higher than in the BxPC-3 GPC1-knockout xenograft (SUVmax, 4.66 ± 0.40 and 2.36 ± 0.36, respectively; P = 0.05). The uptake was significantly inhibited in the blocking group compared with the nonblocking group (percentage injected dose per gram, 7.3 ± 1.3 and 12.4 ± 3.0, respectively; P = 0.05). DNA double-strand breaks were observed by adding 150 kBq of [211At]GPC1 and were significantly suppressed by the internalization inhibitor (dynasore), suggesting a substantial contribution of the internalization ability to the antitumor effect. Tumor growth suppression was observed in PANC-1 mice after the administration of [211At]GPC1 mAb. Internalization inhibitors (prochlorperazine) significantly inhibited the therapeutic effect of [211At]GPC1 mAb, suggesting an essential role in targeted α-therapy. Conclusion: [89Zr]GPC1 mAb PET showed high tumoral uptake in the early phase after administration, and targeted α-therapy using [211At]GPC1 mAb showed tumor growth suppression. GPC1 is a promising target for future applications for the precise diagnosis of pancreatic ductal adenocarcinoma and GPC1-targeted theranostics.
Collapse
Affiliation(s)
- Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan;
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | - Kazuya Kabayama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Sadahiro Naka
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Ryuku Yamamoto
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Kazuko Kaneda
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Suita, Japan
| | | | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, Saitama, Japan
| | - Kenta Kurimoto
- Department of Pharmacy, Osaka University Hospital, Suita, Japan
| | - Takanori Kobayashi
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Noriyuki Tomiyama
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita, Japan; and
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, Suita, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Toyonaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Tetsuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Yahaba, Japan
| |
Collapse
|
17
|
Hernandez MC, Yazaki P, Mortimer JE, Yamauchi D, Poku E, Park J, Frankel P, Kim J, Colcher DM, Wong J, Fong Y, Shively J, Woo Y. Pilot study of HER2 targeted 64 Cu-DOTA-tagged PET imaging in gastric cancer patients. Nucl Med Commun 2023; 44:1151-1155. [PMID: 37901917 PMCID: PMC10872802 DOI: 10.1097/mnm.0000000000001761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
OBJECTIVE Human epidermal growth factor receptor 2 (HER2) is an important biomarker for targeted gastric cancer (GC) immunotherapy. However, heterogeneous HER2 overexpression in GC, loss of HER2 expression during therapy, and inability to non-invasively identify HER2 overexpressing tumors impede effective targeting therapies. Improved HER2-specific functional imaging can address these challenges. Trastuzumab is a HER2-directed mAb to treat HER2 overexpressing cancers. The 64 Cu-DOTA-trastuzumab radiotracer is used to detect HER2+ metastatic breast cancer. We aimed to develop 64 Cu-DOTA-trastuzumab PET-CT to detect and characterize tumor uptake in HER2+ or - GC patients. METHODS We conducted a single-arm phase II pilot study exploring the feasibility of 64 Cu-DOTA-trastuzumab for PET imaging of HER2 overexpressing GC compared to HER2 non-expressing tumors. Eight patients with biopsy-confirmed gastric adenocarcinoma were included. Immunohistochemistry was used to evaluate primary tumor biopsies for HER2 overexpression. Patients were injected with 45 mg of cold trastuzumab followed by 5 mg of 64 Cu-DOTA-trastuzumab. PET-CT scans were performed 24-48 h post radiotracer injection and compared to standard staging CT scans. RESULTS We observed limited toxicity following 64 Cu-DOTA-trastuzumab injections. While there was uptake of the radiotracer in portions of HER2+ lesions, there was no statistically significant distinction between tumor and background by standardized uptake value analysis. CONCLUSION Despite the potential of 64 Cu-DOTA-trastuzumab PET imaging of HER2+ metastatic breast cancer, a 5 mg dose of this radiotracer injected 24-48 h before imaging was insufficient to identify HER2+ GC. These results inform future GC imaging studies to optimize biomarker-targeted therapies based on dosage and timing for more clinically relevant imaging.
Collapse
Affiliation(s)
- Matthew C. Hernandez
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Paul Yazaki
- Department of Molecular Imaging and Therapy, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Joanne E. Mortimer
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA
| | | | - Erasmus Poku
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Jinha Park
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA
| | - Paul Frankel
- Department of Biostatistics, Beckman Research Institute, Duarte, CA
| | - Joseph Kim
- Division of Surgical Oncology, Department of Surgery, UK Healthcare, University of Kentucky, Lexington, KY
| | - David M. Colcher
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA
| | - Jeffrey Wong
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Yuman Fong
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - John Shively
- Department of Immunology, Beckman Research Institute, City of Hope, Duarte, CA
| | - Yanghee Woo
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA
| |
Collapse
|
18
|
Surendra Panikar S, Shmuel S, Lewis JS, Pereira PMR. PET and Optical Imaging of Caveolin-1 in Gastric Tumors. ACS OMEGA 2023; 8:35884-35892. [PMID: 37810678 PMCID: PMC10552508 DOI: 10.1021/acsomega.3c03614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
Previous studies have suggested tumoral caveolin-1 (CAV1) as a predictive biomarker for the response to anti-HER2 antibody drug therapies in gastric tumors. In this study, radiolabeled and fluorescently labeled anti-CAV1 antibodies were developed and tested as an immunoPET or optical imaging agent to detect CAV1 in HER2-positive/CAV1-high NCIN87 gastric tumors. The expression of CAV1 receptors in NCIN87 gastric tumors and nontumor murine organs was determined by Western blot. Binding assays were performed to validate the anti-CAV1 antibody specificity for CAV1-expressing NCIN87 cancer cells. Subcutaneous and orthotopic NCIN87 xenografts were used for PET imaging and ex vivo biodistribution of the radioimmunoconjugate. Additional HER2-PET and CAV1-optical imaging was also performed to determine CAV1 in the HER2-positive tumors. 89Zr-labeled anti-CAV1 antibody was able to bind to CAV1-expressing NCIN87 cells with a Bmax value of 2.7 × 103 CAV1 receptors/cell in vitro. ImmunoPET images demonstrated the localization of the antibody in subcutaneous NCIN87 xenografts. In the orthotopic model, CAV1 expression was also observed by optical imaging in the HER2-positive tumors previously imaged with HER2-PET. Ex vivo biodistribution analysis further confirmed these imaging results. The preclinical data from this study demonstrate the potential of using CAV1-PET and optical imaging for detecting gastric tumors.
Collapse
Affiliation(s)
- Sandeep Surendra Panikar
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Shayla Shmuel
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Patrícia M. R. Pereira
- Department
of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
19
|
Ducharme M, Mansur A, Sligh L, Ulaner GA, Lapi SE, Sorace AG. Human Epidermal Growth Factor Receptor 2/Human Epidermal Growth Factor Receptor 3 PET Imaging: Challenges and Opportunities. PET Clin 2023; 18:543-555. [PMID: 37339919 DOI: 10.1016/j.cpet.2023.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) and HER3 provide actionable targets for both therapy and imaging in breast cancer. Further, clinical trials have shown the prognostic impact of receptor status discordance in breast cancer. Intra- and intertumoral heterogeneity of both HER and hormone receptor expression contributes to inherent errors in tissue sampling, and single biopsies are incapable of identifying discordance in biomarker expression. Numerous PET radiopharmaceuticals have been developed to evaluate (or target for therapy) HER2 and HER3 expression. This review seeks to inform on challenges and opportunities in HER2 and HER3 PET imaging in both clinical and preclinical settings.
Collapse
Affiliation(s)
- Maxwell Ducharme
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ameer Mansur
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Luke Sligh
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Irvine, CA, USA; Department of Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
20
|
Maron SB, Chatila W, Walch H, Chou JF, Ceglia N, Ptashkin R, Do RKG, Paroder V, Pandit-Taskar N, Lewis JS, Biachi De Castria T, Sabwa S, Socolow F, Feder L, Thomas J, Schulze I, Kim K, Elzein A, Bojilova V, Zatzman M, Bhanot U, Nagy RJ, Lee J, Simmons M, Segal M, Ku GY, Ilson DH, Capanu M, Hechtman JF, Merghoub T, Shah S, Schultz N, Solit DB, Janjigian YY. Determinants of Survival with Combined HER2 and PD-1 Blockade in Metastatic Esophagogastric Cancer. Clin Cancer Res 2023; 29:3633-3640. [PMID: 37406106 PMCID: PMC10502449 DOI: 10.1158/1078-0432.ccr-22-3769] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/21/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE We report updated clinical outcomes from a phase II study of pembrolizumab, trastuzumab, and chemotherapy (PTC) in metastatic esophagogastric cancer in conjunction with outcomes from an independent Memorial Sloan Kettering (MSK) cohort. PATIENTS AND METHODS The significance of pretreatment 89Zr-trastuzumab PET, plasma circulating tumor DNA (ctDNA) dynamics, and tumor HER2 expression and whole exome sequencing was evaluated to identify prognostic biomarkers and mechanisms of resistance in patients treated on-protocol with PTC. Additional prognostic features were evaluated using a multivariable Cox regression model of trastuzumab-treated MSK patients (n = 226). Single-cell RNA sequencing (scRNA-seq) data from MSK and Samsung were evaluated for mechanisms of therapy resistance. RESULTS 89Zr-trastuzumab PET, scRNA-seq, and serial ctDNA with CT imaging identified how pre-treatment intrapatient genomic heterogeneity contributes to inferior progression-free survival (PFS). We demonstrated that the presence of intensely avid lesions by 89Zr-trastuzumab PET declines in tumor-matched ctDNA by 3 weeks, and clearance of tumor-matched ctDNA by 9 weeks were minimally invasive biomarkers of durable PFS. Paired pre- and on-treatment scRNA-seq identified rapid clearance of HER2-expressing tumor clones with expansion of clones expressing a transcriptional resistance program, which was associated with MT1H, MT1E, MT2A, and MSMB expression. Among trastuzumab-treated patients at MSK, ERBB2 amplification was associated with improved PFS, while alterations in MYC and CDKN2A/B were associated with inferior PFS. CONCLUSIONS These findings highlight the clinical relevance of identifying baseline intrapatient heterogeneity and serial ctDNA monitoring of HER2-positive esophagogastric cancer patients to identify early evidence of treatment resistance, which could guide proactive therapy escalation or deescalation.
Collapse
Affiliation(s)
- Steven B. Maron
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Walid Chatila
- Tri-Institutional Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Henry Walch
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joanne F. Chou
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicholas Ceglia
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryan Ptashkin
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard Kinh Gian Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tiago Biachi De Castria
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shalom Sabwa
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fiona Socolow
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lara Feder
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jasmine Thomas
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Isabell Schulze
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kwanghee Kim
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Arijh Elzein
- Department of Pharmacology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, New York
| | - Viktoria Bojilova
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew Zatzman
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Umesh Bhanot
- Precision Pathology Center, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Marc Simmons
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michal Segal
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Geoffrey Yuyat Ku
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David H. Ilson
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Marinela Capanu
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jaclyn F. Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Taha Merghoub
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sohrab Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B. Solit
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée & Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yelena Y. Janjigian
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
21
|
Bragina O, Chernov V, Larkina M, Rybina A, Zelchan R, Garbukov E, Oroujeni M, Loftenius A, Orlova A, Sörensen J, Frejd FY, Tolmachev V. Phase I clinical evaluation of 99mTc-labeled Affibody molecule for imaging HER2 expression in breast cancer. Theranostics 2023; 13:4858-4871. [PMID: 37771776 PMCID: PMC10526658 DOI: 10.7150/thno.86770] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/27/2023] [Indexed: 09/30/2023] Open
Abstract
The determination of tumor human epidermal growth factor receptor type 2 (HER2) status is of increasing importance with the recent approval of more efficacious HER2-targeted treatments. There is a lack of suitable methods for clinical in vivo HER2 expression assessment. Affibody molecules are small affinity proteins ideal for imaging detection of receptors, which are engineered using a small (molecular weight 6.5 kDa) nonimmunoglobulin scaffold. Labeling of Affibody molecules with positron emitters enabled the development of sensitive and specific agents for molecular imaging. The development of probes for SPECT would permit the use of Affibody-based imaging in regions where PET is not available. In this first-in-human study, we evaluated the safety, biodistribution, and dosimetry of the 99mTc-ZHER2:41071 Affibody molecule developed for SPECT/CT imaging of HER2 expression. Methods: Thirty-one patients with primary breast cancer were enrolled and divided into three cohorts (injected with 500, 1000, or 1500 µg ZHER2:41071) comprising at least five patients with high (positive) HER2 tumor expression (IHC score 3+ or 2+ and ISH positive) and five patients with low (IHC score 2+ or 1+ and ISH negative) or absent HER2 tumor expression. Patients were injected with 451 ± 71 MBq 99mTc-ZHER2:4107. Planar scintigraphy was performed after 2, 4, 6 and 24 h, and SPECT/CT imaging followed planar imaging 2, 4 and 6 h after injection. Results: Injections of 99mTc-ZHER2:41071 were well tolerated and not associated with adverse events. Normal organs with the highest accumulation were the kidney and liver. The effective dose was 0.019 ± 0.004 mSv/MBq. Injection of 1000 µg provided the best standard discrimination between HER2-positive and HER2-low or HER2-negative tumors 2 h after injection (SUVmax 16.9 ± 7.6 vs. 3.6 ± 1.4, p < 0.005). The 99mTc-ZHER2:41071 uptake in HER2-positive lymph node metastases (SUVmax 6.9 ± 2.4, n = 5) was significantly (p < 0.05) higher than that in HER2-low/negative lymph nodes (SUVmax 3.5 ± 1.2, n = 4). 99mTc-ZHER2:41071 visualized hepatic metastases in a patient with liver involvement. Conclusions: Injections of 99mTc-ZHER2:41071 appear safe and exhibit favorable dosimetry. The protein dose of 1000 µg provides the best discrimination between HER2-positive and HER2-low/negative expression of HER2 according to the definition used for current HER2-targeting drugs.
Collapse
Affiliation(s)
- Olga Bragina
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Vladimir Chernov
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Mariia Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Anstasiya Rybina
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Roman Zelchan
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Eugeniy Garbukov
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | | | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Radiology and Nuclear Medicine, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Fredrik Y. Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
| |
Collapse
|
22
|
Vlastara M, Rossin R, Hoeben FJ, de Roode KE, Boswinkel M, Kleijn LH, Nagarajah J, Rijpkema M, Robillard MS. Click-to-Release: Cleavable Radioimmunoimaging with [ 89Zr]Zr-DFO- Trans-Cyclooctene-Trastuzumab Increases Tumor-to-Blood Ratio. Theranostics 2023; 13:4004-4015. [PMID: 37554267 PMCID: PMC10405837 DOI: 10.7150/thno.84865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 08/10/2023] Open
Abstract
One of the main challenges of PET imaging with 89Zr-labeled monoclonal antibodies (mAbs) remains the long blood circulation of the radiolabeled mAbs, leading to high background signals, decreasing image quality. To overcome this limitation, here we report the use of a bioorthogonal linker cleavage approach (click-to-release chemistry) to selectively liberate [89Zr]Zr-DFO from trans-cyclooctene-functionalized trastuzumab (TCO-Tmab) in blood, following the administration of a tetrazine compound (trigger) in BT-474 tumor-bearing mice. Methods: We created a series of TCO-DFO constructs and evaluated their performance in [89Zr]Zr-DFO release from Tmab in vitro using different trigger compounds. The in vivo behavior of the best performing [89Zr]Zr-TCO-Tmab was studied in healthy mice first to determine the optimal dose of the trigger. To find the optimal time for the trigger administration, the rate of [89Zr]Zr-TCO-Tmab internalization was studied in BT-474 cancer cells. Finally, the trigger was administered 6 h or 24 h after [89Zr]Zr-TCO-Tmab- administration in tumor-bearing mice to liberate the [89Zr]Zr-DFO fragment. PET scans were obtained of tumor-bearing mice that received the trigger 6 h post-[89Zr]Zr-TCO-Tmab administration. Results: The [89Zr]Zr-TCO-Tmab and trigger pair with the best in vivo properties exhibited 83% release in 50% mouse plasma. In tumor-bearing mice the tumor-blood ratios were markedly increased from 1.0 ± 0.4 to 2.3 ± 0.6 (p = 0.0057) and from 2.5 ± 0.7 to 6.6 ± 0.9 (p < 0.0001) when the trigger was administered at 6 h and 24 h post-mAb, respectively. Same day PET imaging clearly showed uptake in the tumor combined with a strongly reduced background due to the fast clearance of the released [89Zr]Zr-DFO-containing fragment from the circulation through the kidneys. Conclusions: This is the first demonstration of the use of trans-cyclooctene-tetrazine click-to-release chemistry to release a radioactive chelator from a mAb in mice to increase tumor-to-blood ratios. Our results suggest that click-cleavable radioimmunoimaging may allow for substantially shorter intervals in PET imaging with full mAbs, reducing radiation doses and potentially even enabling same day imaging.
Collapse
Affiliation(s)
- Maria Vlastara
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Raffaella Rossin
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | - Kim E. de Roode
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Milou Boswinkel
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | | | - James Nagarajah
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Marc S. Robillard
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| |
Collapse
|
23
|
Lumish MA, Maron SB, Paroder V, Chou JF, Capanu M, Philemond S, O'Donoghue JA, Schöder H, Lewis JS, Lyashchenko SK, Pandit-Taskar N, Janjigian YY. Noninvasive Assessment of Human Epidermal Growth Factor Receptor 2 (HER2) in Esophagogastric Cancer Using 89Zr-Trastuzumab PET: A Pilot Study. J Nucl Med 2023; 64:724-730. [PMID: 36418168 PMCID: PMC10152123 DOI: 10.2967/jnumed.122.264470] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Variations in human epidermal growth factor receptor 2 (HER2) expression between the primary tumor and metastases may contribute to drug resistance in HER2-positive (HER2+) metastatic esophagogastric cancer (mEGC). 89Zr-trastuzumab PET (HER2 PET) holds promise for noninvasive assessment of variations in HER2 expression and target engagement. The aim of this study was to describe HER2 PET findings in patients with mEGC. Methods: Patients with HER2+ mEGC were imaged with HER2 PET, 18F-FDG PET, and CT. Lesions were annotated using measurements (on CT) and maximum SUVs (on HER2 PET). Correlation of visualized disease burden among imaging modalities with clinical and pathologic characteristics was performed. Results: Thirty-three patients with HER2+ mEGC were imaged with HER2 PET and CT (12% esophageal, 64% gastroesophageal junction, and 24% gastric adenocarcinoma), 26 of whom were also imaged with 18F-FDG PET. More lesions were identified on 18F-FDG PET (median, 7 [range, 1-14]) than HER2 PET (median, 4 [range, 0-11]). Of the 8 lesions identified on HER2 but not on 18F-FDG PET, 3 (38%) were in bone and 1 was in the brain. Of the 68 lesions identified on 18F-FDG but not on HER2 PET, 4 (6%) were in bone and the remainder were in the lymph nodes (35, 51%) and liver (16, 24%). Of the 33 total patients, 23 (70%) were HER2 imaging-positive (≥50% of tumor load positive). Only 10 patients had 100% of the tumor load positive; 2 had 0% positive. When only patients receiving HER2-directed therapy as first-line treatment were considered (n = 13), median progression-free survival (PFS) therapy was not significantly different between HER2 imaging-positive and -negative patients. Median PFS for patients with at least 1 intense or very intense lesion (SUV ≥ 10) was 16 (95% CI: 11-not reached) mo (n = 7), compared with 12 (95% CI: 6.3-not reached) mo for patients without an intense or very intense lesion (n = 6) (P = 0.35). Conclusion: HER2 PET may identify heterogeneity of HER2 expression and allow assessment of lesions throughout the entire body. A potential application of HER2 PET is noninvasive evaluation of HER2 status including assessment of intrapatient disease heterogeneity not captured by standard imaging or single-site biopsies.
Collapse
Affiliation(s)
- Melissa A Lumish
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven B Maron
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Viktoriya Paroder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joanne F Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven Philemond
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schöder
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York and
| | - Serge K Lyashchenko
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York and
| | - Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Yelena Y Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
24
|
Melendez-Alafort L, Ferro-Flores G, De Nardo L, Ocampo-García B, Bolzati C. Zirconium immune-complexes for PET molecular imaging: Current status and prospects. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.215005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
25
|
Ma C, Wang X, Guo J, Yang B, Li Y. Challenges and future of HER2-positive gastric cancer therapy. Front Oncol 2023; 13:1080990. [PMID: 36793592 PMCID: PMC9924067 DOI: 10.3389/fonc.2023.1080990] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Gastric cancer is the fifth most common cancer worldwide, and the treatment of advanced gastric cancer has relatively little progress. With the continuous development of molecularly targeted therapy for tumors, it has been discovered that human epidermal growth factor receptor 2 (HER2) contributes to the poor prognosis and pathogenesis of various cancers. In order to treat HER2-positive advanced gastric cancer, Trastuzumab has emerged as the first first-line targeted medication used in conjunction with chemotherapy. The consequent trastuzumab resistance has become an important issue, and various new HER2-targeted gastric cancer drugs are emerging to address this challenge. This review's primary concern is the drug mechanism of various HER2-positive gastric cancer targeted therapy and fresh techniques of detection.
Collapse
Affiliation(s)
- Chenzhe Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiao Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiwu Guo
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Yumin Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
26
|
Chen Y, Hou X, Li D, Ding J, Liu J, Wang Z, Teng F, Li H, Zhang F, Gu Y, Yu S, Qian X, Yang Z, Zhu H. Development of a CLDN18.2-targeting Immuno-PET Probe for Non-invasive Imaging in Gastrointestinal Tumors. J Pharm Anal 2023; 13:367-375. [PMID: 37181294 PMCID: PMC10173170 DOI: 10.1016/j.jpha.2023.02.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023] Open
Abstract
Claudin18.2 (CLDN18.2) is a tight junction protein that is overexpressed in a variety of solid tumors such as gastrointestinal cancer and oesophageal cancer. It has been identified as a promising target and a potential biomarker to diagnose tumor, evaluate efficacy, and determine patient prognosis. TST001 is a recombinant humanized CLDN18.2 antibody that selectively binds to the extracellular loop of human Claudin18.2. In this study, we constructed a solid target radionuclide zirconium-89 (89Zr) labled-TST001 to detect the expression of in the human stomach cancer BGC823CLDN18.2 cell lines. The [89Zr]Zr-desferrioxamine (DFO)-TST001 showed high radiochemical purity (RCP, >99%) and specific activity (24.15 ± 1.34 GBq/μmol), and was stable in 5% human serum albumin, and phosphate buffer saline (>85% RCP at 96 h). The EC50 values of TST001 and DFO-TST001 were as high as 0.413 ± 0.055 and 0.361 ± 0.058 nM (P > 0.05), respectively. The radiotracer had a significantly higher average standard uptake values in CLDN18.2-positive tumors than in CLDN18.2-negative tumors (1.11 ± 0.02 vs. 0.49 ± 0.03, P = 0.0016) 2 days post injection (p.i.). BGC823CLDN18.2 mice models showed high tumor/muscle ratios 96 h p.i. with [89Zr]Zr-DFO-TST001 was much higher than those of the other imaging groups. Immunohistochemistry results showed that BGC823CLDN18.2 tumors were highly positive (+++) for CLDN18.2, while those in the BGC823 group did not express CLDN18.2 (-). The results of ex vivo biodistribution studies showed that there was a higher distribution in the BGC823CLDN18.2 tumor bearing mice (2.05 ± 0.16 %ID/g) than BGC823 mice (0.69 ± 0.02 %ID/g) and blocking group (0.72 ± 0.02 %ID/g). A dosimetry estimation study showed that the effective dose of [89Zr]Zr-DFO-TST001 was 0.0705 mSv/MBq, which is within the range of acceptable doses for nuclear medicine research. Taken together, these results suggest that Good Manufacturing Practices produced by this immuno-positron emission tomography probe can detect CLDN18.2-overexpressing tumors.
Collapse
|
27
|
The Role of Radiolabeled Monoclonal Antibodies in Cancer Imaging and ADC Treatment. Cancer J 2022; 28:446-453. [DOI: 10.1097/ppo.0000000000000625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
28
|
Augustin JE, Soussan P, Bass AJ. Targeting the complexity of ERBB2 biology in gastroesophageal carcinoma. Ann Oncol 2022; 33:1134-1148. [PMID: 35963482 DOI: 10.1016/j.annonc.2022.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 12/20/2022] Open
Abstract
ERBB2 is the most prominent therapeutic target in gastroesophageal adenocarcinoma (GEA). For two decades, trastuzumab was the only treatment available for GEA overexpressing ERBB2. Several drugs showing evidence of efficacy over or in complement to trastuzumab in breast cancer failed to show clinical benefit in GEA. This resistance to anti-ERBB2 therapy is peculiarly recurrent in GEA and is mostly due to tumor heterogeneity with the existence of low expressing ERBB2 tumor clones and loss of ERBB2 over time. The development of new ERBB2 testing strategies and the use of antibody-drug conjugates having a bystander effect are providing new tools to fight heterogeneity in ERBB2-positive GEA. Co-amplifications of tyrosine kinase receptors, alterations in mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3-kinase (PI3K) signaling pathways and in proteins controlling cell cycle are well known to contribute resistance to anti-ERBB2 therapy, and they can be targeted by dual therapy. Recently described, NF1 mutations are responsible for Ras phosphorylation and activation and can also be targeted by MEK/ERK inhibition along with anti-ERBB2 therapy. Multiple lines of evidence suggest that immune mechanisms involving antibody-dependent cell-mediated cytotoxicity are preponderant over intracellular signaling in anti-ERBB2 therapy action. A better comprehension of these mechanisms could leverage immune action of anti-ERBB2 therapy and elucidate efficacy of combinations associating immunotherapy and anti-ERBB2 therapy, as suggested by the recent intermediate positive results of the KEYNOTE-811 trial.
Collapse
Affiliation(s)
- J E Augustin
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, USA; Department of Pathology, Henri Mondor University Hospital, Assistance Publique - Hôpitaux de Paris, Créteil, France; INSERM U955 Team 18, Université Paris Est Créteil - Faculté de Médecine, Créteil, France
| | - P Soussan
- Institut National de la Santé et de la Recherche Médicale U938, Centre de Recherche Saint-Antoine, Sorbonne Université - Faculté Saint Antoine, Paris, France; Department of Virology, GHU Paris-Est, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - A J Bass
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, USA.
| |
Collapse
|
29
|
Lugat A, Bailly C, Chérel M, Rousseau C, Kraeber-Bodéré F, Bodet-Milin C, Bourgeois M. Immuno-PET: Design options and clinical proof-of-concept. Front Med (Lausanne) 2022; 9:1026083. [PMID: 36314010 PMCID: PMC9613928 DOI: 10.3389/fmed.2022.1026083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Radioimmunoconjugates have been used for over 30 years in nuclear medicine applications. In the last few years, advances in cancer biology knowledge have led to the identification of new molecular targets specific to certain patient subgroups. The use of these targets in targeted therapies approaches has allowed the developments of specifically tailored therapeutics for patients. As consequence of the PET-imaging progresses, nuclear medicine has developed powerful imaging tools, based on monoclonal antibodies, to in vivo characterization of these tumor biomarkers. This imaging modality known as immuno-positron emission tomography (immuno-PET) is currently in fastest-growing and its medical value lies in its ability to give a non-invasive method to assess the in vivo target expression and distribution and provide key-information on the tumor targeting. Currently, immuno-PET presents promising probes for different nuclear medicine topics as staging/stratification tool, theranostic approaches or predictive/prognostic biomarkers. To develop a radiopharmaceutical drug that can be used in immuno-PET approach, it is necessary to find the best compromise between the isotope choice and the immunologic structure (full monoclonal antibody or derivatives). Through some clinical applications, this paper review aims to discuss the most important aspects of the isotope choice and the usable proteic structure that can be used to meet the clinical needs.
Collapse
Affiliation(s)
- Alexandre Lugat
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France
| | - Clément Bailly
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Caroline Rousseau
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France,ARRONAX Cyclotron, Saint-Herblain, France,*Correspondence: Mickaël Bourgeois
| |
Collapse
|
30
|
Wijngaarden JE, Huisman MC, Pouw JEE, Menke-van der Houven van Oordt CW, Jauw YWS, Boellaard R. Optimal imaging time points considering accuracy and precision of Patlak linearization for 89Zr-immuno-PET: a simulation study. EJNMMI Res 2022; 12:54. [PMID: 36065038 PMCID: PMC9445120 DOI: 10.1186/s13550-022-00927-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022] Open
Abstract
Purpose Zirconium-89-immuno-positron emission tomography (89Zr-immuno-PET) has enabled visualization of zirconium-89 labelled monoclonal antibody (89Zr-mAb) uptake in organs and tumors in vivo. Patlak linearization of 89Zr-immuno-PET quantification data allows for separation of reversible and irreversible uptake, by combining multiple blood samples and PET images at different days. As one can obtain only a limited number of blood samples and scans per patient, choosing the optimal time points is important. Tissue activity concentration curves were simulated to evaluate the effect of imaging time points on Patlak results, considering different time points, input functions, noise levels and levels of reversible and irreversible uptake. Methods Based on 89Zr-mAb input functions and reference values for reversible (VT) and irreversible (Ki) uptake from literature, multiple tissue activity curves were simulated. Three different 89Zr-mAb input functions, five time points between 24 and 192 h p.i., noise levels of 5, 10 and 15%, and three reference Ki and VT values were considered. Simulated Ki and VT were calculated (Patlak linearization) for a thousand repetitions. Accuracy and precision of Patlak linearization were evaluated by comparing simulated Ki and VT with reference values. Results Simulations showed that Ki is always underestimated. Inclusion of time point 24 h p.i. reduced bias and variability in VT, and slightly reduced bias and variability in Ki, as compared to combinations of three later time points. After inclusion of 24 h p.i., minimal differences were found in bias and variability between different combinations of later imaging time points, despite different input functions, noise levels and reference values. Conclusion Inclusion of a blood sample and PET scan at 24 h p.i. improves accuracy and precision of Patlak results for 89Zr-immuno-PET; the exact timing of the two later time points is not critical. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00927-6.
Collapse
Affiliation(s)
- Jessica E Wijngaarden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands. .,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Marc C Huisman
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Johanna E E Pouw
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - C Willemien Menke-van der Houven van Oordt
- Department of Medical Oncology, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Yvonne W S Jauw
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Boelelaan 1117, Amsterdam, The Netherlands.,Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
31
|
Mortimer JE, Bading JR, Frankel PH, Carroll MI, Yuan Y, Park JM, Tumyan L, Gidwaney N, Poku EK, Shively JE, Colcher DM. Use of 64Cu-DOTA-Trastuzumab PET to Predict Response and Outcome of Patients Receiving Trastuzumab Emtansine for Metastatic Breast Cancer: A Pilot Study. J Nucl Med 2022; 63:1145-1148. [PMID: 34857660 PMCID: PMC9364339 DOI: 10.2967/jnumed.121.262940] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/08/2021] [Indexed: 02/03/2023] Open
Abstract
We hypothesized that functional imaging with 64Cu-DOTA-trastuzumab PET/CT would predict the response to the antibody-drug conjugate trastuzumab-emtansine (T-DM1). Methods: Ten women with metastatic human epidermal growth factor receptor 2-positive breast cancer underwent 18F-FDG PET/CT and 64Cu-DOTA-trastuzumab PET/CT on days 1 and 2 before treatment with T-DM1. Results: T-DM1-responsive patients had higher uptake than nonresponsive patients. Day 1 minimum SUVmax (5.6 vs. 2.8, P < 0.02), day 2 minimum SUVmax (8.1 vs. 3.2, P < 0.01), and day 2 average SUVmax (8.5 vs. 5.4, P < 0.05) for 64Cu-DOTA-trastuzumab all favored responding patients. Tumor-level response suggested threshold dependence on SUVmax Patients with a day 2 minimum SUVmax above versus below the threshold had a median time to treatment failure of 28 mo versus 2 mo (P < 0.02). Conclusion: Measurement of trastuzumab uptake in tumors via PET/CT is promising for identifying patients with metastatic breast cancer who will benefit from T-DM1.
Collapse
Affiliation(s)
- Joanne E. Mortimer
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California
| | - James R. Bading
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California
| | - Paul H. Frankel
- Department of Information Sciences, City of Hope, Duarte, California
| | - Mary I. Carroll
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California
| | - Yuan Yuan
- Department of Medical Oncology and Experimental Therapeutics, City of Hope, Duarte, California
| | - Jinha M. Park
- Department of Radiology, City of Hope, Duarte, California; and
| | - Lusine Tumyan
- Department of Radiology, City of Hope, Duarte, California; and
| | - Nikita Gidwaney
- Department of Radiology, City of Hope, Duarte, California; and
| | - Erasmus K. Poku
- Department of Cancer Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California
| | - John E. Shively
- Department of Cancer Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California
| | - David M. Colcher
- Department of Cancer Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California
| |
Collapse
|
32
|
Li L, Liu T, Shi L, Zhang X, Guo X, Hu B, Yao M, Zhu H, Yang Z, Jia B, Wang F. HER2-targeted dual radiotracer approach with clinical potential for noninvasive imaging of trastuzumab-resistance caused by epitope masking. Am J Cancer Res 2022; 12:5551-5563. [PMID: 35910795 PMCID: PMC9330517 DOI: 10.7150/thno.74154] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023] Open
Abstract
Rationale: The decreased HER2-accessibility by epitope masking is a primary trastuzumab-resistance mechanism. In this study, we developed a HER2-targeted dual radiotracer approach to predict the HER2-trastuzumab engagement noninvasively. Methods: Two novel HER2-specific VHHs, MIRC208 and MIRC213, were acquired by immunizing alpaca with human HER2 protein, and were site-specifically labeled with 99mTc. Biodistribution and SPECT/CT imaging studies were performed in mice bearing HER2-positive and HER2-negative tumors. The HER2 binding sites of 99mTc-MIRC208 and 99mTc-MIRC213 were investigated by cell binding and SPECT/CT imaging studies. We evaluated the therapeutic predictive ability of our dual-radiotracer imaging approach for trastuzumab treatment in mice bearing MUC4-positive tumors (trastuzumab-resistant JIMT-1 and 87MUC4) and MUC4-negative tumors (trastuzumab-sensitive 7HER2 and NCI-N87). The preliminary clinical studies of 99mTc-MIRC208 were performed in two patients with HER2-positive breast tumors. Results:99mTc-MIRC208 and 99mTc-MIRC213 clearly visualized HER2-positive tumors, but not HER2-negative tumors. 99mTc-MIRC208 competes with trastuzumab for HER2-binding while 99mTc-MIRC213 recognizes HER2 on an epitope that is not masked by MUC4. The SPECT/CT studies with 99mTc-MIRC208 and 99mTc-MIRC213 clearly showed that the MUC4-negative and trastuzumab-sensitive 7HER2 and NCI-N87 tumors had very similar tumor uptake with the SUV208/SUV213 (2 h) ratios of 1.11 ± 0.17 in 7HER2 and 1.25 ± 0.22 in NCI-N87. However, the MUC4-positive JIMT-1 tumors showed the decreased SUV208/SUV213 (2 h) ratio (0.63 ± 0.07), which correlated well with the low response rate to trastuzumab therapy. The SUV208/SUV213 (2 h) ratio was reduced to 0.72 ± 0.02 in MUC4-expressing NCI-N87 cells, and resulting in the decreased trastuzumab sensitivity, further supporting the correlation between the SUV208/SUV213 (2 h) ratio and trastuzumab-sensitivity. The primary and metastatic HER2-positive lesions of patients were clearly visualized by 99mTc-MIRC208 SPECT at 2 h post injection. Conclusion: Overall, we demonstrated that the dual radiotracer imaging strategy is a valid noninvasive approach for the cancer patient selection before trastuzumab therapy. 99mTc-MIRC213 SPECT is utilized to quantify the tumor HER2 expression and screen HER2-positive cancer patients, while 99mTc-MIRC208 SPECT is used to determine the HER2-accessibility of trastuzumab. The SUV208/SUV213 (2 h) ratio is an important biomarker to determine the responsiveness of trastuzumab therapy.
Collapse
Affiliation(s)
- Liqiang Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Linqing Shi
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xin Zhang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Biao Hu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Meinan Yao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,✉ Corresponding author:
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| |
Collapse
|
33
|
Manafi-Farid R, Ataeinia B, Ranjbar S, Jamshidi Araghi Z, Moradi MM, Pirich C, Beheshti M. ImmunoPET: Antibody-Based PET Imaging in Solid Tumors. Front Med (Lausanne) 2022; 9:916693. [PMID: 35836956 PMCID: PMC9273828 DOI: 10.3389/fmed.2022.916693] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a molecular imaging modality combining the high sensitivity of PET with the specific targeting ability of monoclonal antibodies. Various radioimmunotracers have been successfully developed to target a broad spectrum of molecules expressed by malignant cells or tumor microenvironments. Only a few are translated into clinical studies and barely into clinical practices. Some drawbacks include slow radioimmunotracer kinetics, high physiologic uptake in lymphoid organs, and heterogeneous activity in tumoral lesions. Measures are taken to overcome the disadvantages, and new tracers are being developed. In this review, we aim to mention the fundamental components of immunoPET imaging, explore the groundbreaking success achieved using this new technique, and review different radioimmunotracers employed in various solid tumors to elaborate on this relatively new imaging modality.
Collapse
Affiliation(s)
- Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Ataeinia
- Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shaghayegh Ranjbar
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Zahra Jamshidi Araghi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mobin Moradi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
34
|
Roviello G, Catalano M, Iannone LF, Marano L, Brugia M, Rossi G, Aprile G, Antonuzzo L. Current status and future perspectives in HER2 positive advanced gastric cancer. Clin Transl Oncol 2022; 24:981-996. [PMID: 35091998 DOI: 10.1007/s12094-021-02760-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023]
Abstract
Gastric cancer is one of the most common malignancy worldwide with a prognosis less than 1 year in unresectable or metastatic disease. HER2 expression is the main biomarker to lead the addition of trastuzumab to first line systemic chemotherapy improving the overall survival in advanced HER2-positivegastric adenocarcinoma. The inevitable development of resistance to trastuzumab remains a great problem inasmuch several treatment strategies that have proven effective in breast cancer failed to show clinical benefit in advanced gastric cancer. In this review, we summarize the available data on the mechanisms underlying primary and secondary resistance toHER2-targeted therapy and current challenges in the treatment of HER2-positive advanced gastric cancer refractory to trastuzumab. Further, we describe the prognostic value of new non-invasive screening techniques, the current development of novel agents such us HER2 antibody-drug conjugates and bispecific antibodies, and the strategies with antitumor activity on going.
Collapse
Affiliation(s)
- G Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139, Florence, Italy.
| | - M Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - L F Iannone
- Department of Health Science, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - L Marano
- Department of Medical, Surgical and NeuroSciences, Section of Surgery, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
| | - M Brugia
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - G Rossi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - G Aprile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - L Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, 50134, Florence, Italy
- Medical Oncology Unit, Careggi University Hospital, 50134, Florence, Italy
| |
Collapse
|
35
|
Altunay B, Morgenroth A, Mottaghy FM. Use of Radionuclide-Based Imaging Methods in Breast Cancer. Semin Nucl Med 2022; 52:561-573. [PMID: 35624034 DOI: 10.1053/j.semnuclmed.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 12/21/2022]
Abstract
Breast cancer is one of the most commonly occurring cancers in women globally and is the primary cause of cancer mortality in females. Thus, early and effective breast cancer diagnosis is crucial for enhancing the survival rate. Current standard diagnostic techniques to assess the hormone receptor status in biopsies include immunohistochemistry and fluorescence in situ hybridization. However, in recent years, there has been an increase in research on noninvasive techniques for molecular imaging of hormone receptors. These methods offer many advantages over conventional imaging, as repeated measurements can be used to capture heterogeneous tumor expression throughout the body, as well as transformations in receptor status during disease progression. Thus, the noninvasive method, as an adjunct to conventional imaging, offers the potential to improve patient selection, optimize dose and schedule, and streamline the assessment of response.
Collapse
Affiliation(s)
- Betül Altunay
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, Germany; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.
| |
Collapse
|
36
|
Bauer D, Visca H, Weerakkody A, Carter LM, Samuels Z, Kaminsky S, Andreev OA, Reshetnyak YK, Lewis JS. PET Imaging of Acidic Tumor Environment With 89Zr-labeled pHLIP Probes. Front Oncol 2022; 12:882541. [PMID: 35664740 PMCID: PMC9160799 DOI: 10.3389/fonc.2022.882541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
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.
Collapse
Affiliation(s)
- David Bauer
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hannah Visca
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Anuradha Weerakkody
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Lukas M. Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Zachary Samuels
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Spencer Kaminsky
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Oleg A. Andreev
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Yana K. Reshetnyak
- Department of Physics, University of Rhode Island, Kingston, RI, United States
| | - Jason S. Lewis
- Department of Radiology and the Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
- Department of Pharmacology Program, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
37
|
Firth G, Blower JE, Bartnicka JJ, Mishra A, Michaels AM, Rigby A, Darwesh A, Al-Salemee F, Blower PJ. Non-invasive radionuclide imaging of trace metal trafficking in health and disease: "PET metallomics". RSC Chem Biol 2022; 3:495-518. [PMID: 35656481 PMCID: PMC9092424 DOI: 10.1039/d2cb00033d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/07/2022] [Indexed: 12/05/2022] Open
Abstract
Several specific metallic elements must be present in the human body to maintain health and function. Maintaining the correct quantity (from trace to bulk) and location at the cell and tissue level is essential. The study of the biological role of metals has become known as metallomics. While quantities of metals in cells and tissues can be readily measured in biopsy and autopsy samples by destructive analytical techniques, their trafficking and its role in health and disease are poorly understood. Molecular imaging with radionuclides - positron emission tomography (PET) and single photon emission computed tomography (SPECT) - is emerging as a means to non-invasively study the acute trafficking of essential metals between organs, non-invasively and in real time, in health and disease. PET scanners are increasingly widely available in hospitals, and methods for producing radionuclides of some of the key essential metals are developing fast. This review summarises recent developments in radionuclide imaging technology that permit such investigations, describes the radiological and physicochemical properties of key radioisotopes of essential trace metals and useful analogues, and introduces current and potential future applications in preclinical and clinical investigations to study the biology of essential trace metals in health and disease.
Collapse
Affiliation(s)
- George Firth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Julia E Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Joanna J Bartnicka
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aishwarya Mishra
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Aidan M Michaels
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Alex Rigby
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Afnan Darwesh
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Fahad Al-Salemee
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St. Thomas' Hospital London UK
| |
Collapse
|
38
|
Aragon-Sanabria V, Aditya A, Zhang L, Chen F, Yoo B, Cao T, Madajewski B, Lee R, Turker MZ, Ma K, Monette S, Chen P, Wu J, Ruan S, Overholtzer M, Zanzonico P, Rudin CM, Brennan C, Wiesner U, Zhang L. Ultrasmall Nanoparticle Delivery of Doxorubicin Improves Therapeutic Index for High-Grade Glioma. Clin Cancer Res 2022; 28:2938-2952. [PMID: 35499557 DOI: 10.1158/1078-0432.ccr-21-4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/11/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Despite dramatic growth in the number of small molecule drugs developed to treat solid tumors, durable therapeutic options to control primary central nervous system malignancies are relatively scarce. Chemotherapeutic agents which appear biologically potent in model systems have often been found to be marginally effective at best when given systemically in clinical trials. This work presents for the first time an ultrasmall (< 8 nm) multimodal core-shell silica nanoparticle, Cornell prime dots (or C' dots), for the efficacious treatment of high-grade gliomas. Experimental Design: This work presents first-in-kind renally-clearable ultrasmall (< 8 nm) multimodal Cornell prime dots (or C' dots) with surface-conjugated doxorubicin via pH-sensitive linkers for the efficacious treatment in two different clinically relevant high-grade glioma models. Results: Optimal drug-per-particle ratios of as-developed nanoparticle-drug conjugates were established and used to obtain favorable pharmacokinetic profiles. The in vivo efficacy results showed significantly improved biological, therapeutic, and toxicological properties over the native drug after intravenous administration in platelet-derived growth factor-driven genetically engineered mouse model, and an epidermal growth factor expressing patient-derived xenograft (EGFR PDX) model. Conclusions: Ultrasmall C' dot-drug conjugates showed great translational potential over doxorubicin for improving the therapeutic outcome of patients with high-grade gliomas, even without a cancer-targeting moiety.
Collapse
Affiliation(s)
| | - Anusha Aditya
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Feng Chen
- Memorial Sloan Kettering Cancer Center, United States
| | | | - Tianye Cao
- Memorial Sloan Kettering Cancer Center, New York, United States
| | - Brian Madajewski
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | | | - Kai Ma
- Cornell University, Ithaca, NY, United States
| | - Sebastien Monette
- Memorial Sloan Kettering Cancer Center, The Rockefeller University, Weill Cornell Medicine, New York, New York, United States
| | - Peiming Chen
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jing Wu
- Hunter College, United States
| | - Shutian Ruan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Pat Zanzonico
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Charles M. Rudin
- Memorial Sloan Kettering Cancer Center, New York, New York, United States
| | - Cameron Brennan
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Li Zhang
- Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
39
|
Crișan G, Moldovean-Cioroianu NS, Timaru DG, Andrieș G, Căinap C, Chiș V. Radiopharmaceuticals for PET and SPECT Imaging: A Literature Review over the Last Decade. Int J Mol Sci 2022; 23:5023. [PMID: 35563414 PMCID: PMC9103893 DOI: 10.3390/ijms23095023] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) uses radioactive tracers and enables the functional imaging of several metabolic processes, blood flow measurements, regional chemical composition, and/or chemical absorption. Depending on the targeted processes within the living organism, different tracers are used for various medical conditions, such as cancer, particular brain pathologies, cardiac events, and bone lesions, where the most commonly used tracers are radiolabeled with 18F (e.g., [18F]-FDG and NA [18F]). Oxygen-15 isotope is mostly involved in blood flow measurements, whereas a wide array of 11C-based compounds have also been developed for neuronal disorders according to the affected neuroreceptors, prostate cancer, and lung carcinomas. In contrast, the single-photon emission computed tomography (SPECT) technique uses gamma-emitting radioisotopes and can be used to diagnose strokes, seizures, bone illnesses, and infections by gauging the blood flow and radio distribution within tissues and organs. The radioisotopes typically used in SPECT imaging are iodine-123, technetium-99m, xenon-133, thallium-201, and indium-111. This systematic review article aims to clarify and disseminate the available scientific literature focused on PET/SPECT radiotracers and to provide an overview of the conducted research within the past decade, with an additional focus on the novel radiopharmaceuticals developed for medical imaging.
Collapse
Affiliation(s)
- George Crișan
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | | | - Diana-Gabriela Timaru
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
| | - Gabriel Andrieș
- Department of Nuclear Medicine, County Clinical Hospital, Clinicilor 3-5, 400006 Cluj-Napoca, Romania;
| | - Călin Căinap
- The Oncology Institute “Prof. Dr. Ion Chiricuţă”, Republicii 34-36, 400015 Cluj-Napoca, Romania;
| | - Vasile Chiș
- Faculty of Physics, Babeş-Bolyai University, Str. M. Kogălniceanu 1, 400084 Cluj-Napoca, Romania; (G.C.); (N.S.M.-C.); (D.-G.T.)
- Institute for Research, Development and Innovation in Applied Natural Sciences, Babeș-Bolyai University, Str. Fântânele 30, 400327 Cluj-Napoca, Romania
| |
Collapse
|
40
|
Modulation of Secondary Cancer Risks from Radiation Exposure by Sex, Age and Gonadal Hormone Status: Progress, Opportunities and Challenges. J Pers Med 2022; 12:jpm12050725. [PMID: 35629147 PMCID: PMC9146871 DOI: 10.3390/jpm12050725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/18/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022] Open
Abstract
Available data on cancer secondary to ionizing radiation consistently show an excess (2-fold amount) of radiation-attributable solid tumors in women relative to men. This excess risk varies by organ and age, with the largest sex differences (6- to more than 10-fold) found in female thyroid and breasts exposed between birth until menopause (~50 years old) relative to age-matched males. Studies in humans and animals also show large changes in cell proliferation rates, radiotracer accumulation and target density in female reproductive organs, breast, thyroid and brain in conjunction with physiological changes in gonadal hormones during the menstrual cycle, puberty, lactation and menopause. These sex differences and hormonal effects present challenges as well as opportunities to personalize radiation-based treatment and diagnostic paradigms so as to optimize the risk/benefit ratios in radiation-based cancer therapy and diagnosis. Specifically, Targeted Radionuclide Therapy (TRT) is a fast-expanding cancer treatment modality utilizing radiopharmaceuticals with high avidity to specific molecular tumor markers, many of which are influenced by sex and gonadal hormone status. However, past and present dosimetry studies of TRT agents do not stratify results by sex and hormonal environment. We conclude that cancer management using ionizing radiation should be personalized and informed by the patient sex, age and hormonal status.
Collapse
|
41
|
Fay R, Törő I, Schinke AL, Simic B, Schaefer JV, Dreier B, Plückthun A, Holland JP. Sortase-Mediated Site-Specific Conjugation and 89Zr-Radiolabeling of Designed Ankyrin Repeat Proteins for PET. Mol Pharm 2022; 19:3576-3585. [DOI: 10.1021/acs.molpharmaceut.2c00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Rachael Fay
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Imre Törő
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anna-Lena Schinke
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Branko Simic
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jonas V. Schaefer
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Birgit Dreier
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jason P. Holland
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
42
|
A Physiologically Based Pharmacokinetic Framework for Quantifying Antibody Distribution Gradients from Tumors to Tumor-Draining Lymph Nodes. Antibodies (Basel) 2022; 11:antib11020028. [PMID: 35466281 PMCID: PMC9036243 DOI: 10.3390/antib11020028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Immune checkpoint blockades prescribed in the neoadjuvant setting are now under active investigation for many types of tumors, and many have shown early success. The primary tumor (PT) and tumor-draining lymph node (TDLN) immune factors, along with adequate therapeutic antibody distributions to the PT and TDLN, are critical for optimal immune activation and anti-tumor efficacy in neoadjuvant immunotherapy. However, it remains largely unknown how much of the antibody can be distributed into the PT-TDLN axis at different clinical scenarios. The goal of the current work is to build a physiologically based pharmacokinetic (PBPK) model framework capable of characterizing antibody distribution gradients in the PT-TDLN axis across various clinical and pathophysiological scenarios. The model was calibrated using clinical data from immuno-PET antibody-imaging studies quantifying antibody pharmacokinetics (PK) in the blood, PTs, and TDLNs. The effects of metastatic lesion location, tumor-induced compression, and inflammation, as well as surgery, on antibody concentration gradients in the PT-TDLN axis were characterized. The PBPK model serves as a valuable tool to predict antibody exposures in various types of tumors, metastases, and the associated lymph node, supporting effective immunotherapy.
Collapse
|
43
|
Sharma SK, Suzuki M, Xu H, Korsen JA, Samuels Z, Guo H, Nemieboka B, Piersigilli A, Edwards KJ, Cheung NKV, Lewis JS. Influence of Fc Modifications and IgG Subclass on Biodistribution of Humanized Antibodies Targeting L1CAM. J Nucl Med 2022; 63:629-636. [PMID: 34353869 PMCID: PMC8973293 DOI: 10.2967/jnumed.121.262383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/15/2021] [Indexed: 11/16/2022] Open
Abstract
Immuno-PET is a powerful tool to noninvasively characterize the in vivo biodistribution of engineered antibodies. Methods: L1 cell adhesion molecule-targeting humanized (HuE71) IgG1 and IgG4 antibodies bearing identical variable heavy- and light-chain sequences but different fragment crystallizable (Fc) portions were radiolabeled with 89Zr, and the in vivo biodistribution was studied in SKOV3 ovarian cancer xenografted nude mice. Results: In addition to showing uptake in L1 cell adhesion molecule-expressing SKOV3 tumors, as does its parental counterpart HuE71 IgG1, the afucosylated variant having enhanced Fc-receptor affinity showed high nonspecific uptake in lymph nodes. On the other hand, aglycosylated HuE71 IgG1 with abrogated Fc-receptor binding did not show lymphoid uptake. The use of the IgG4 subclass showed high nonspecific uptake in the kidneys, which was prevented by mutating serine at position 228 to proline in the hinge region of the IgG4 antibody to mitigate in vivo fragment antigen-binding arm exchange. Conclusion: Our findings highlight the influence of Fc modifications and the choice of IgG subclass on the in vivo biodistribution of antibodies and the potential outcomes thereof.
Collapse
Affiliation(s)
- Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maya Suzuki
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Hong Xu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua A Korsen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pharmacology, Weill Cornell Medical College, New York, New York
| | - Zachary Samuels
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hongfen Guo
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon Nemieboka
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alessandra Piersigilli
- Tri-Institutional Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, and Rockefeller University, New York, New York
| | - Kimberly J Edwards
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Pharmacology, Weill Cornell Medical College, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Radiology, Weill Cornell Medical College, New York, New York; and
- Radiochemistry and Molecular Imaging Probes Core, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
44
|
Hu G, Zhu W, Liu Y, Wang Y, Zhang Z, Zhu S, Duan W, Zhou P, Fu C, Li F, Huo L. Development and comparison of three 89Zr-labeled anti-CLDN18.2 antibodies to noninvasively evaluate CLDN18.2 expression in gastric cancer: a preclinical study. Eur J Nucl Med Mol Imaging 2022; 49:2634-2644. [DOI: 10.1007/s00259-022-05739-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/17/2022] [Indexed: 01/28/2023]
|
45
|
Parakh S, Lee ST, Gan HK, Scott AM. Radiolabeled Antibodies for Cancer Imaging and Therapy. Cancers (Basel) 2022; 14:1454. [PMID: 35326605 PMCID: PMC8946248 DOI: 10.3390/cancers14061454] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Radioimmunoconjugates consist of a monoclonal antibody (mAb) linked to a radionuclide. Radioimmunoconjugates as theranostics tools have been in development with success, particularly in hematological malignancies, leading to approval by the US Food and Drug Administration (FDA) for the treatment of non-Hodgkin's lymphoma. Radioimmunotherapy (RIT) allows for reduced toxicity compared to conventional radiation therapy and enhances the efficacy of mAbs. In addition, using radiolabeled mAbs with imaging methods provides critical information on the pharmacokinetics and pharmacodynamics of therapeutic agents with direct relevance to the optimization of the dose and dosing schedule, real-time antigen quantitation, antigen heterogeneity, and dynamic antigen changes. All of these parameters are critical in predicting treatment responses and identifying patients who are most likely to benefit from treatment. Historically, RITs have been less effective in solid tumors; however, several strategies are being investigated to improve their therapeutic index, including targeting patients with minimal disease burden; using pre-targeting strategies, newer radionuclides, and improved labeling techniques; and using combined modalities and locoregional application. This review provides an overview of the radiolabeled intact antibodies currently in clinical use and those in development.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
| | - Sze Ting Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
| | - Hui K. Gan
- Department of Medical Oncology, Heidelberg, VIC 3084, Australia; (S.P.); (H.K.G.)
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| | - Andrew M. Scott
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3086, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC 3010, Australia
| |
Collapse
|
46
|
Han J, Chen Y, Zhao Y, Zhao X, Zhang J, Wang J, Zhang Z. Pre-Clinical Study of the [ 18F]AlF-Labeled HER2 Affibody for Non-Invasive HER2 Detection in Gastric Cancer. Front Med (Lausanne) 2022; 9:803005. [PMID: 35252244 PMCID: PMC8890119 DOI: 10.3389/fmed.2022.803005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is an important biomarker in gastric cancer (GC) and directly influences the therapeutic effect. Fluorine is firmly bound to Al3+ forming [18F]AlF-1,4,7-triazacyclononanetriacetic acid (NOTA)-HER2 affibody is a promising radiolabeled tracer that can monitor the changes of HER2 expression combining the advantages of simple preparation and the properties of 18F. The aim of this study was to develop a quick method for the synthesis of [18F]AlF-NOTA-HER2 affibody and evaluate its utility for HER2+ GC imaging in mouse models. Moreover, 68Ga-NOTA-HER2 affibody imaging was also performed to highlight the superiority of [18F]AlF-NOTA-HER2 affibody imaging in resolution. The HER2 affibody was conjugated with NOTA and labeled using 18F based on the complexation of [18F]AlF by NOTA. Its quality control and stability were performed by high-pressure liquid chromatography (HPLC). The molecular specificity and binding affinity of the novel radiotracer were evaluated in the GC cell line with HER2 overexpression (NCI-N87) and negative expression (MKN74). Distribution studies and PET/CT imaging were performed in mouse models. 68Ga-NOTA-HER2 affibody PET/CT imaging was also performed. [18F]AlF-NOTA-HER2 affibody was efficiently prepared within 30 min with a non-decay-corrected maximum yield of 32.69% and a radiochemical purity of more than 98%. [18F]AlF-NOTA-HER2 affibody was highly stable in incubation medium for 4 h in vitro and in the blood of nude mice at 30 min post-injection (p.i.). In vitro studies revealed specific binding and high binding affinity of the probe in NCI-N87 cells, while no binding was seen in MKN74 cells. PET imaging showed that NCI-N87 xenografts were differentiated from MKN74 xenografts with excellent contrast and low abdominal background, which was confirmed by the distribution results. High-level accumulation of the [18F]AlF-NOTA-HER2 affibody in HER2+ tumors was blocked by excess unlabeled NOTA-HER2 affibody. [18F]AlF-NOTA-HER2 affibody has a higher image resolution than that of 68Ga-NOTA-HER2 affibody. [18F]AlF-NOTA-HER2 affibody could be produced facilely with high radiochemical yield and may serve as a novel molecular probe with tremendous clinical potential for the non-invasive whole-body detection of the HER2 status in GC with good image contrast and resolution. This method could provide an in vivo understanding of GC biology that will ultimately guide the accurate diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yang Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Zhao
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, China
| | - Jingmian Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianfang Wang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
47
|
Mini-review: Antibody-PET of receptor tyrosine kinase interplay and heterogeneity. Nucl Med Biol 2022; 108-109:70-75. [DOI: 10.1016/j.nucmedbio.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Accepted: 03/11/2022] [Indexed: 11/21/2022]
|
48
|
Review: Radionuclide Molecular Imaging Targeting HER2 in Breast Cancer with a Focus on Molecular Probes into Clinical Trials and Small Peptides. Molecules 2021; 26:molecules26216482. [PMID: 34770887 PMCID: PMC8588233 DOI: 10.3390/molecules26216482] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
As the most frequently occurring cancer worldwide, breast cancer (BC) is the leading cause of cancer-related death in women. The overexpression of HER2 (human epidermal growth factor receptor 2) is found in about 15% of BC patients, and it is often associated with a poor prognosis due to the effect on cell proliferation, migration, invasion, and survival. As a result of the heterogeneity of BC, molecular imaging with HER2 probes can non-invasively, in real time, and quantitatively reflect the expression status of HER2 in tumors. This will provide a new approach for patients to choose treatment options and monitor treatment response. Furthermore, radionuclide molecular imaging has the potential of repetitive measurements, and it can help solve the problem of heterogeneous expression and conversion of HER2 status during disease progression or treatment. Different imaging probes of targeting proteins, such as monoclonal antibodies, antibody fragments, nanobodies, and affibodies, are currently in preclinical and clinical development. Moreover, in recent years, HER2-specific peptides have been widely developed for molecular imaging techniques for HER2-positive cancers. This article summarized different types of molecular probes targeting HER2 used in current clinical applications and the developmental trend of some HER2-specific peptides.
Collapse
|
49
|
Sabanathan D, Lund ME, Campbell DH, Walsh BJ, Gurney H. Radioimmunotherapy for solid tumors: spotlight on Glypican-1 as a radioimmunotherapy target. Ther Adv Med Oncol 2021; 13:17588359211022918. [PMID: 34646364 PMCID: PMC8504276 DOI: 10.1177/17588359211022918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Radioimmunotherapy (i.e., the use of radiolabeled tumor targeting antibodies) is an emerging approach for the diagnosis, therapy, and monitoring of solid tumors. Often using paired agents, each targeting the same tumor molecule, but labelled with an imaging or therapeutic isotope, radioimmunotherapy has achieved promising clinical results in relatively radio-resistant solid tumors such as prostate. Several approaches to optimize therapeutic efficacy, such as dose fractionation and personalized dosimetry, have seen clinical success. The clinical use and optimization of a radioimmunotherapy approach is, in part, influenced by the targeted tumor antigen, several of which have been proposed for different solid tumors. Glypican-1 (GPC-1) is a heparan sulfate proteoglycan that is expressed in a variety of solid tumors, but whose expression is restricted in normal adult tissue. Here, we discuss the preclinical and clinical evidence for the potential of GPC-1 as a radioimmunotherapy target. We describe the current treatment paradigm for several solid tumors expressing GPC-1 and suggest the potential clinical utility of a GPC-1 directed radioimmunotherapy for these tumors.
Collapse
Affiliation(s)
- Dhanusha Sabanathan
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | | | | | | | - Howard Gurney
- Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, Sydney, NSW 2109, Australia
| |
Collapse
|
50
|
Radiopharmaceuticals developed for 89Zr-Immuno-PET. J Radioanal Nucl Chem 2021. [DOI: 10.1007/s10967-021-07922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|