1
|
Chandekar KR, Satapathy S, Dharmashaktu Y, Ballal S, Ranjan P, Batra A, Gogia A, Mathur S, Bal C. Somatostatin receptor-targeted theranostics in patients with estrogen receptor-positive metastatic breast cancer-a prospective exploratory study. Breast Cancer Res Treat 2025; 211:363-373. [PMID: 40000538 DOI: 10.1007/s10549-025-07651-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Somatostatin receptor (SSTR) expression has been reported in estrogen receptor-positive (ER +) metastatic breast cancer (mBC) by pathology and immunohistochemistry studies. We aimed to investigate whether SSTR could be a viable target for PET imaging and potential theranostics in ER + mBC. METHODS Thirty prospectively recruited patients with ER + mBC underwent PET/CT imaging with [18F]FDG and [68Ga]Ga-DOTATATE (within three weeks). Detection rates (per-patient, per-region), number of lesions detected, SUVmax values, Krenning scores, SSTR-FDG visual scores, and PET-based staging with both radiotracers were compared. RESULTS [18F]FDG and [68Ga]Ga-DOTATATE PET/CT had similar per-patient detection rates (100% vs 96.7%, P = 1.0). Per-region and per-lesion analyses revealed comparable detection of local/breast lesions, nodal, and skeletal metastases. However, [18F]FDG outperformed [68Ga]Ga-DOTATATE in detecting visceral/other metastases (235 vs 128 lesions, P = 0.003). [68Ga]Ga-DOTATATE resulted in a lower PET-based M-stage compared to [18F]FDG in 10% of patients, although T-/N-stages were concordant in all patients. HER2- patients showed a trend of higher [68Ga]Ga-DOTATATE lesional SUVmax values compared to the HER2 + sub-group (median 9.0 vs 3.8, P = 0.078). 3/30 (10%) participants had a patient-level Krenning score ≥ 3 ([68Ga]Ga-DOTATATE uptake higher than liver background in majority of the lesions), potentially making them suitable for SSTR-targeted radionuclide therapy. CONCLUSIONS SSTR-targeted theranostics may represent a novel potential alternative in a subset of patients with ER + mBC. Its generalized applicability is limited by poor sensitivity for visceral metastases and significant inter-lesion heterogeneity. Future studies must identify how tumor subtype, proliferation, and prior systemic therapies impact SSTR expression levels in these patients to ensure meaningful clinical translation. CLINICAL TRIAL REGISTRATION Clinical Trials Registry-India: CTRI/2023/03/051025 (prospectively registered on 23.03.2023).
Collapse
Affiliation(s)
- Kunal Ramesh Chandekar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Swayamjeet Satapathy
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Yamini Dharmashaktu
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sanjana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Piyush Ranjan
- Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Atul Batra
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ajay Gogia
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
2
|
Morgat C, Duan H, Dalm S, Hindié E, Günther T, Krause BJ, Kramer V, Cavelier F, Stephens AW, Moran S, Lamb L, Iagaru A. A Vision for Gastrin-Releasing Peptide Receptor Targeting for Imaging and Therapy: Perspective from Academia and Industry. J Nucl Med 2025:jnumed.124.269444. [PMID: 40341094 DOI: 10.2967/jnumed.124.269444] [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: 12/29/2024] [Accepted: 03/25/2025] [Indexed: 05/10/2025] Open
Abstract
The gastrin-releasing peptide receptor (GRPR) is overexpressed in various cancers, including prostate cancer, breast cancer, small cell and non-small cell lung cancer, uterine and ovarian cancer, colon cancer, and gastrointestinal stromal tumors. This makes GRPR a multicancer target for theranostics, that is, molecular imaging and therapy. Here, we explore the current state of GRPR-targeted theranostics from bench to bedside, highlighting the preclinical development of various GRPR-targeting compounds and clinical applications. We review the role of GRPR-targeted molecular imaging for all stages of prostate cancer, breast cancer, and other tumors and provide a quo vadis GRPR. We aimed to offer a comprehensive overview of GRPR-targeted theranostics to inform researchers, clinicians, pharma, and regulators of the potential benefits and emerging opportunities in the pursuit of personalized precision cancer care.
Collapse
Affiliation(s)
- Clément Morgat
- Nuclear Medicine Department, Bordeaux University Hospital, Bordeaux, France
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, Bordeaux, France
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Simone Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Elif Hindié
- Nuclear Medicine Department, Bordeaux University Hospital, Bordeaux, France
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, Bordeaux, France
- Institut Universitaire de France, Paris, France
| | - Thomas Günther
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | | | - Vasko Kramer
- Nuclear Medicine and PET/CT Center PositronMed, Santiago, Chile
- Positronpharma SA, Santiago, Chile
| | - Florine Cavelier
- Pôle Chime Balard, IBMM, UMR 5247 CNRS, Université Montpellier ENSCM, Montpellier, France
| | | | | | - Laura Lamb
- Clarity Pharmaceuticals Ltd., Sydney, Australia
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California;
| |
Collapse
|
3
|
Giugliano F, Giordano E, Gilardi L, Salimbeni BT, Zagami P, Esposito A, Marra A, Trapani D, Berton Giachetti PPM, Malagutti B, Henry T, Deandreis D, Curigliano G, Ceci F, Criscitiello C. Radioligand Therapy in Metastatic Breast Cancer: Harnessing Precision Oncology. Cancer Treat Rev 2025; 136:102940. [PMID: 40228448 DOI: 10.1016/j.ctrv.2025.102940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Radioligand therapy (RLT) represents a promising advancement in precision oncology and enables the targeted delivery of radiation to cancer cells. This approach has shown success in other tumor types, such as prostate cancer and neuroendocrine tumors. Its potential in metastatic breast cancer (mBC) is currently under investigation. This review discusses RLT mechanism of action, therapeutic potential, and integration into the existing therapeutic landscape of mBC. While clinical trials have shown promising results, challenges remain regarding target heterogeneity, implementation, and optimizing treatment strategies. Further research is essential to integrate RLT into clinical practice and improve patient outcomes fully.
Collapse
Affiliation(s)
- Federica Giugliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Inserm U981, Gustave Roussy Cancer Campus, Villejuif, France
| | - Elisa Giordano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Laura Gilardi
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Paola Zagami
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Angela Esposito
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Antonio Marra
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Pier Paolo Maria Berton Giachetti
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Bianca Malagutti
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Théophraste Henry
- Division of Nuclear Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Desiree Deandreis
- Division of Nuclear Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Francesco Ceci
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy.
| |
Collapse
|
4
|
Lynch SE, Crawford CI, Houson HA, Omweri JM, Pukkanasut P, Gallegos CA, Whitt JD, Jaskula-Sztul R, Lapi SE, Sorace AG. Characterizing SSTR2 expression and modulation for targeted imaging and therapy in preclinical models of triple-negative breast cancer. Sci Rep 2025; 15:9988. [PMID: 40121305 PMCID: PMC11929780 DOI: 10.1038/s41598-025-94578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Patients with breast cancer which lack molecular targets, such as human epidermal growth factor receptor 2 (HER2) or hormone receptors, have limited access to targeted therapies. Somatostatin receptor 2 (SSTR2) is overexpressed in some cancers, and SSTR2-targeted radiopharmaceuticals are FDA-approved for theranostic targeted imaging and therapy in neuroendocrine tumors (NETs). Importantly, histone deacetylase (HDAC) inhibitors can epigenetically modulate SSTR2 expression in NETs with low or variable basal expression. The goal of this study is to characterize SSTR2 basal expression and induction via HDAC inhibition as a potential target for imaging and therapy in preclinical models of triple-negative breast cancer (TNBC). SSTR2 expression in mouse samples was assessed via Western blot and immunohistochemistry. Real-time quantitative PCR (qRT-PCR), flow cytometry, and cell binding assays were utilized to determine if HDAC inhibition can upregulate SSTR2 expression. [68Ga]Ga-DOTATATE positron emission tomography (PET) imaging, which targets SSTR2, was used to non-invasively characterize SSTR2 expression and variability in the EO771 and 4T1 TNBC models before and after HDAC inhibition. These studies demonstrate that HDAC inhibition can upregulate SSTR2 at the transcriptional, translational, and functional levels in breast cancer. Importantly, SSTR2 expression can be characterized non-invasively via PET imaging and modulation with HDAC inhibitors can be monitored longitudinally. Our findings highlight SSTR2 as a promising therapeutic molecular target in TNBC.
Collapse
Affiliation(s)
- Shannon E Lynch
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Corinne I Crawford
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey A Houson
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
| | - James M Omweri
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Carlos A Gallegos
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason D Whitt
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, VH G082, 1670 University Blvd, Birmingham, AL, 35233, USA.
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Lichtenfels M, Almeida Lima Nunes R, Mendoza López RV, Alves da Silva C, Zeferino LC, de Souza Lino V, Longatto-Filho A, De Brot L, Rabelo-Santos SH, Cornelio DB, Boccardo E, de Farias CB, Termini L. Gastrin-releasing peptide receptor: a promising new biomarker to identify cervical precursor lesions and cancer. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2025; 47:e-rbgo4. [PMID: 40242010 PMCID: PMC12002723 DOI: 10.61622/rbgo/2025rbgo4] [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: 05/22/2024] [Accepted: 10/21/2024] [Indexed: 04/18/2025] Open
Abstract
Objective This study aimed to verify the relation between gastrin-releasing peptide receptor (GRPR), oncogenic Human Papillomavirus (HPV) and cervical lesions severity. Methods GRPR mRNA levels were evaluated in cervical cancer-derived cell lines and in primary keratinocytes expressing HPV16 oncogenes by RT-PCR. GRPR protein expression was assessed by immunohistochemistry in organotypic cell cultures derived from keratinocytes transduced with HPV16 oncogenes and in 208 cervical samples, including 59 non-neoplastic tissue, 28 cervical intraepithelial neoplasia grade 3 (CIN3), 44 squamous cell carcinomas (SCC) and 77 adenocarcinomas (ADC). Generic primers (GP5+/GP6+) were used to identify HPV infection in tissue samples. Experiments involving cell lines were analyzed through non-parametric tests (Kruskal Wallis), and Fisher's Exact Test for human tissues samples. All statistical tests were considered significant at p <0.05. Immunohistochemical evaluation was conducted independently and blindly by two observers (AD- LO). Any discordant findings were resolved through discussion to reach a consensus score. Results GRPR mRNA levels were not increased in cells expressing HPV16 or HPV18 oncogenes. However, at the protein level, GRPR was upregulated in organotypic cell cultures containing HPV oncogenes. Besides, it was identified an association between GRPR expression and cervical lesion severity (p < 0.0001). The detection rate of high-risk HPV DNA was directly correlated with cervical disease. Nonetheless, HPV infection was not directly associated with GRPR in cervical samples. Conclusion GRPR expression is highly predictive of cervical lesion severity, irrespective of HPV infection and might contribute to improving patient's therapeutic management as well as being used a marker of disease progression.
Collapse
Affiliation(s)
- Martina Lichtenfels
- Ziel BiosciencesPorto AlegreRio Grande do SulBrazilZiel Biosciences, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Rafaella Almeida Lima Nunes
- Universidade de Sao PauloFaculdade de MedicinaHospital das Clinicas HCFMUSPSão PauloBrazilCenter for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil.
- Universidade de Sao PauloComprehensive Center for Precision OncologySão PauloBrazilComprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil.
| | - Rossana Veronica Mendoza López
- Universidade de Sao PauloFaculdade de MedicinaHospital das Clinicas HCFMUSPSão PauloBrazilCenter for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil.
- Universidade de Sao PauloComprehensive Center for Precision OncologySão PauloBrazilComprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil.
| | - Camila Alves da Silva
- Ziel BiosciencesPorto AlegreRio Grande do SulBrazilZiel Biosciences, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Luiz Carlos Zeferino
- State University of CampinasFaculty of Medical SciencesDivision of Gynecologic and Breast OncologySão PauloBrazilDepartment of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, Faculty of Medical Sciences, State University of Campinas (UNICAMP – Universidade Estadual de Campinas), São Paulo, Brazil.
| | - Vanesca de Souza Lino
- Universidade de São PauloInstituto de Ciências BiomédicasDepartment of MicrobiologySão PauloBrazilDepartment of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.
| | - Adhemar Longatto-Filho
- University of São PauloSchool of MedicineDepartment of PathologySão PauloBrazilLaboratory of Medical Investigation (LIM) 14, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil;
- University of MinhoSchool of MedicineLife and Health Sciences Research InstituteBragaPortugalLife and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's;
- Pio XII FoundationBarretos Cancer HospitalMolecular Oncology Research CenterSão PauloBrazilMolecular Oncology Research Center, Barretos Cancer Hospital, Pio XII Foundation, Barretos, São Paulo, Brazil.
| | - Louise De Brot
- AC Camargo Cancer CenterDepartment of PathologySão PauloBrazilDepartment of Pathology, AC Camargo Cancer Center, São Paulo, Brazil.
| | - Silvia Helena Rabelo-Santos
- Universidade Federal de GoiásFaculdade de FarmáciaGoiásBrazilFaculdade de Farmácia, Universidade Federal de Goiás (UFG), Goiás, Brazil.
| | - Daniela Baumann Cornelio
- Irmandade Santa Casa de Porto AlegreRio Grande do SulBrazilIrmandade Santa Casa de Porto Alegre, Rio Grande do Sul, Brazil
| | - Enrique Boccardo
- Universidade de São PauloInstituto de Ciências BiomédicasDepartment of MicrobiologySão PauloBrazilDepartment of Microbiology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil.
| | - Caroline Brunetto de Farias
- Ziel BiosciencesPorto AlegreRio Grande do SulBrazilZiel Biosciences, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Lara Termini
- Universidade de Sao PauloFaculdade de MedicinaHospital das Clinicas HCFMUSPSão PauloBrazilCenter for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil.
- Universidade de Sao PauloComprehensive Center for Precision OncologySão PauloBrazilComprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil.
| |
Collapse
|
6
|
Bokhout L, Campeiro JD, Dalm SU. Exploring the landscape of current in vitro and in vivo models and their relevance for targeted radionuclide theranostics. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07123-3. [PMID: 40016527 DOI: 10.1007/s00259-025-07123-3] [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: 10/07/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains a leading cause of mortality globally, driving ongoing research into innovative treatment strategies. Preclinical research forms the base for developing these novel treatments, using both in vitro and in vivo model systems that are, ideally, as clinically representative as possible. Emerging as a promising approach for cancer management, targeted radionuclide theranostics (TRT) uses radiotracers to deliver (cytotoxic) radionuclides specifically to cancer cells. Since the field is relatively new, more advanced preclinical models are not yet regularly applied in TRT research. This narrative review examines the currently applied in vitro, ex vivo and in vivo models for oncological research, discusses if and how these models are now applied for TRT studies, and whether not yet applied models can be of benefit for the field. A selection of different models is discussed, ranging from in vitro two-dimensional (2D) and three-dimensional (3D) cell models, including spheroids, organoids and tissue slice cultures, to in vivo mouse cancer models, such as cellline-derived models, patient-derived xenograft models and humanized models. Each of the models has advantages and limitations for studying human cancer biology, radiopharmaceutical assessment and treatment efficacy. Overall, there is a need to apply more advanced models in TRT research that better address specific TRT phenomena, such as crossfire and abscopal effects, to enhance the clinical relevance and effectiveness of preclinical TRT evaluations.
Collapse
Affiliation(s)
- Lisa Bokhout
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joana D Campeiro
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Zhang T, An W, You S, Chen S, Zhang S. G protein-coupled receptors and traditional Chinese medicine: new thinks for the development of traditional Chinese medicine. Chin Med 2024; 19:92. [PMID: 38956679 PMCID: PMC11218379 DOI: 10.1186/s13020-024-00964-4] [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: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
G protein-coupled receptors (GPCRs) widely exist in vivo and participate in many physiological processes, thus emerging as important targets for drug development. Approximately 30% of the Food and Drug Administration (FDA)-approved drugs target GPCRs. To date, the 'one disease, one target, one molecule' strategy no longer meets the demands of drug development. Meanwhile, small-molecule drugs account for 60% of FDA-approved drugs. Traditional Chinese medicine (TCM) has garnered widespread attention for its unique theoretical system and treatment methods. TCM involves multiple components, targets and pathways. Centered on GPCRs and TCM, this paper discusses the similarities and differences between TCM and GPCRs from the perspectives of syndrome of TCM, the consistency of TCM's multi-component and multi-target approaches and the potential of GPCRs and TCM in the development of novel drugs. A novel strategy, 'simultaneous screening of drugs and targets', was proposed and applied to the study of GPCRs. We combine GPCRs with TCM to facilitate the modernisation of TCM, provide valuable insights into the rational application of TCM and facilitate the research and development of novel drugs. This study offers theoretical support for the modernisation of TCM and introduces novel ideas for development of safe and effective drugs.
Collapse
Affiliation(s)
- Ting Zhang
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China
| | - Wenqiao An
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China
| | - Shengjie You
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shilin Chen
- Institute of Herbgenomics, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sanyin Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611100, China.
| |
Collapse
|
8
|
Ma Y, Gao F. Advances of radiolabeled GRPR ligands for PET/CT imaging of cancers. Cancer Imaging 2024; 24:19. [PMID: 38279185 PMCID: PMC10811881 DOI: 10.1186/s40644-024-00658-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
GRPR is a type of seven-transmembrane G-protein coupled receptor that belongs to the bombesin protein receptor family. It is highly expressed in various cancers, including prostate cancer, breast cancer, lung cancer, gastrointestinal cancer, and so on. As a result, molecular imaging studies have been conducted using radiolabeled GRPR ligands for tumor diagnosis, as well as monitoring of recurrence and metastasis. In this paper, we provided a comprehensive overview of relevant literature from the past two decades, with a specific focus on the advancements made in radiolabeled GRPR ligands for imaging prostate cancer and breast cancer.
Collapse
Affiliation(s)
- Yuze Ma
- Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Feng Gao
- Research Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
9
|
D’Onofrio A, Engelbrecht S, Läppchen T, Rominger A, Gourni E. GRPR-targeting radiotheranostics for breast cancer management. Front Med (Lausanne) 2023; 10:1250799. [PMID: 38020178 PMCID: PMC10657217 DOI: 10.3389/fmed.2023.1250799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Breast Cancer (BC) is the most common cancer worldwide and, despite the advancements made toward early diagnosis and novel treatments, there is an urgent need to reduce its mortality. The Gastrin-Releasing Peptide Receptor (GRPR) is a promising target for the development of theranostic radioligands for luminal BC with positive estrogen receptor (ER) expression, because GRPR is expressed not only in primary lesions but also in lymph nodes and distant metastasis. In the last decades, several GRPR-targeting molecules have been evaluated both at preclinical and clinical level, however, most of the studies have been focused on prostate cancer (PC). Nonetheless, given the relevance of non-invasive diagnosis and potential treatment of BC through Peptide Receptor Radioligand Therapy (PRRT), this review aims at collecting the available preclinical and clinical data on GRPR-targeting radiopeptides for the imaging and therapy of BC, to better understand the current state-of-the-art and identify future perspectives and possible limitations to their clinical translation. In fact, since luminal-like tumors account for approximately 80% of all BC, many BC patients are likely to benefit from the development of GRPR-radiotheranostics.
Collapse
Affiliation(s)
| | | | | | | | - Eleni Gourni
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
10
|
Verhoeven M, Ruigrok EAM, van Leenders GJLH, van den Brink L, Balcioglu HE, van Weerden WM, Dalm SU. GRPR versus PSMA: expression profiles during prostate cancer progression demonstrate the added value of GRPR-targeting theranostic approaches. Front Oncol 2023; 13:1199432. [PMID: 37719014 PMCID: PMC10502172 DOI: 10.3389/fonc.2023.1199432] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/02/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Central to targeted radionuclide imaging and therapy of prostate cancer (PCa) are prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals. Gastrin-releasing peptide receptor (GRPR) targeting has been proposed as a potential additional approach for PCa theranostics. The aim of this study was to investigate to what extent and at what stage of the disease GRPR-targeting applications can complement PSMA-targeting theranostics in the management of PCa. Methods Binding of the GRPR- and PSMA-targeting radiopharmaceuticals [177Lu]Lu-NeoB and [177Lu]Lu-PSMA-617, respectively, was evaluated and compared on tissue sections of 20 benign prostatic hyperplasia (BPH), 16 primary PCa and 17 progressive castration-resistant PCa (CRPC) fresh frozen tissue specimens. Hematoxylin-eosin and alpha-methylacyl-CoA racemase stains were performed to identify regions of prostatic adenocarcinoma and potentially high-grade prostatic intraepithelial neoplasia. For a subset of primary PCa samples, RNA in situ hybridization (ISH) was used to identify target mRNA expression in defined tumor regions. Results The highest median [177Lu]Lu-NeoB binding was observed in primary PCa samples, while median and overall [177Lu]Lu-PSMA-617 binding was highest in CRPC samples. The highest [177Lu]Lu-NeoB binding was observed in 3/17 CRPC samples of which one sample showed no [177Lu]Lu-PSMA-617 binding. RNA ISH analyses showed a trend between mRNA expression and radiopharmaceutical binding, and confirmed the distinct GRPR and PSMA expression patterns in primary PCa observed with radiopharmaceutical binding. Conclusion Our study emphasizes that GRPR-targeting approaches can contribute to improved PCa management and complement currently applied PSMA-targeting strategies in both early and late stage PCa.
Collapse
Affiliation(s)
- Marjolein Verhoeven
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Eline A. M. Ruigrok
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Geert J. L. H. van Leenders
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lilian van den Brink
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hayri E. Balcioglu
- Department of Medical Oncology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wytske M. van Weerden
- Department of Experimental Urology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Simone U. Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
11
|
Günther T, Deiser S, Felber V, Beck R, Wester HJ. Substitution of l-Tryptophan by α-Methyl-l-Tryptophan in 177Lu-RM2 Results in 177Lu-AMTG, a High-Affinity Gastrin-Releasing Peptide Receptor Ligand with Improved In Vivo Stability. J Nucl Med 2022; 63:1364-1370. [PMID: 35027371 PMCID: PMC9454457 DOI: 10.2967/jnumed.121.263323] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/28/2021] [Indexed: 01/26/2023] Open
Abstract
Theranostic applications targeting the gastrin-releasing peptide receptor (GRPR) have shown promising results. When compared with other peptide ligands for radioligand therapy, the most often used GRPR ligand, DOTA-Pip5-d-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Sta13-Leu14-NH2 (RM2), may be clinically impacted by limited metabolic stability. With the aim of improving the metabolic stability of RM2, we investigated whether the metabolically unstable Gln7-Trp8 bond within the pharmacophore of RM2 can be stabilized via substitution of l-Trp8 by α-methyl-l-tryptophan (α-Me-l-Trp) and whether the corresponding DOTAGA analog might also be advantageous. A comparative preclinical evaluation of 177Lu-α-Me-l-Trp8-RM2 (177Lu-AMTG) and its DOTAGA counterpart (177Lu-AMTG2) was performed using 177Lu-RM2 and 177Lu-NeoBOMB1 as reference compounds. Methods: Peptides were synthesized by solid-phase peptide synthesis and labeled with 177Lu. Lipophilicity was determined at pH 7.4 (logD 7.4). Receptor-mediated internalization was investigated on PC-3 cells (37°C, 60 min), whereas GRPR affinity (half-maximal inhibitory concentration) was determined on both PC-3 and T-47D cells. Stability toward peptidases was examined in vitro (human plasma, 37°C, 72 ± 2 h) and in vivo (murine plasma, 30 min after injection). Biodistribution studies were performed at 24 h after injection, and small-animal SPECT/CT was performed on PC-3 tumor-bearing mice at 1, 4, 8, 24, and 28 h after injection. Results: Solid-phase peptide synthesis yielded 9%-15% purified labeling precursors. 177Lu labeling proceeded quantitatively. Compared with 177Lu-RM2, 177Lu-AMTG showed slightly improved GRPR affinity, a similar low internalization rate, slightly increased lipophilicity, and considerably improved stability in vitro and in vivo. In vivo, 177Lu-AMTG exhibited the highest tumor retention (11.45 ± 0.43 percentage injected dose/g) and tumor-to-blood ratio (2,702 ± 321) at 24 h after injection, as well as a favorable biodistribution profile. As demonstrated by small-animal SPECT/CT imaging, 177Lu-AMTG also revealed a less rapid clearance from tumor tissue. Compared with 177Lu-AMTG, 177Lu-AMTG2 did not show any further benefits. Conclusion: The results of this study, particularly the superior metabolic stability of 177Lu-AMTG, strongly recommend a clinical evaluation of this novel GRPR-targeted ligand to investigate its potential for radioligand therapy of GRPR-expressing malignancies.
Collapse
Affiliation(s)
- Thomas Günther
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Sandra Deiser
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Veronika Felber
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Roswitha Beck
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| |
Collapse
|
12
|
Kurth J, Potratz M, Heuschkel M, Krause BJ, Schwarzenböck SM. GRPr Theranostics: Current Status of Imaging and Therapy using GRPr Targeting Radiopharmaceuticals. Nuklearmedizin 2022; 61:247-261. [PMID: 35668669 DOI: 10.1055/a-1759-4189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Addressing molecular targets, that are overexpressed by various tumor entities, using radiolabeled molecules for a combined diagnostic and therapeutic (theranostic) approach is of increasing interest in oncology. The gastrin-releasing peptide receptor (GRPr), which is part of the bombesin family, has shown to be overexpressed in a variety of tumors, therefore, serving as a promising target for those theranostic applications. A large amount of differently radiolabeled bombesin derivatives addressing the GRPr have been evaluated in the preclinical as well as clinical setting showing fast blood clearance and urinary excretion with selective GRPr-binding. Most of the available studies on GRPr-targeted imaging and therapy have evaluated the theranostic approach in prostate and breast cancer applying bombesin derivatives tagged with the predominantly used theranostic pair of 68Ga/177Lu which is the focus of this review.
Collapse
Affiliation(s)
- Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Madlin Potratz
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Martin Heuschkel
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - Bernd J Krause
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | | |
Collapse
|
13
|
Nguyen A, Fullard K, Sheehan-Dare G, Tang R, Chan L, Ho B, Dear R, Keane J, Hickey A, Nandurkar R, Chen J, Chen A, Lim E, Emmett L. Diagnostic value of 68 Ga-DOTATATE PET-CT imaging for staging of ER + /PR + HER2- breast cancer patients with metastatic disease: Comparison with conventional imaging with bone scan, diagnostic CT and 18 F-FDG PET-CT in a prospective pilot trial. J Med Imaging Radiat Oncol 2021; 66:731-737. [PMID: 34676675 DOI: 10.1111/1754-9485.13342] [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: 12/30/2020] [Accepted: 08/22/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION 18 F-Fludeoxyglucose PET-CT (FDG) is increasingly used to stage breast cancer. Most breast cancers express the Oestrogen Receptor (ER) and Progesterone Receptor (PR), and this subtype demonstrates lower activity on FDG imaging. Somatostatin receptors (SSTR) offer a potentially improved radiotracer target for ER+ /PR+ breast cancer. We present the first in␣vivo clinical study comparing 68 Ga-DOTATATE PET-CT (DOTA) to FDG and conventional imaging (bone scan and diagnostic CT), in metastatic ER+ /PR+ human epidermal growth factor receptor 2 (HER2) negative breast cancer. METHODS Patients with clinically progressive metastatic ER+ /PR+ HER2- breast cancer underwent restaging with DOTA, FDG and conventional imaging. Scans were analysed visually, and semi-quantitatively. Wilcoxon-Rank Scoring was used to assess significance. RESULTS Ten women (mean age 57 years) underwent imaging. 8/10 demonstrated disease on both DOTA and FDG. 2/10 positive on conventional imaging, but DOTA- /FDG- , and had no disease progression at 1-year follow-up. Heterogeneity of uptake was seen between DOTA and FDG with 5 bone lesions DOTA+ /FDG- and 1 bone lesion FDG+ /DOTA- . Twenty-one visceral lesions were FDG+ /DOTA- (2 patients), with 10/21 identified on conventional imaging. Maximum standard uptake values (SUV max) of DOTA were greater than FDG (10.9 vs. 6.6, P = ns). Four sites were biopsied (3 patients). 3/4 had high ER/PR expression (mean DOTA SUV max 9.4) and 1/4 low ER/PR expression (DOTA SUV max 3.1). CONCLUSION Whilst we have not demonstrated DOTA to be superior to FDG in staging of ER+ /PR+ breast cancers, DOTA may have a role in assessing HR status and treatment decisions; further evaluation of this is warranted.
Collapse
Affiliation(s)
- Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia
| | - Karen Fullard
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Gemma Sheehan-Dare
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Reuben Tang
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Lyn Chan
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia
| | - Bao Ho
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia
| | - Rachel Dear
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Joanne Keane
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Adam Hickey
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Rohan Nandurkar
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Julia Chen
- St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Andrew Chen
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Elgene Lim
- St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia.,St Vincent's Clinical School, UNSW, Sydney, New South Wales, Australia.,Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Masab M, Gross A, Flanagan M, Goldberg R, Malla M. Neuroendocrine Neoplasm of the Breast Presenting as a Liver Metastasis: A Rare Diagnostic Challenge. Cureus 2021; 13:e16860. [PMID: 34513437 PMCID: PMC8413109 DOI: 10.7759/cureus.16860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 11/05/2022] Open
Abstract
We present a case of a 58-year-old female who presented initially to an outside institution with abdominal pain and was diagnosed on liver biopsy with a well-differentiated neuroendocrine tumor of an unknown primary source. She was referred to our academic institution for a second opinion after disease progression on the initial chemotherapy regimen. Through additional evaluation, diagnostics, and multi-disciplinary tumor board discussion she was diagnosed with metastases from a well-differentiated neuroendocrine neoplasm of the breast (NENB). Consequently, her treatment plan was modified leading to significant clinical and radiological improvement.
Collapse
Affiliation(s)
- Muhammad Masab
- Hematology and Medical Oncology, West Virginia University, Morgantown, USA
| | - Alexander Gross
- Pathology and Laboratory Medicine, West Virginia University, Morgantown, USA
| | - Melina Flanagan
- Pathology and Laboratory Medicine, West Virginia University, Morgantown, USA
| | - Richard Goldberg
- Hematology and Medical Oncology, West Virginia University, Morgantown, USA
| | - Midhun Malla
- Hematology and Medical Oncology, West Virginia University, Morgantown, USA
| |
Collapse
|
15
|
Chastel A, Vimont D, Claverol S, Zerna M, Bodin S, Berndt M, Chaignepain S, Hindié E, Morgat C. 68Ga-Radiolabeling and Pharmacological Characterization of a Kit-Based Formulation of the Gastrin-Releasing Peptide Receptor (GRP-R) Antagonist RM2 for Convenient Preparation of [ 68Ga]Ga-RM2. Pharmaceutics 2021; 13:pharmaceutics13081160. [PMID: 34452121 PMCID: PMC8398231 DOI: 10.3390/pharmaceutics13081160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022] Open
Abstract
Background: [68Ga]Ga-RM2 is a potent Gastrin-Releasing Peptide-receptor (GRP-R) antagonist for imaging prostate cancer and breast cancer, currently under clinical evaluation in several specialized centers around the world. Targeted radionuclide therapy of GRP-R-expressing tumors is also being investigated. We here report the characteristics of a kit-based formulation of RM2 that should ease the development of GRP-R imaging and make it available to more institutions and patients. Methods: Stability of the investigated kits over one year was determined using LC/MS/MS and UV-HPLC. Direct 68Ga-radiolabeling was optimized with respect to buffer (pH), temperature, reaction time and shaking time. Conventionally prepared [68Ga]Ga-RM2 using an automated synthesizer was used as a comparator. Finally, the [68Ga]Ga-RM2 product was assessed with regards to hydrophilicity, affinity, internalization, membrane bound fraction, calcium mobilization assay and efflux, which is a valuable addition to the in vivo literature. Results: The kit-based formulation, kept between 2 °C and 8 °C, was stable for over one year. Using acetate buffer pH 3.0 in 2.5–5.1 mL total volume, heating at 100 °C during 10 min and cooling down for 5 min, the [68Ga]Ga-RM2 produced by kit complies with the requirements of the European Pharmacopoeia. Compared with the module production route, the [68Ga]Ga-RM2 produced by kit was faster, displayed higher yields, higher volumetric activity and was devoid of ethanol. In in vitro evaluations, the [68Ga]Ga-RM2 displayed sub-nanomolar affinity (Kd = 0.25 ± 0.19 nM), receptor specific and time dependent membrane-bound fraction of 42.0 ± 5.1% at 60 min and GRP-R mediated internalization of 24.4 ± 4.3% at 30 min. The [natGa]Ga-RM2 was ineffective in stimulating intracellular calcium mobilization. Finally, the efflux of the internalized activity was 64.3 ± 6.5% at 5 min. Conclusion: The kit-based formulation of RM2 is suitable to disseminate GRP-R imaging and therapy to distant hospitals without complex radiochemistry equipment.
Collapse
Affiliation(s)
- Adrien Chastel
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, F-33000 Bordeaux, France; (A.C.); (D.V.); (S.B.); (E.H.)
- Nuclear Medicine Department, University Hospital of Bordeaux, F-33000 Bordeaux, France
| | - Delphine Vimont
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, F-33000 Bordeaux, France; (A.C.); (D.V.); (S.B.); (E.H.)
| | - Stephane Claverol
- Proteome Platform, University Bordeaux, F-33000 Bordeaux, France; (S.C.); (S.C.)
| | - Marion Zerna
- Life Molecular Imaging (Formely Piramal Imaging) GmbH, 13353 Berlin, Germany; (M.Z.); (M.B.)
| | - Sacha Bodin
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, F-33000 Bordeaux, France; (A.C.); (D.V.); (S.B.); (E.H.)
- Nuclear Medicine Department, University Hospital of Bordeaux, F-33000 Bordeaux, France
| | - Mathias Berndt
- Life Molecular Imaging (Formely Piramal Imaging) GmbH, 13353 Berlin, Germany; (M.Z.); (M.B.)
| | - Stéphane Chaignepain
- Proteome Platform, University Bordeaux, F-33000 Bordeaux, France; (S.C.); (S.C.)
| | - Elif Hindié
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, F-33000 Bordeaux, France; (A.C.); (D.V.); (S.B.); (E.H.)
- Nuclear Medicine Department, University Hospital of Bordeaux, F-33000 Bordeaux, France
| | - Clément Morgat
- INCIA, University of Bordeaux, CNRS, EPHE, UMR 5287, F-33000 Bordeaux, France; (A.C.); (D.V.); (S.B.); (E.H.)
- Nuclear Medicine Department, University Hospital of Bordeaux, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
16
|
Damiana TST, Dalm SU. Combination Therapy, a Promising Approach to Enhance the Efficacy of Radionuclide and Targeted Radionuclide Therapy of Prostate and Breast Cancer. Pharmaceutics 2021; 13:pharmaceutics13050674. [PMID: 34067215 PMCID: PMC8151894 DOI: 10.3390/pharmaceutics13050674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
In recent years, radionuclide therapy (RT) and targeted radionuclide therapy (TRT) have gained great interest in cancer treatment. This is due to promising results obtained in both preclinical and clinical studies. However, a complete response is achieved in only a small percentage of patients that receive RT or TRT. As a consequence, there have been several strategies to improve RT and TRT outcomes including the combination of these treatments with other well-established anti-cancer therapies, for example, chemotherapy. Combinations of RT and TRT with other therapies with distinct mechanisms of action represent a promising strategy. As for prostate cancer and breast cancer, the two most prevalent cancer types worldwide, several combination-based therapies have been evaluated. In this review, we will provide an overview of the RT and TRT agents currently used or being investigated in combination with hormone therapy, chemotherapy, immunotherapy, and external beam radiation therapy for the treatment of prostate cancer and breast cancer.
Collapse
|
17
|
Meester EJ, de Blois E, Krenning BJ, van der Steen AFW, Norenberg JP, van Gaalen K, Bernsen MR, de Jong M, van der Heiden K. Autoradiographical assessment of inflammation-targeting radioligands for atherosclerosis imaging: potential for plaque phenotype identification. EJNMMI Res 2021; 11:27. [PMID: 33730311 PMCID: PMC7969682 DOI: 10.1186/s13550-021-00772-z] [Citation(s) in RCA: 5] [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: 12/27/2020] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Many radioligands have been developed for the visualization of atherosclerosis by targeting inflammation. However, interpretation of in vivo signals is often limited to plaque identification. We evaluated binding of some promising radioligands in an in vitro approach in atherosclerotic plaques with different phenotypes. METHODS Tissue sections of carotid endarterectomy tissue were characterized as early plaque, fibro-calcific plaque, or phenotypically vulnerable plaque. In vitro binding assays for the radioligands [111In]In-DOTATATE; [111In]In-DOTA-JR11; [67Ga]Ga-Pentixafor; [111In]In-DANBIRT; and [111In]In-EC0800 were conducted, the expression of the radioligand targets was assessed via immunohistochemistry. Radioligand binding and expression of radioligand targets was investigated and compared. RESULTS In sections characterized as vulnerable plaque, binding was highest for [111In]In-EC0800; followed by [111In]In-DANBIRT; [67Ga]Ga-Pentixafor; [111In]In-DOTA-JR11; and [111In]In-DOTATATE (0.064 ± 0.036; 0.052 ± 0.029; 0.011 ± 0.003; 0.0066 ± 0.0021; 0.00064 ± 0.00014 %Added activity/mm2, respectively). Binding of [111In]In-DANBIRT and [111In]In-EC0800 was highest across plaque phenotypes, binding of [111In]In-DOTA-JR11 and [67Ga]Ga-Pentixafor differed most between plaque phenotypes. Binding of [111In]In-DOTATATE was the lowest across plaque phenotypes. The areas positive for cells expressing the radioligand's target differed between plaque phenotypes for all targets, with lowest percentage area of expression in early plaque sections and highest in phenotypically vulnerable plaque sections. CONCLUSIONS Radioligands targeting inflammatory cell markers showed different levels of binding in atherosclerotic plaques and among plaque phenotypes. Different radioligands might be used for plaque detection and discerning early from vulnerable plaque. [111In]In-EC0800 and [111In]In-DANBIRT appear most suitable for plaque detection, while [67Ga]Ga-Pentixafor and [111In]In-DOTA-JR11 might be best suited for differentiation between plaque phenotypes.
Collapse
Affiliation(s)
- Eric J Meester
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Antonius F W van der Steen
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Jeff P Norenberg
- Radiopharmaceutical Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Kim van Gaalen
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Monique R Bernsen
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
18
|
Fu H, Du B, Chen Z, Li Y. Radiolabeled Peptides for SPECT and PET Imaging in the Detection of Breast Cancer: Preclinical and Clinical Perspectives. Curr Med Chem 2021; 27:6987-7002. [PMID: 32003658 DOI: 10.2174/0929867327666200128110827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer in women worldwide. Due to the heterogeneous nature of breast cancer, the optimal treatment and expected response for each patient may not necessarily be universal. Molecular imaging techniques could play an important role in the early detection and targeted therapy evaluation of breast cancer. This review focuses on the development of peptides labeled with SPECT and PET radionuclides for breast cancer imaging. We summarized the current status of radiolabeled peptides for different receptors in breast cancer. The characteristics of radionuclides and major techniques for peptide labeling are also briefly discussed.
Collapse
Affiliation(s)
- Hao Fu
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Bulin Du
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zijun Chen
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| |
Collapse
|
19
|
Baratto L, Duan H, Mäcke H, Iagaru A. Imaging the Distribution of Gastrin-Releasing Peptide Receptors in Cancer. J Nucl Med 2020; 61:792-798. [DOI: 10.2967/jnumed.119.234971] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/06/2020] [Indexed: 01/01/2023] Open
|
20
|
Advanced approaches to imaging primary breast cancer: an update. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
Morgat C, Schollhammer R, Macgrogan G, Barthe N, Vélasco V, Vimont D, Cazeau AL, Fernandez P, Hindié E. Comparison of the binding of the gastrin-releasing peptide receptor (GRP-R) antagonist 68Ga-RM2 and 18F-FDG in breast cancer samples. PLoS One 2019; 14:e0210905. [PMID: 30645633 PMCID: PMC6333408 DOI: 10.1371/journal.pone.0210905] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/03/2019] [Indexed: 12/30/2022] Open
Abstract
The Gastrin-Releasing Peptide Receptor (GRPR) is over-expressed in estrogen receptor (ER) positive breast tumors and related metastatic lymph nodes offering the opportunity of imaging and therapy of luminal tumors. 68Ga-RM2 binding and 18F-FDG binding in tumoral zones were measured and compared using tissue micro-imaging with a beta imager on 14 breast cancer samples (10 primaries and 4 associated metastatic lymph nodes). Results were then assessed against ER expression, progesterone receptor (PR) expression, HER2 over-expression or not and Ki-67 expression. GRPR immunohistochemistry (IHC) was also performed on all samples. We also retrospectively compared 68Ga-RM2 and 18F-FDG bindings to 18F-FDG SUVmax on the pre-therapeutic PET/CT examination, if available. 68Ga-RM2 binding was significantly higher in tumors expressing GRPR on IHC than in GRPR-negative tumors (P = 0.022). In ER+ tumors, binding of 68Ga-RM2 was significantly higher than 18F-FDG (P = 0.015). In tumors with low Ki-67, 68Ga-RM2 binding was also significantly increased compared to 18F-FDG (P = 0.029). Overall, the binding of 68Ga-RM2 and 18F-FDG displayed an opposite pattern in tumor samples and 68Ga-RM2 binding was significantly higher in tumors that had low 18F-FDG binding (P = 0.021). This inverse correlation was also documented in the few patients in whom a 18F-FDG PET/CT examination before surgery was available. Findings from this in vitro study suggest that GRPR targeting can be an alternative to 18F-FDG imaging in ER+ breast tumors. Moreover, because GRPR antagonists can also be labeled with lutetium-177 this opens new avenues for targeted radionuclide therapy in the subset of patients with progressive metastatic disease following conventional treatments.
Collapse
Affiliation(s)
- Clément Morgat
- Nuclear Medicine Department, University Hospital of Bordeaux, Bordeaux, France
- Univ. Bordeaux, INCIA, UMR-CNRS 5287, Talence, France
- CNRS, INCIA, UMR 5287, Talence, France
- * E-mail:
| | - Romain Schollhammer
- Nuclear Medicine Department, University Hospital of Bordeaux, Bordeaux, France
- Univ. Bordeaux, INCIA, UMR-CNRS 5287, Talence, France
- CNRS, INCIA, UMR 5287, Talence, France
| | - Gaétan Macgrogan
- Surgical Pathology Unit, Department of BioPathology, Institut Bergonié, Bordeaux, France
- INSERM, ACTION U1218, Bordeaux, France
| | | | - Valérie Vélasco
- Surgical Pathology Unit, Department of BioPathology, Institut Bergonié, Bordeaux, France
- INSERM, ACTION U1218, Bordeaux, France
| | - Delphine Vimont
- Univ. Bordeaux, INCIA, UMR-CNRS 5287, Talence, France
- CNRS, INCIA, UMR 5287, Talence, France
| | | | - Philippe Fernandez
- Nuclear Medicine Department, University Hospital of Bordeaux, Bordeaux, France
- Univ. Bordeaux, INCIA, UMR-CNRS 5287, Talence, France
- CNRS, INCIA, UMR 5287, Talence, France
| | - Elif Hindié
- Nuclear Medicine Department, University Hospital of Bordeaux, Bordeaux, France
- Univ. Bordeaux, INCIA, UMR-CNRS 5287, Talence, France
- CNRS, INCIA, UMR 5287, Talence, France
| |
Collapse
|
22
|
Liang X, Briaux A, Becette V, Benoist C, Boulai A, Chemlali W, Schnitzler A, Baulande S, Rivera S, Mouret-Reynier MA, Bouvet LV, De La Motte Rouge T, Lemonnier J, Lerebours F, Callens C. Molecular profiling of hormone receptor-positive, HER2-negative breast cancers from patients treated with neoadjuvant endocrine therapy in the CARMINA 02 trial (UCBG-0609). J Hematol Oncol 2018; 11:124. [PMID: 30305115 PMCID: PMC6180434 DOI: 10.1186/s13045-018-0670-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Postmenopausal women with large, hormone receptor (HR)-positive/HER2-negative and low-proliferative breast cancer derived a benefit from neoadjuvant endocrine therapy (NET) in the CARMINA02 trial. This study was designed to correlate gene expression and mutation profiles with both response to NET and prognosis. METHODS Gene expression profiling using RNA sequencing was performed in 86 pre-NET and post-NET tumor samples. Targeted next-generation sequencing of 91 candidate breast cancer-associated genes was performed on DNA samples from 89 patients. Molecular data were correlated with radiological response and relapse-free survival. RESULTS The transcriptional profile of tumors to NET in responders involved immune-associated genes enriched in activated Th1 pathway, which remained unchanged in non-responders. Immune response was confirmed by analysis of tumor-infiltrating lymphocytes (TILs). The percentage of TILs was significantly increased post-NET compared to pre-NET samples in responders (p = 0.0071), but not in non-responders (p = 0.0938). Gene expression revealed that lipid metabolism was the main molecular function related to prognosis, while PPARγ is the most important upstream regulator gene. The most frequently mutated genes were PIK3CA (48.3%), CDH1 (20.2%), PTEN (15.7%), TP53 (10.1%), LAMA2 (10.1%), BRCA2 (9.0%), MAP3K1 (7.9%), ALK (6.7%), INPP4B (6.7%), NCOR1 (6.7%), and NF1 (5.6%). Cell cycle and apoptosis pathway and PIK3CA/AKT/mTOR pathway were altered significantly more frequently in non-responders than in responders (p = 0.0017 and p = 0.0094, respectively). The average number of mutations per sample was significantly higher in endocrine-resistant tumors (2.88 vs. 1.64, p = 0.03), but no difference was observed in terms of prognosis. ESR1 hotspot mutations were detected in 3.4% of treatment-naive tumors. CONCLUSIONS The Th1-related immune system and lipid metabolism appear to play key roles in the response to endocrine therapy and prognosis in HR-positive/HER2-negative breast cancer. Deleterious somatic mutations in the cell cycle and apoptosis pathway and PIK3CA/AKT/mTOR pathway may be relevant for clinical management. TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov ( NCT00629616 ) on March 6, 2008, retrospectively registered.
Collapse
Affiliation(s)
- Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, China.,Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Adrien Briaux
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Véronique Becette
- Department of Biopathology, Curie Institute, René Huguenin Hospital, Saint-Cloud, France
| | - Camille Benoist
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Anais Boulai
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Walid Chemlali
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Anne Schnitzler
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Curie Institute, PSL Research University, Paris, France
| | - Sofia Rivera
- Department of Radiotherapy, Gustave Roussy, Villejuif, France
| | | | | | | | | | - Florence Lerebours
- Department of Medical Oncology, Curie Institute, René Huguenin Hospital, Saint-Cloud, France
| | - Céline Callens
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France.
| |
Collapse
|
23
|
Zang J, Mao F, Wang H, Zhang J, Liu Q, Peng L, Li F, Lang L, Chen X, Zhu Z. 68Ga-NOTA-RM26 PET/CT in the Evaluation of Breast Cancer: A Pilot Prospective Study. Clin Nucl Med 2018; 43:663-669. [PMID: 30036253 PMCID: PMC6076351 DOI: 10.1097/rlu.0000000000002209] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND This prospective pilot study investigated the value of Ga-NOTA-RM26, an antagonist targeting gastrin-releasing peptide receptor, in evaluation of breast cancer. METHODS Thirty-five women in suspicion of breast cancer based on mammography or ultrasonography were recruited with informed consent. They underwent PET/CT scans 30 minutes after intravenous injection of Ga-NOTA-RM26 in a dose of 1.85 MBq (0.05 mCi) per kilogram body weight within 1 week before surgery. The Ga-NOTA-RM26 uptake was correlated with the pathological and immunohistochemical findings. RESULTS Ga-NOTA-RM26 positivity was found correlated with estrogen receptor (ER) expression (P = 0.006) and menstrual status (P = 0.019). In 34 patients diagnosed with breast cancer, the SUVmax was found significantly higher in the ER-positive breast cancer (4.97 ± 1.89) than in the ER-negative breast cancer (2.78 ± 0.65, P < 0.001). Ga-NOTA-RM26 was also found accumulated in normal breast tissue, with the SUVmax significantly higher in patients at the secretory phase of menstrual cycle (2.27 ± 0.71) than in those at the nonsecretory phase (1.59 ± 0.49, P = 0.017) and postmenopause (1.43 ± 0.44, P = 0.002). If the secretory phase patients were excluded, the sensitivity, specificity, and accuracy for differentiation of breast cancer from breast tissue increased from 85.3%, 86.8%, and 86.1% to 100%, 90.9%, and 95.5%, respectively. CONCLUSIONS This pilot study indicates that the diagnostic accuracy of Ga-NOTA-RM26 PET/CT in breast cancer may correlate with ER expression and menstrual status of the patient. It may be better to avoid performing this examination during the menstrual secretory phase to reduce physiological uptake in normal breast tissue.
Collapse
Affiliation(s)
- Jie Zang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Feng Mao
- Department of Breast Surgery, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
| | - Hao Wang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Jingjing Zhang
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Qingxing Liu
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Li Peng
- Department of Breast Surgery, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Lixin Lang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Science and PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| |
Collapse
|
24
|
Large set data mining reveals overexpressed GPCRs in prostate and breast cancer: potential for active targeting with engineered anti-cancer nanomedicines. Oncotarget 2018; 9:24882-24897. [PMID: 29861840 PMCID: PMC5982759 DOI: 10.18632/oncotarget.25427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/24/2018] [Indexed: 01/29/2023] Open
Abstract
Over 800 G-protein-coupled receptors (GPCRs) are encoded by the human genome and many are overexpressed in tumors. GPCRs are triggered by ligand molecules outside the cell and activate internal signal transduction pathways driving cellular responses. The receptor signals are desensitized by receptor internalization and this mechanism can be exploited for the specific delivery of ligand-linked drug molecules directly into cells. Detailed expression analysis in cancer tissue can inform the design of GPCR-ligand decorated drug carriers for active tumor cell targeting. The active targeting process utilizes ligand receptor interactions leading to binding and in most cases internalization of the ligand-attached drug carrier resulting in effective targeting of cancer cells. In this report public microarray data from the Gene Expression Omnibus (GEO) repository was used to identify overexpressed GPCRs in prostate and breast cancer tissues. The analyzed data confirmed previously known cancer receptor associations and identified novel candidates for potential active targeting. Prioritization of the identified targeting receptors is also presented based on high expression levels and frequencies in cancer samples but low expression in healthy tissue. Finally, some selected examples were used in ligand docking studies to assess the feasibility for chemical conjugation to drug nanocarriers without interference of receptor binding and activation. The presented data demonstrate a large untapped potential to improve efficacy and safety of current and future anti-cancer compounds through active targeting of GPCRs on cancer cells.
Collapse
|
25
|
Dalm SU, Haeck J, Doeswijk GN, de Blois E, de Jong M, van Deurzen CHM. SSTR-Mediated Imaging in Breast Cancer: Is There a Role for Radiolabeled Somatostatin Receptor Antagonists? J Nucl Med 2017; 58:1609-1614. [PMID: 28450563 DOI: 10.2967/jnumed.116.189035] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/18/2017] [Indexed: 11/16/2022] Open
Abstract
Recent studies have shown enhanced tumor targeting by novel somatostatin receptor (SSTR) antagonists compared with clinically widely used agonists. However, these results have been obtained mostly in neuroendocrine tumors, and only limited data are available for cancer types with lower SSTR expression, including breast cancer (BC). To date, two studies have reported higher binding of the antagonist than the agonist in BC, but in both studies only a limited number of cases were evaluated. In this preclinical study, we further investigated whether the application of an SSTR antagonist can improve SSTR-mediated BC imaging in a large panel of BC specimens. We also generated an in vivo BC mouse model and performed SPECT/MRI and biodistribution studies. Methods: Binding of 111In-DOTA-Tyr3-octreotate (SSTR agonist) and 111In-DOTA-JR11 (SSTR antagonist) to 40 human BC specimens was compared using in vitro autoradiography. SSTR2 immunostaining was performed to confirm SSTR2 expression of the tumor cells. Furthermore, binding of the radiolabeled SSTR agonist and antagonist was analyzed in tissue material from 6 patient-derived xenografts. One patient-derived xenograft, the estrogen receptor-positive model T126, was chosen to generate in vivo mouse models containing orthotopic breast tumors for in vivo SPECT/MRI and biodistribution studies after injection with 177Lu-DOTA-Tyr3-octreotate or 177Lu-DOTA-JR11. Results:111In-DOTA-JR11 binding to human BC tissue was significantly higher than 111In-DOTA-Tyr3-octreotate binding (P < 0.001). The median ratio of antagonist binding versus agonist binding was 3.39 (interquartile range, 2-5). SSTR2 immunostaining confirmed SSTR2 expression on the tumor cells. SPECT/MRI of the mouse model found better tumor visualization with the antagonist. This result was in line with the significantly higher tumor uptake of the radiolabeled antagonist than of the agonist as measured in biodistribution studies 285 min after radiotracer injection (percentage injected dose per gram of tissue: 1.92 ± 0.43 vs. 0.90 ± 0.17; P = 0.002). Conclusion: SSTR antagonists are promising candidates for BC imaging.
Collapse
Affiliation(s)
- Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Joost Haeck
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Gabriela N Doeswijk
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; and
| | | |
Collapse
|
26
|
Dude I, Zhang Z, Rousseau J, Hundal-Jabal N, Colpo N, Merkens H, Lin KS, Bénard F. Evaluation of agonist and antagonist radioligands for somatostatin receptor imaging of breast cancer using positron emission tomography. EJNMMI Radiopharm Chem 2017; 2:4. [PMID: 29503845 PMCID: PMC5824694 DOI: 10.1186/s41181-017-0023-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/28/2017] [Indexed: 01/21/2023] Open
Abstract
Background The somatostatin receptor subtype 2 (sstr2) is expressed on a majority of luminal breast cancers, however SPECT and scintigraphy imaging with agonistic sstr2 probes has been sub-optimal. High affinity antagonists can access more binding sites on the cell surface, resulting in higher tumor uptake and improved sensitivity. We compared the tumor uptake and biodistribution of the antagonist 68Ga-NODAGA-JR11 with two agonists 68Ga-DOTA-Tyr3-octreotide (68Ga-DOTATOC) and 68Ga-DOTA-Tyr3-octreotate (68Ga-DOTATATE), in the human, sstr2-positive, luminal breast cancer model: ZR-75-1. Results Peptides were assayed for binding affinity using a filtration-based competitive assay to sstr2. natGa-DOTATOC and natGa-DOTATATE had excellent affinity (inhibition constant Ki: 0.9 ± 0.1 nM and 1.4 ± 0.3 nM respectively) compared to natGa-NODAGA-JR11 (25.9 ± 0.2 nM). The number of binding sites on ZR-75-1 cells was determined in vitro by saturation assays. Agonist 67/natGa-DOTATOC bound to 6.64 ± 0.39 × 104 sites/cells, which was 1.5-fold higher than 67/natGa-NODAGA-JR11 and 2.3-fold higher than 67/natGa-DOTATATE. All three 68Ga-labeled peptides were obtained in good decay-corrected radiochemical yield (61-68%) and were purified by high performance liquid chromatography to ensure high specific activity (137 – 281 MBq/nmol at the end of synthesis). NOD scid gamma mice bearing ZR-75-1 tumors were injected intravenously with the labeled peptides and used for PET/CT imaging and biodistribution at 1 h post-injection. We found that 68Ga-DOTATOC had the highest tumor uptake (18.4 ± 2.9%ID/g), followed by 68Ga-DOTATATE (15.2 ± 2.2%ID/g) and 68Ga-NODAGA-JR11 (12.2 ± 0.8%ID/g). Tumor-to-blood and tumor-to-muscle ratios were also higher for the agonists (>40 and >150 respectively), compared to the antagonist (15.6 ± 2.2 and 45.2 ± 11.6 respectively). Conclusions The antagonist 68Ga-NODAGA-JR11 had the lowest tumor uptake and contrast compared to agonists 68Ga-DOTATOC and 68Ga-DOTATATE in ZR-75-1 xenografts. The main contributing factor to this result could be the use of an endogenously expressing cell line, which may differ from previously published transfected models in the number of low-affinity, antagonist-specific binding sites. The relative merit of agonists versus antagonists for sstr2 breast cancer imaging warrants further investigation, first in preclinical models with other sstr2-positive breast cancer xenografts, and ultimately in luminal breast cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s41181-017-0023-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iulia Dude
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Zhengxing Zhang
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Julie Rousseau
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Navjit Hundal-Jabal
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Nadine Colpo
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Helen Merkens
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada
| | - Kuo-Shyan Lin
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada.,2Department of Radiology, University of British Columbia, Vancouver, BC Canada
| | - François Bénard
- 1Department of Molecular Oncology, BC Cancer Agency Research Centre, 675 West 10th Ave, Vancouver, V5Z 1 L3 BC Canada.,2Department of Radiology, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
27
|
Morgat C, MacGrogan G, Brouste V, Vélasco V, Sévenet N, Bonnefoi H, Fernandez P, Debled M, Hindié E. Expression of Gastrin-Releasing Peptide Receptor in Breast Cancer and Its Association with Pathologic, Biologic, and Clinical Parameters: A Study of 1,432 Primary Tumors. J Nucl Med 2017; 58:1401-1407. [DOI: 10.2967/jnumed.116.188011] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022] Open
|
28
|
Review: Receptor Targeted Nuclear Imaging of Breast Cancer. Int J Mol Sci 2017; 18:ijms18020260. [PMID: 28134770 PMCID: PMC5343796 DOI: 10.3390/ijms18020260] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/17/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022] Open
Abstract
Receptor targeted nuclear imaging directed against molecular markers overexpressed on breast cancer (BC) cells offers a sensitive and specific method for BC imaging. Currently, a few targets such as estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), somatostatin receptor (SSTR), and the gastrin releasing peptide receptor (GRPR) are being investigated for this purpose. Expression of these targets is BC subtype dependent and information that can be gained from lesion visualization is dependent on the target; ER-targeting radiotracers, e.g., can be used to monitor response to anti-estrogen treatment. Here we give an overview of the studies currently under investigation for receptor targeted nuclear imaging of BC. Main findings of imaging studies are summarized and (potential) purposes of lesion visualization by targeting these molecular markers are discussed. Since BC is a very heterogeneous disease and molecular target expression can vary per subtype, but also during disease progression or under influence of treatment, radiotracers for selected imaging purposes should be chosen carefully.
Collapse
|
29
|
Dalm SU, Schrijver WAME, Sieuwerts AM, Look MP, Ziel - van der Made ACJ, de Weerd V, Martens JW, van Diest PJ, de Jong M, van Deurzen CHM. Prospects of Targeting the Gastrin Releasing Peptide Receptor and Somatostatin Receptor 2 for Nuclear Imaging and Therapy in Metastatic Breast Cancer. PLoS One 2017; 12:e0170536. [PMID: 28107508 PMCID: PMC5249060 DOI: 10.1371/journal.pone.0170536] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/05/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The gastrin releasing peptide receptor (GRPR) and the somatostatin receptor 2 (SSTR2) are overexpressed on primary breast cancer (BC), making them ideal candidates for receptor-mediated nuclear imaging and therapy. The aim of this study was to determine whether these receptors are also suitable targets for metastatic BC. METHODS mRNA expression of human BC samples were studied by in vitro autoradiography and associated with radioligand binding. Next, GRPR and SSTR2 mRNA levels of 60 paired primary BCs and metastases from different sites were measured by quantitative reverse transcriptase polymerase chain reaction. Receptor mRNA expression levels were associated with clinico-pathological factors and expression levels of primary tumors and corresponding metastases were compared. RESULTS Binding of GRPR and SSTR radioligands to tumor tissue correlated significantly with receptor mRNA expression. High GRPR and SSTR2 mRNA levels were associated with estrogen receptor (ESR1)-positive tumors (p<0.001 for both receptors). There was no significant difference in GRPR mRNA expression of primary tumors versus paired metastases. Regarding SSTR2 mRNA expression, there was also no significant difference in the majority of cases, apart from liver and ovarian metastases which showed a significantly lower expression compared to the corresponding primary tumors (p = 0.02 and p = 0.03, respectively). CONCLUSION Targeting the GRPR and SSTR2 for nuclear imaging and/or treatment has the potential to improve BC care in primary as well as metastatic disease.
Collapse
Affiliation(s)
- Simone U. Dalm
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- * E-mail:
| | | | - Anieta M. Sieuwerts
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Maxime P. Look
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | | | - Vanja de Weerd
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - John W. Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Paul J. van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marion de Jong
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
30
|
Dalm SU, Melis M, Emmering J, Kwekkeboom DJ, de Jong M. Breast cancer imaging using radiolabelled somatostatin analogues. Nucl Med Biol 2016; 43:559-565. [DOI: 10.1016/j.nucmedbio.2016.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/12/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
|
31
|
Stoykow C, Erbes T, Maecke HR, Bulla S, Bartholomä M, Mayer S, Drendel V, Bronsert P, Werner M, Gitsch G, Weber WA, Stickeler E, Meyer PT. Gastrin-releasing Peptide Receptor Imaging in Breast Cancer Using the Receptor Antagonist (68)Ga-RM2 And PET. Theranostics 2016; 6:1641-50. [PMID: 27446498 PMCID: PMC4955063 DOI: 10.7150/thno.14958] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/08/2016] [Indexed: 01/07/2023] Open
Abstract
Introduction: The gastrin-releasing peptide receptor (GRPR) is overexpressed in breast cancer. The present study evaluates GRPR imaging as a novel imaging modality in breast cancer by employing positron emission tomography (PET) and the GRPR antagonist 68Ga-RM2. Methods: Fifteen female patients with biopsy confirmed primary breast carcinoma (3 bilateral tumors; median clinical stage IIB) underwent 68Ga-RM2-PET/CT for pretreatment staging. In vivo tumor uptake of 68Ga-RM2 was correlated with estrogen (ER) and progesterone (PR) receptor expression, HER2/neu status and MIB-1 proliferation index in breast core biopsy specimens. Results: 13/18 tumors demonstrated strongly increased 68Ga-RM2 uptake compared to normal breast tissue (defined as PET-positive). All PET-positive primary tumors were ER- and PR-positive (13/13) in contrast to only 1/5 PET-negative tumors. Mean SUVMAX of ER-positive tumors was 10.6±6.0 compared to 2.3±1.0 in ER-negative tumors (p=0.016). In a multivariate analysis including ER, PR, HER2/neu and MIB-1, only ER expression predicted 68Ga-RM2 uptake (model: r2=0.55, p=0.025). Normal breast tissue showed inter- and intraindividually variable, moderate GRPR binding (SUVMAX 2.3±1.0), while physiological uptake of other organs was considerably less except pancreas. Of note, 68Ga-RM2-PET/CT detected internal mammary lymph nodes with high 68Ga-RM2 uptake (n=8), a contralateral axillary lymph node metastasis (verified by biopsy) and bone metastases (n=1; not detected by bone scan and CT). Conclusion: Our study demonstrates that 68Ga-RM2-PET/CT is a promising imaging method in ER-positive breast cancer. In vivo GRPR binding assessed by 68Ga-RM2-PET/CT correlated with ER expression in primary tumors of untreated patients.
Collapse
|
32
|
Affiliation(s)
- Fan Pu
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA.,Inlighta Biosciences LLC, Marietta, GA 30068, USA
| | - Shenghui Xue
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
33
|
Liu Y, An S, Ward R, Yang Y, Guo XX, Li W, Xu TR. G protein-coupled receptors as promising cancer targets. Cancer Lett 2016; 376:226-39. [PMID: 27000991 DOI: 10.1016/j.canlet.2016.03.031] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
G protein-coupled receptors (GPCRs) regulate an array of fundamental biological processes, such as growth, metabolism and homeostasis. Specifically, GPCRs are involved in cancer initiation and progression. However, compared with the involvement of the epidermal growth factor receptor in cancer, that of GPCRs have been largely ignored. Recent findings have implicated many GPCRs in tumorigenesis, tumor progression, invasion and metastasis. Moreover, GPCRs contribute to the establishment and maintenance of a microenvironment which is permissive for tumor formation and growth, including effects upon surrounding blood vessels, signaling molecules and the extracellular matrix. Thus, GPCRs are considered to be among the most useful drug targets against many solid cancers. Development of selective ligands targeting GPCRs may provide novel and effective treatment strategies against cancer and some anticancer compounds are now in clinical trials. Here, we focus on tumor related GPCRs, such as G protein-coupled receptor 30, the lysophosphatidic acid receptor, angiotensin receptors 1 and 2, the sphingosine 1-phosphate receptors and gastrin releasing peptide receptor. We also summarize their tissue distributions, activation and roles in tumorigenesis and discuss the potential use of GPCR agonists and antagonists in cancer therapy.
Collapse
Affiliation(s)
- Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Richard Ward
- Molecular Pharmacology Group, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, Scotland, United Kingdom
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Wei Li
- Kidney Cancer Research, Diagnosis and Translational Technology Center of Yunnan Province, Department of Urology, The People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China.
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|