1
|
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
|
2
|
Arslan E, Baloğlu MC, Özkan İA, Alçin G, Akgün E, Çermik TF. 18 F-FDG PET/CT Positive and 68 Ga-DOTA-Bombesin PET/CT Negative Focus of Benign Apocrine Metaplasia Mimicking Malignancy. Clin Nucl Med 2025; 50:e300-e302. [PMID: 40025668 DOI: 10.1097/rlu.0000000000005709] [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: 09/24/2024] [Accepted: 12/17/2024] [Indexed: 03/04/2025]
Abstract
Gastrin-releasing peptide receptor (GRPR) is a promising agent for imaging and development of theranostic radioligands in estrogen receptor (ER)-positive luminal type breast cancer (BC) and may show expression not only in primary malignant lesions but also in lymph node metastases and distant organ metastases. We would like to present a lesion diagnosed as benign apocrine metaplasia in a 45-year-old woman diagnosed with BC, which was negative in 68 Ga-DOTA-Bombesin PET-CT and false positive in 18 F-FDG PET-CT imaging, within the scope of the ongoing study.
Collapse
Affiliation(s)
- Esra Arslan
- Istanbul Training and Research Hospital, Clinic of Nuclear Medicine, University of Health Sciences
| | - Mehmet Can Baloğlu
- Istanbul Training and Research Hospital, Clinic of Nuclear Medicine, University of Health Sciences
| | - İrem Aylin Özkan
- Department of Pathology, Istanbul Training and Research Hospital, University of Health Sciences
| | - Göksel Alçin
- Istanbul Training and Research Hospital, Clinic of Nuclear Medicine, University of Health Sciences
| | - Elife Akgün
- Istanbul Training and Research Hospital, Clinic of Nuclear Medicine, University of Health Sciences
| | | |
Collapse
|
3
|
Plichta KA, Buatti JM. The Emerging Potential of Lead-212 Theranostics. Hematol Oncol Clin North Am 2025; 39:221-236. [PMID: 39827042 DOI: 10.1016/j.hoc.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The field of theranostics uses radiopharmaceuticals to diagnose and treat disease, allowing for a personalized approach to treatment. Most theranostic therapies involve the use of beta-emitting radiopharmaceuticals. Because of their higher energies and decreased range, the use of alpha-emitting radiopharmaceuticals offers potential advantages over beta-emitting radiopharmaceuticals, including the potential for improved cell kill and decreased toxicity to normal tissues. This article focuses on the potential use of lead-212 as a theranostic treatment agent.
Collapse
Affiliation(s)
- Kristin A Plichta
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
4
|
Wang L, Chen CC, Chapple D, Wong AAWL, Kurkowska S, Lau WS, Uribe CF, Bénard F, Lin KS. Synthesis and Evaluation of 68Ga- and 177Lu-Labeled [diF-Pro 14]Bombesin(6-14) Analogs for Detection and Radioligand Therapy of Gastrin-Releasing Peptide Receptor-Expressing Cancer. Pharmaceuticals (Basel) 2025; 18:234. [PMID: 40006047 PMCID: PMC11859184 DOI: 10.3390/ph18020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Overexpressed in various solid tumors, the gastrin-releasing peptide receptor (GRPR) is a promising target for cancer diagnosis and therapy. However, the high pancreas uptake of the current clinically evaluated GRPR-targeted radiopharmaceuticals limits their applications. In this study, we replaced the Pro14 residue in our previously reported GRPR-targeted LW02056 and ProBOMB5 with 4,4-difluoroproline (diF-Pro) to obtain an agonist LW02060 (DOTA-Pip-[D-Phe6,Tle10,NMe-His12,diF-Pro14]Bombesin(6-14)) and an antagonist LW02080 (DOTA-Pip-[D-Phe6,NMe-Gly11,Leu13(ψ)diF-Pro14]Bombesin(6-14)), respectively. Methods/Results: The binding affinities (Ki) of Ga-LW02060, Ga-LW02080, Lu-LW02060, and Lu-LW02080 were measured by in vitro competition binding assays using PC-3 cells and were found to be 5.57 ± 2.47, 21.7 ± 6.69, 8.00 ± 2.61, and 32.1 ± 8.14 nM, respectively. The 68Ga- and 177Lu-labeled ligands were obtained in 36-75% decay-corrected radiochemical yields with >95% radiochemical purity. PET imaging, SPECT imaging, and ex vivo biodistribution studies were conducted in PC-3 tumor-bearing mice. Both [68Ga]Ga-LW02060 and [68Ga]Ga-LW02080 enabled clear tumor visualization in PET images at 1 h post-injection (pi). Tumor uptake values of [68Ga]Ga-LW02060 and [68Ga]Ga-LW02080 at 1 h pi were 16.8 ± 2.70 and 7.36 ± 1.33 %ID/g, respectively, while their pancreas uptake values were 3.12 ± 0.89 and 0.38 ± 0.04 %ID/g, respectively. Compared to [177Lu]Lu-LW02080, [177Lu]Lu-LW02060 showed higher tumor uptake at all time points (1, 4, 24, 72, and 120 h pi). However, fast tumor clearance was observed for both [177Lu]Lu-LW02060 and [177Lu]Lu-LW02080. Conclusions: Our data demonstrate that [68Ga]Ga-LW02060 is promising for clinical translation for the detection of GRPR-expressing tumor lesions. However, further optimizations are needed for [177Lu]Lu-LW02060 and [177Lu]Lu-LW02080 to prolong tumor retention for therapeutic applications.
Collapse
Affiliation(s)
- Lei Wang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
| | - Chao-Cheng Chen
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
| | - Devon Chapple
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
| | - Antonio A. W. L. Wong
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
| | - Sara Kurkowska
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.K.); (C.F.U.)
- Department of Nuclear Medicine, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Wing Sum Lau
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
| | - Carlos F. Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.K.); (C.F.U.)
- Department of Molecular Imaging and Therapy, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
- Department of Molecular Imaging and Therapy, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (L.W.); (C.-C.C.); (D.C.); (A.A.W.L.W.); (W.S.L.); (F.B.)
- Department of Molecular Imaging and Therapy, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
5
|
Kim DG, Choi EY, Ahn HM, Kim YJ. GRPR Drives Metastasis via CRABP2 and FNDC4 Pathways in Lung Adenocarcinoma. Cells 2024; 13:2128. [PMID: 39768218 PMCID: PMC11674891 DOI: 10.3390/cells13242128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Metastasis is a leading cause of lung adenocarcinoma (LUAD)-related mortality and presents significant challenges for treatment. The gastrin-releasing peptide receptor (GRPR), a member of the G protein-coupled receptor (GPCR) family, has an unclear role in LUAD progression. This study aimed to investigate the function and underlying mechanisms of GRPR in LUAD metastasis. Our findings revealed that GRPR levels were significantly elevated in tumor tissues, and higher GRPR expression was associated with worse overall survival outcomes. Functional assays demonstrated that GRPR overexpression enhanced LUAD cell invasion, while GRPR knockdown inhibited invasion both in vitro and in vivo. RNA sequencing and gene set enrichment analysis (GSEA) identified an enrichment of metastasis-promoting genes in GRPR-overexpressing cells, with CRABP2 and FNDC4 emerging as key targets. Clinical analyses further confirmed a positive correlation between GRPR expression and the levels of CRABP2 and FNDC4 in LUAD patients. These results suggest that GRPR could serve as both a prognostic marker and a therapeutic target to inhibit metastasis in LUAD.
Collapse
Affiliation(s)
| | | | | | - Youn-Jae Kim
- Targeted Therapy Branch, Division of Rare and Refractory Cancer, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea; (D.-G.K.); (E.-Y.C.); (H.-M.A.)
| |
Collapse
|
6
|
Saidi A, Stallons TA, Wong AG, Torgue JJ. Preclinical Investigation of [ 212Pb]Pb-DOTAM-GRPR1 for Peptide Receptor Radionuclide Therapy in a Prostate Tumor Model. J Nucl Med 2024; 65:1769-1775. [PMID: 39327021 PMCID: PMC11533912 DOI: 10.2967/jnumed.124.268101] [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: 05/30/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The role of gastrin-releasing peptide receptor (GRPR) in various diseases, including cancer, has been extensively studied and has emerged as a promising therapeutic target. In this study, we successfully achieved the use of [212Pb]Pb-DOTAM-GRPR1, comprising the α-particle generator, 212Pb, combined with a GRPR-targeting peptide, GRPR1, in a prostate cancer model. Methods: Pharmacokinetics, toxicity, radiation dosimetry, and efficacy were assessed in GRPR-positive prostate tumor-bearing mice after intravenous administration of [212Pb]Pb-DOTAM-GRPR1 (where DOTAM is 1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane). Results: Preclinical studies have shown tumor targeting of up to 5 percent injected dose per gram over 24 h, and optimization of the drug formulation and quantity has led to minimized oxidation and off-target binding, respectively. Particularly, an increase in peptide amount from 28 to 280 ng was shown to reduce off-target uptake, especially at the level of the pancreas, by about 30%. Furthermore, dosimetry studies confirmed the kidney as the dose-limiting organ, and toxicity studies revealed that a nontoxic dose of up to 1,665 kBq could be injected into mice. Efficacy studies indicated a median survival time of 9 wk in the control group, which received only a buffer solution, compared with 19 wk in the group that received 4 injections of 370 kBq at 3-wk intervals. Conclusion: Taken together, these combined data demonstrate the safety, tolerability, and efficacy of [212Pb]Pb-DOTAM-GRPR1, thus warranting further exploration in clinical trials.
Collapse
|
7
|
Dalm S, Duan H, Iagaru A. Gastrin Releasing Peptide Receptors-targeted PET Diagnostics and Radionuclide Therapy for Prostate Cancer Management: Preclinical and Clinical Developments of the Past 5 Years. PET Clin 2024; 19:401-415. [PMID: 38644111 DOI: 10.1016/j.cpet.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Each tumor has its own distinctive molecular identity. Treatment, therefore, should be tailored to this unique cancer phenotype. Theragnostics uses the same compound for targeted imaging and treatment, radiolabeled to an appropriate radionuclide, respectively. Gastrin-releasing peptide receptors (GRPRs) are overexpressed in prostate cancer, and radiolabeled GRPR antagonists have shown high diagnostic performance at staging and biochemical recurrence. Several GRPR-targeting theragnostic compounds have been developed preclinically. Their translation into clinics is underway with 4 clinical trials recruiting participants with GRPR-expressing tumors.
Collapse
Affiliation(s)
- Simone Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, Rotterdam 3015 GD, The Netherlands
| | - Heying Duan
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA 94305, USA
| | - Andrei Iagaru
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Wen X, Wang R, Xu P, Shi M, Shang Q, Zeng X, Zeng X, Liu J, Wang X, Zhu Z, Guo Z, Chen X, Zhang J. Synthesis, preclinical, and initial clinical evaluation of integrin α Vβ 3 and gastrin-releasing peptide receptor (GRPR) dual-targeting radiotracer [ 68Ga]Ga-RGD-RM26-03. Eur J Nucl Med Mol Imaging 2024; 51:2023-2035. [PMID: 38376806 DOI: 10.1007/s00259-024-06634-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024]
Abstract
UNLABELLED Integrin receptor αvβ3 and gastrin-releasing peptide receptor (GRPR) expression of tumors could be detected using PET imaging with radiolabeled Arg-Gly-Asp (RGD) and the antagonistic bombesin analog RM26, respectively. The purpose of this study was to investigate the dual receptor-targeting property of the heterodimer RGD-RM26-03 (denoted as LNC1015), demonstrate the tumor diagnostic value of [68Ga]Ga-LNC1015 in preclinical experiments, and evaluate its preliminary clinical feasibility. METHODS LNC1015 was designed and synthesized by linking cyclic RGD and the RM26 peptide. Preclinical pharmacokinetics were detected in a PC3 xenograft model using microPET and biodistribution studies. The clinical feasibility of [68Ga]Ga-LNC1015 PET/CT was performed in patients with breast cancer, and the results were compared with those of 18F-fluorodeoxyglucose (FDG). RESULTS [68Ga]Ga-LNC1015 had good stability in saline for at least 2 h, and favorable binding affinity and specificity were demonstrated in vitro and in vivo. The tumor uptake and retention of [68Ga]Ga-LNC1015 during PET imaging were improved compared with its monomeric counterparts [68Ga]Ga-RGD and [68Ga]Ga-RM26 at all the time points examined. In our initial clinical studies, the tumor uptake and tumor-to-background ratio (TBR) of primary and metastatic lesions in [68Ga]Ga-LNC1015 PET/CT were significantly higher than those in [18F]FDG PET/CT, resulting in high lesion detection rate and tumor delineation. CONCLUSION The dual targeting radiotracer [68Ga]Ga-LNC1015 showed significantly improved tumor uptake and retention, as well as lower liver uptake than [68Ga]Ga-RGD and [68Ga]Ga-RM26 monomer. The first-in-human study showed high TBRs in patients, suggesting favorable pharmacokinetics and high clinical feasibility for PET/CT imaging of cancer.
Collapse
Affiliation(s)
- Xuejun Wen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Pengfei Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Mengqi Shi
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Qingyao Shang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xueyuan Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xinying Zeng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Jia Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Zhide Guo
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
9
|
Xiao T, Lee J, Gauntner TD, Velegraki M, Lathia JD, Li Z. Hallmarks of sex bias in immuno-oncology: mechanisms and therapeutic implications. Nat Rev Cancer 2024; 24:338-355. [PMID: 38589557 DOI: 10.1038/s41568-024-00680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/10/2024]
Abstract
Sex differences are present across multiple non-reproductive organ cancers, with male individuals generally experiencing higher incidence of cancer with poorer outcomes. Although some mechanisms underlying these differences are emerging, the immunological basis is not well understood. Observations from clinical trials also suggest a sex bias in conventional immunotherapies with male individuals experiencing a more favourable response and female individuals experiencing more severe adverse events to immune checkpoint blockade. In this Perspective article, we summarize the major biological hallmarks underlying sex bias in immuno-oncology. We focus on signalling from sex hormones and chromosome-encoded gene products, along with sex hormone-independent and chromosome-independent epigenetic mechanisms in tumour and immune cells such as myeloid cells and T cells. Finally, we highlight opportunities for future studies on sex differences that integrate sex hormones and chromosomes and other emerging cancer hallmarks such as ageing and the microbiome to provide a more comprehensive view of how sex differences underlie the response in cancer that can be leveraged for more effective immuno-oncology approaches.
Collapse
Affiliation(s)
- Tong Xiao
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy D Gauntner
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Maria Velegraki
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
- Rose Ella Burkhardt Brain Tumour Center, Cleveland Clinic, Cleveland, OH, USA.
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA.
| |
Collapse
|
10
|
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
|
11
|
O'Shea A, Iravani A, Saboury B, Jadvar H, Catalano O, Mahmood U, Heidari P. Integrating Theranostics Into Patient Care Pathways: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2023; 220:619-629. [PMID: 36321986 PMCID: PMC10133840 DOI: 10.2214/ajr.22.28237] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Theranostics describes the coupling of a diagnostic biomarker and a therapeutic agent (i.e., a theranostic pair) that have a common target in tumor cells or their microenvironment. The term is increasingly associated with in vivo nuclear medicine oncologic applications that couple diagnostic imaging by means of gamma radiation with concomitant localized high-energy particulate radiation to a tissue expressing the common target. Several theranostic pairs have been translated into clinical practice in the United States and are poised to become a mainstay of cancer treatment. The purposes of this article are to review experience with theranostics for solid-organ malignancies and to address the practical integration into care pathways of β-emitting therapies that include somatostatin analogue radioligands for neuroendocrine tumors, PSMA-directed therapy for prostate cancer, and 131I-MIBG therapy for tumors of neural crest origin. Toxicities related to theranostics administration and indications for cessation of therapy in patients who experience adverse events are also discussed. A multidisciplinary team-based approach for identifying patients most likely to respond to these agents, determining the optimal time for therapy delivery, and managing patient care throughout the therapeutic course is critical to the success of a radiotheranostic program.
Collapse
Affiliation(s)
- Aileen O'Shea
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, White 427, Boston, MA 02115
| | | | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, MD
- Department of Computer Science and Electrical Engineering, University of Maryland, Baltimore County, Baltimore, MD
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Hossein Jadvar
- Division of Nuclear Medicine and Molecular Imaging Center, Keck School of Medicine and Viterbi School of Engineering, University of Southern California, Los Angeles, CA
| | - Onofrio Catalano
- Division of Abdominal Imaging, Massachusetts General Hospital, Boston, MA
| | - Umar Mahmood
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, White 427, Boston, MA 02115
| | - Pedram Heidari
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, White 427, Boston, MA 02115
| |
Collapse
|
12
|
Nock BA, Kanellopoulos P, Joosten L, Mansi R, Maina T. Peptide Radioligands in Cancer Theranostics: Agonists and Antagonists. Pharmaceuticals (Basel) 2023; 16:ph16050674. [PMID: 37242457 DOI: 10.3390/ph16050674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical success of radiolabeled somatostatin analogs in the diagnosis and therapy-"theranostics"-of tumors expressing the somatostatin subtype 2 receptor (SST2R) has paved the way for the development of a broader panel of peptide radioligands targeting different human tumors. This approach relies on the overexpression of other receptor-targets in different cancer types. In recent years, a shift in paradigm from internalizing agonists to antagonists has occurred. Thus, SST2R-antagonist radioligands were first shown to accumulate more efficiently in tumor lesions and clear faster from the background in animal models and patients. The switch to receptor antagonists was soon adopted in the field of radiolabeled bombesin (BBN). Unlike the stable cyclic octapeptides used in the case of somatostatin, BBN-like peptides are linear, fast to biodegradable and elicit adverse effects in the body. Thus, the advent of BBN-like antagonists provided an elegant way to obtain effective and safe radiotheranostics. Likewise, the pursuit of gastrin and exendin antagonist-based radioligands is advancing with exciting new outcomes on the horizon. In the present review, we discuss these developments with a focus on clinical results, commenting on challenges and opportunities for personalized treatment of cancer patients by means of state-of-the-art antagonist-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Berthold A Nock
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| | | | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| |
Collapse
|
13
|
Trencsényi G, Képes Z. Scandium-44: Diagnostic Feasibility in Tumor-Related Angiogenesis. Int J Mol Sci 2023; 24:ijms24087400. [PMID: 37108559 PMCID: PMC10138813 DOI: 10.3390/ijms24087400] [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: 03/15/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Angiogenesis-related cell-surface molecules, including integrins, aminopeptidase N, vascular endothelial growth factor, and gastrin-releasing peptide receptor (GRPR), play a crucial role in tumour formation. Radiolabelled imaging probes targeting angiogenic biomarkers serve as valuable vectors in tumour identification. Nowadays, there is a growing interest in novel radionuclides other than gallium-68 (68Ga) or copper-64 (64Cu) to establish selective radiotracers for the imaging of tumour-associated neo-angiogenesis. Given its ideal decay characteristics (Eβ+average: 632 KeV) and a half-life (T1/2 = 3.97 h) that is well matched to the pharmacokinetic profile of small molecules targeting angiogenesis, scandium-44 (44Sc) has gained meaningful attention as a promising radiometal for positron emission tomography (PET) imaging. More recently, intensive research has been centered around the investigation of 44Sc-labelled angiogenesis-directed radiopharmaceuticals. Previous studies dealt with the evaluation of 44Sc-appended avb3 integrin-affine Arg-Gly-Asp (RGD) tripeptides, GRPR-selective aminobenzoyl-bombesin analogue (AMBA), and hypoxia-associated nitroimidazole derivatives in the identification of various cancers using experimental tumour models. Given the tumour-related hypoxia- and angiogenesis-targeting capability of these PET probes, 44Sc seems to be a strong competitor of the currently used positron emitters in radiotracer development. In this review, we summarize the preliminary preclinical achievements with 44Sc-labelled angiogenesis-specific molecular probes.
Collapse
Affiliation(s)
- György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| |
Collapse
|
14
|
Sun HL, Ma QY, Bian HG, Meng XM, Jin J. Novel insight on GRP/GRPR axis in diseases. Biomed Pharmacother 2023; 161:114497. [PMID: 36933382 DOI: 10.1016/j.biopha.2023.114497] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/26/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The gastrin-releasing peptide receptor (GRPR), a member of the G protein-coupled receptors (GPCRs), binds to ligands such as gastrin-releasing peptide (GRP) and plays a variety of biological roles. GRP/GRPR signalling is involved in the pathophysiological processes of many diseases, including inflammatory diseases, cardiovascular diseases, neurological diseases, and various cancers. In the immune system, the unique function of GRP/GRPR in neutrophil chemotaxis suggests that GRPR can be directly stimulated through GRP-mediated neutrophils to activate selective signalling pathways, such as PI3K, PKC, and MAPK, and participate in the occurrence and development of inflammation-related diseases. In the cardiovascular system, GRP increases intercellular adhesion molecule 1 (ICAM-1) and induces vascular cell adhesion molecule-1 (VCAM-1). GRP activates ERK1/2, MAPK, and AKT, leading to cardiovascular diseases, including myocardial infarction. Central nervous system signal transduction mediated by the GRP/GRPR axis plays a vital role in emotional responses, social interaction, and memory. The GRP/GRPR axis is elevated in various cancers, including lung, cervical, colorectal, renal cell, and head and neck squamous cell carcinomas. GRP is a mitogen in a variety of tumour cell lines. Its precursor, pro-gastrin-releasing peptide (ProGRP), may play an important role as an emerging tumour marker in early tumour diagnosis. GPCRs serve as therapeutic targets for drug development, but their function in each disease remains unclear, and their involvement in disease progression has not been well explored or summarised. This review lays out the above mentioned pathophysiological processes based on previous research conclusions. The GRP/GRPR axis may be a potential target for treating multiple diseases, and the study of this signalling axis is particularly important.
Collapse
Affiliation(s)
- Hao-Lu Sun
- School of Basic Medical Sciences, Anhui Medical University, Anhui, China
| | - Qiu-Ying Ma
- Department of pharmacy, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, No. 100 Huaihai Road, Hefei, Anhui, 230012, China
| | - He-Ge Bian
- School of Basic Medical Sciences, Anhui Medical University, Anhui, China
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei 230032, China.
| | - Juan Jin
- School of Basic Medical Sciences, Anhui Medical University, Anhui, China.
| |
Collapse
|
15
|
Kunos CA, Fabian D, Napier D, Stonecypher MS, Duncan RM, Hurt J. Human gastrin- releasing peptide receptor expression in women with uterine cervix cancer. Front Oncol 2023; 13:1126426. [PMID: 36761980 PMCID: PMC9905715 DOI: 10.3389/fonc.2023.1126426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction 212Pb-DOTAM-GRPR1 is a pharmaceutical radioimmunoconjugate consisiting of an α-particle-emitting radionuclide lead-212 (212Pb), a metal chelator DOTAM (1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane), and a gastrin-releasing peptide receptor (GRPR)-targeted antagonist currently being evaluated as therapy in uterine cervix and other cancer types. Previous studies have revealed that a variable proportion of uterine cervix cancer tumors overexpress the radiopharmaceutical target GRPR when assessed by cell proportion and staining intensity immunoreactive scores (IRS). Tumor response to 212Pb-DOTAM-GRPR1 strongly associates with GRPR overexpression, and therefore, it seems reasonable to assess uterine cervix cancer GRPR immunoreactivity for greater insight into the feasibility of using 212Pb-DOTAM-GRPR1 as a radiopharmaceutical treatment. Methods We examined a series of 33 uterine cervix cancer paraffin-embedded tumors in order to establish whether this tumor type overexpresses GRPR at an IRS score of 6 or higher, as 212Pb-DOTAM-GRPR1 is currently being evaluated in clinical trials against tumors showing such a level of expression. Results The results show that five of five (100%) primary adenocarcinomas and 10 of 16 (63%) primary squamous cell tumors overexpress GRPR at an IRS score of 6 or higher. Discussion The frequency of overexpression in this study suggests that 212Pb-DOTAM-GRPR1 radiopharmaceutical treatment may be useful in the management of persistent, recurrent, or metastatic uterine cervix cancer patients. A phase I clinical trial involving patients with metastatic uterine cervix cancer is currently underway (NCT05283330).
Collapse
Affiliation(s)
- Charles A. Kunos
- Department of Radiation Medicine, University of Kentucky, Lexington, KY, United States,*Correspondence: Charles A. Kunos,
| | - Denise Fabian
- Department of Radiation Medicine, University of Kentucky, Lexington, KY, United States
| | - Dana Napier
- Biospecimen Procurement & Translational Pathology, University of Kentucky, Lexington, KY, United States
| | | | - Ravyn M. Duncan
- Molecular Pathology Laboratory Network, Inc., Maryville, TN, United States
| | | |
Collapse
|
16
|
Duan H, Baratto L, Fan RE, Soerensen SJC, Liang T, Chung BI, Thong AEC, Gill H, Kunder C, Stoyanova T, Rusu M, Loening AM, Ghanouni P, Davidzon GA, Moradi F, Sonn GA, Iagaru A. Correlation of 68Ga-RM2 PET with Postsurgery Histopathology Findings in Patients with Newly Diagnosed Intermediate- or High-Risk Prostate Cancer. J Nucl Med 2022; 63:1829-1835. [PMID: 35552245 DOI: 10.2967/jnumed.122.263971] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Indexed: 01/11/2023] Open
Abstract
68Ga-RM2 targets gastrin-releasing peptide receptors (GRPRs), which are overexpressed in prostate cancer (PC). Here, we compared preoperative 68Ga-RM2 PET to postsurgery histopathology in patients with newly diagnosed intermediate- or high-risk PC. Methods: Forty-one men, 64.0 ± 6.7 y old, were prospectively enrolled. PET images were acquired 42-72 min (median ± SD, 52.5 ± 6.5 min) after injection of 118.4-247.9 MBq (median ± SD, 138.0 ± 22.2 MBq) of 68Ga-RM2. PET findings were compared with preoperative multiparametric MRI (mpMRI) (n = 36) and 68Ga-PSMA11 PET (n = 17) and correlated to postprostatectomy whole-mount histopathology (n = 32) and time to biochemical recurrence. Nine participants decided to undergo radiation therapy after study enrollment. Results: All participants had intermediate- (n = 17) or high-risk (n = 24) PC and were scheduled for prostatectomy. Prostate-specific antigen was 8.8 ± 77.4 (range, 2.5-504) and 7.6 ± 5.3 ng/mL (range, 2.5-28.0 ng/mL) when participants who ultimately underwent radiation treatment were excluded. Preoperative 68Ga-RM2 PET identified 70 intraprostatic foci of uptake in 40 of 41 patients. Postprostatectomy histopathology was available in 32 patients in which 68Ga-RM2 PET identified 50 of 54 intraprostatic lesions (detection rate = 93%). 68Ga-RM2 uptake was recorded in 19 nonenlarged pelvic lymph nodes in 6 patients. Pathology confirmed lymph node metastases in 16 lesions, and follow-up imaging confirmed nodal metastases in 2 lesions. 68Ga-PSMA11 and 68Ga-RM2 PET identified 27 and 26 intraprostatic lesions, respectively, and 5 pelvic lymph nodes each in 17 patients. Concordance between 68Ga-RM2 and 68Ga-PSMA11 PET was found in 18 prostatic lesions in 11 patients and 4 lymph nodes in 2 patients. Noncongruent findings were observed in 6 patients (intraprostatic lesions in 4 patients and nodal lesions in 2 patients). Sensitivity and accuracy rates for 68Ga-RM2 and 68Ga-PSMA11 (98% and 89% for 68Ga-RM2 and 95% and 89% for 68Ga-PSMA11) were higher than those for mpMRI (77% and 77%, respectively). Specificity was highest for mpMRI with 75% followed by 68Ga-PSMA11 (67%) and 68Ga-RM2 (65%). Conclusion: 68Ga-RM2 PET accurately detects intermediate- and high-risk primary PC, with a detection rate of 93%. In addition, 68Ga-RM2 PET showed significantly higher specificity and accuracy than mpMRI and a performance similar to 68Ga-PSMA11 PET. These findings need to be confirmed in larger studies to identify which patients will benefit from one or the other or both radiopharmaceuticals.
Collapse
Affiliation(s)
- Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Lucia Baratto
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Richard E Fan
- Department of Urology, Stanford University, Stanford, California
| | - Simon John Christoph Soerensen
- Department of Urology, Stanford University, Stanford, California.,Department of Epidemiology and Population Health, Stanford University, Stanford, California
| | - Tie Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | | | | | - Harcharan Gill
- Department of Urology, Stanford University, Stanford, California
| | - Christian Kunder
- Department of Pathology, Stanford University, Stanford, California
| | - Tanya Stoyanova
- Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, California
| | - Mirabela Rusu
- Division of Integrative Biomedical Imaging, Department of Radiology, Stanford University, Stanford, California; and
| | - Andreas M Loening
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, California
| | - Pejman Ghanouni
- Division of Body MRI, Department of Radiology, Stanford University, Stanford, California
| | - Guido A Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Geoffrey A Sonn
- Department of Urology, Stanford University, Stanford, California
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California;
| |
Collapse
|
17
|
Abstract
Molecular imaging is advancing rapidly with promising new molecular targets emerging for theragnostic, ie, imaging and treatment with the same compound, to provide targeted, personalized medicine. Gastrin-releasing peptide receptors (GRPR) are overexpressed in prostate cancer. Gallium-68 (68Ga) RM2 is a GRPR antagonist and shows high sensitivity and specificity for the detection of primary prostate cancer and recurrent disease. However, compared with the widely used 68Ga-PSMA11 and 18F-DCFPyL, a discordance in uptake pattern is seen reflecting the heterogeneity in tumor biology of prostate cancer. In this review, we present the background, current status, and future perspectives of PET imaging using 68Ga-RM2.
Collapse
Affiliation(s)
- Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA 94305, USA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, 300 Pasteur Drive, H2200, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
Lyu S, Zhang X, Tu Z, Zhou H, Ke X, Qu Y. GPR108 is required for gambogic acid inhibiting NF-κB signaling in cancer. Pharmacol Res 2022; 182:106279. [PMID: 35659621 DOI: 10.1016/j.phrs.2022.106279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/14/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
GPCRs are the most potential targets for drug discovery, however, their role in oncology is underappreciated and GPCR-based anti-cancer drug is not fully investigated. Herein, we identified GPR108, a GPCR protein described in innate immune system, is a potential therapeutic target of cancer. Depletion of GPR108 dramatically inhibited the survival of various cancers. Notably, TNFα activation of NF-κB was totally impaired after GPR108 knockout. We identified gambogic acid (GA), a natural prenylated xanthone, selectively targeting GPR108. Importantly, GA engaged with GPR108 and promoted its degradation, knockout of GPR108 remarkably blocked GA inhibition of NF-κB signaling. Furthermore, in vitro and in vivo assays demonstrated that GA was dependent on GPR108 to exert anti-cancer activity. Overall, our findings supported GPR108 as a promising therapeutic target of cancer, and provided a small molecule inhibitor GA directly and selectively targeting GPR108 for cancer therapy.
Collapse
Affiliation(s)
- Song Lyu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xue Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhenzhen Tu
- Department of Biochemistry and Molecular Biology, Anhui Medical University, No. 69 Mei Shan Road, Hefei, China
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, Anhui Medical University, No. 69 Mei Shan Road, Hefei, China
| | - Xisong Ke
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yi Qu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
19
|
Chuang CC, Lan YH, Lu YJ, Weng YL, Chen JP. Targeted delivery of irinotecan and SLP2 shRNA with GRP-conjugated magnetic graphene oxide for glioblastoma treatment. Biomater Sci 2022; 10:3201-3222. [PMID: 35579261 DOI: 10.1039/d2bm00420h] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Magnetic nanoparticles (MNPs) are useful for magnetic targeted drug delivery while ligand-mediated active targeting is another common delivery strategy for cancer therapy. In this work, we intend to prepare magnetic graphene oxide (mGO) by chemical co-precipitation of MNPs on the GO surface, followed by conjugation of the gastrin releasing peptide (GRP) as a targeting ligand, for dual targeted drug/gene delivery in invasive brain glioma treatment. mGO was grafted with chitosan, complexed with shRNA plasmid DNA for stomatin-like protein 2 (SLP2) gene silencing, modified with urocanic acid for plasmid DNA endosomal escape, PEGylated for GRP conjugation, and loaded with the chemotherapeutic drug irinotecan (CPT-11) by π-π interaction for pH-responsive drug release (mGOCUG/CPT-11/shRNA). In addition to the in depth characterization of the physico-chemical and biological properties during each preparation step, we also study the loading/pH-responsive release behavior of CPT-11 and the shRNA plasmid loading/cell transfection efficiency. The targeting and antitumor efficacies of the nanocomposite were studied with U87 human glioblastoma cells in vitro. An in vivo study revealed that intravenous administration followed by magnetic guidance results in the efficient targeted delivery of mGOCUG/CPT-11/shRNA to orthotopic brain tumors in nude mice, and it exhibits excellent antitumor efficacy with a reduced tumor growth rate and prolonged animal survival time. Our work thus highlights a multifunctional mGO-based drug/gene delivery platform for effective combination cancer therapy in glioblastoma treatment.
Collapse
Affiliation(s)
- Chi-Cheng Chuang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yu-Hsiang Lan
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yu-Lun Weng
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan 33305, Taiwan
| | - Jyh-Ping Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan.
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan 33305, Taiwan
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
20
|
Zhu Y, Wang H, Yu M, Li C, Meng X, He M, Yao R. Design, synthesis and biological evaluation of novel 1, 3, 4-oxadiazole PD176252 analogues as potential GRPR inhibitors. Anticancer Agents Med Chem 2022; 22:3009-3024. [DOI: 10.2174/1871520622666220501162813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/08/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022]
Abstract
Background:
GRPR is over-expressed in cancer cells and is a potential drug target for the treatment of cancer. PD176252, as the most representative non-peptide inhibitor of GRPR, can inhibit the growth of cancer cells, but its low selectivity to cancer cells and normal cells limits its further application.
Objective:
The aim of this study was to design and synthesize novel GRPR inhibitor with stronger anti-cancer activity and higher affinity with GRPR than the lead compound PD176252.
Methods:
A series of 1, 3, 4-oxadiazole derivatives as PD176252 analogues (4a-4j, 6a-6q) were synthesized and investigated their cytotoxic activity on four cancer lines with high expression of GRPR (gastric (HGC-27), colon (HCT-116), prostate (PC-3), and lung (A549)) and one human cell line (gastric mucosal epithelial (GES-1)) by MTT assay. Flow cytometry analysis and Western Blot were used to determine whether the compound induced programmed apoptosis of cancer cells. Competitive binding experiment was used to verify the affinity between GRPR and the optimal compound.
Results:
Compound 6m exhibited significant growth inhibition on all tested cancer cell lines, especially gastric cancer cells (HGC-27 cellular IC50 0.37±0.04μM). Also, the selectivity of 6m to HGC-27 was much higher than that of PD176252. Flow cytometric analysis and Western Blot proved that 6m significantly promoted the apoptosis of HGC-27 cells. Moreover, competitive binding experiment confirmed the close binding of 6m with GRPR, which indicated 6m with a higher affinity than lead compound PD176252.
Conclusion:
Our results suggested that 6m, as a novel GRPR inhibitor, had a higher affinity with GRPR and potential anti-cancer effect than PD176252, which can be used as a template for further optimization.
Collapse
Affiliation(s)
- Yuting Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Huai Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Mingjun Yu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
- Traditional Chinese Medicine College, Bozhou University, Bozhou, 236800, China
| | - Chao Li
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Meng He
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Risheng Yao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| |
Collapse
|
21
|
Mansi R, Nock BA, Dalm SU, Busstra MB, van Weerden WM, Maina T. Radiolabeled Bombesin Analogs. Cancers (Basel) 2021; 13:cancers13225766. [PMID: 34830920 PMCID: PMC8616220 DOI: 10.3390/cancers13225766] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Recent medical advancements have strived for a personalized medicine approach to patients, aimed at optimizing therapy outcomes with minimum toxicity. In this respect, nuclear medicine methodologies have been playing increasingly important roles. For example, the overexpression of peptide receptors, such as the gastrin-releasing peptide receptor (GRPR), on tumor cells as opposed to their lack of expression in healthy surrounding tissues can be elegantly exploited with the aid of “smart” peptide carriers, such as the analogs of the amphibian 14-peptide bombesin (BBN). These molecules can bring clinically attractive radionuclides to malignant lesions in prostate, breast, and other human cancers, sparing healthy tissues. Depending upon the radionuclide in question, diagnostic imaging with single-photon emission computed tomography (SPECT) or positron emission tomography (PET) has been pursued, identifying patients who are eligible for peptide radionuclide receptor therapy (PRRT) in an integrated “theranostic” approach. In the present review, we (i) discuss the major steps taken in the development of anti-GRPR theranostic radioligands, with a focus on those selected for clinical testing; (ii) comment on the present status in this field of research; and (iii) reflect on the current limitations as well as on new opportunities for their broader and more successful clinical applications. Abstract The gastrin-releasing peptide receptor (GRPR) is expressed in high numbers in a variety of human tumors, including the frequently occurring prostate and breast cancers, and therefore provides the rationale for directing diagnostic or therapeutic radionuclides on cancer lesions after administration of anti-GRPR peptide analogs. This concept has been initially explored with analogs of the frog 14-peptide bombesin, suitably modified at the N-terminus with a number of radiometal chelates. Radiotracers that were selected for clinical testing revealed inherent problems associated with these GRPR agonists, related to low metabolic stability, unfavorable abdominal accumulation, and adverse effects. A shift toward GRPR antagonists soon followed, with safer analogs becoming available, whereby, metabolic stability and background clearance issues were gradually improved. Clinical testing of three main major antagonist types led to promising outcomes, but at the same time brought to light several limitations of this concept, partly related to the variation of GRPR expression levels across cancer types, stages, previous treatments, and other factors. Currently, these parameters are being rigorously addressed by cell biologists, chemists, nuclear medicine physicians, and other discipline practitioners in a common effort to make available more effective and safe state-of-the-art molecular tools to combat GRPR-positive tumors. In the present review, we present the background, current status, and future perspectives of this endeavor.
Collapse
Affiliation(s)
- Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine University Hospital Basel, 4031 Basel, Switzerland;
| | - Berthold A. Nock
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, 15310 Athens, Greece;
| | - Simone U. Dalm
- Erasmus Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (S.U.D.); (M.B.B.); (W.M.v.W.)
| | - Martijn B. Busstra
- Erasmus Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (S.U.D.); (M.B.B.); (W.M.v.W.)
| | - Wytske M. van Weerden
- Erasmus Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands; (S.U.D.); (M.B.B.); (W.M.v.W.)
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, 15310 Athens, Greece;
- Correspondence: ; Tel.: +30-650-3908/3891
| |
Collapse
|
22
|
[ 99mTc]Tc-DB15 in GRPR-Targeted Tumor Imaging with SPECT: From Preclinical Evaluation to the First Clinical Outcomes. Cancers (Basel) 2021; 13:cancers13205093. [PMID: 34680243 PMCID: PMC8533986 DOI: 10.3390/cancers13205093] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Diagnostic imaging and radionuclide therapy of prostate (PC) and breast cancer (BC) using radiolabeled gastrin-releasing peptide receptor (GRPR)-antagonists represents a promising approach. We herein propose the GRPR-antagonist based radiotracer [99mTc]Tc-DB15 ([99mTc]Tc-N4-AMA-DGA-DPhe6,Sar11,LeuNHEt13]BBN(6-13); N4: 6-carboxy-1,4,8,11-tetraazaundecane, AMA: aminomethyl-aniline, DGA: diglycolic acid) as a new diagnostic tool for GRPR-positive tumors applying SPECT/CT. The uptake of [99mTc]Tc-DB15 was tested in vitro in mammary (T-47D) and prostate cancer (PC-3) cells and in vivo in T-47D or PC-3 xenograft-bearing mice as well as in BC patients. DB15 showed high GRPR-affinity (IC50 = 0.37 ± 0.03 nM) and [99mTc]Tc-DB15 strongly bound to the cell-membrane of T-47D and PC-3 cells, according to a radiolabeled antagonist profile. In mice, the radiotracer showed high and prolonged GRPR-specific uptake in PC-3 (e.g., 25.56 ± 2.78 %IA/g vs. 0.72 ± 0.12 %IA/g in block; 4 h pi) and T-47D (e.g., 15.82 ± 3.20 %IA/g vs. 3.82 ± 0.30 %IA/g in block; 4 h pi) tumors, while rapidly clearing from background. In patients with advanced BC, the tracer could reveal several bone and soft tissue metastases on SPECT/CT. The attractive pharmacokinetic profile of [99mTc]DB15 in mice and its capability to target GRPR-positive BC lesions in patients highlight its prospects for a broader clinical use, an option currently being explored by ongoing clinical studies.
Collapse
|
23
|
[ 99mTc]Tc-DB1 Mimics with Different-Length PEG Spacers: Preclinical Comparison in GRPR-Positive Models. Molecules 2020; 25:molecules25153418. [PMID: 32731473 PMCID: PMC7435657 DOI: 10.3390/molecules25153418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background: The frequent overexpression of gastrin-releasing peptide receptors (GRPRs) in human cancers provides the rationale for delivering clinically useful radionuclides to tumor sites using peptide carriers. Radiolabeled GRPR antagonists, besides being safer for human use, have often shown higher tumor uptake and faster background clearance than agonists. We herein compared the biological profiles of the GRPR-antagonist-based radiotracers [99mTc]Tc-[N4-PEGx-DPhe6,Leu-NHEt13]BBN(6-13) (N4: 6-(carboxy)-1,4,8,11-tetraazaundecane; PEG: polyethyleneglycol): (i) [99mTc]Tc-DB7 (x = 2), (ii) [99mTc]Tc-DB13 (x = 3), and (iii) [99mTc]Tc-DB14 (x = 4), in GRPR-positive cells and animal models. The impact of in situ neprilysin (NEP)-inhibition on in vivo stability and tumor uptake was also assessed by treatment of mice with phosphoramidon (PA). Methods: The GRPR affinity of DB7/DB13/DB14 was determined in PC-3 cell membranes, and cell binding of the respective [99mTc]Tc-radioligands was assessed in PC-3 cells. Each of [99mTc]Tc-DB7, [99mTc]Tc-DB13, and [99mTc]Tc-DB14 was injected into mice without or with PA coinjection and 5 min blood samples were analyzed by HPLC. Biodistribution was conducted at 4 h postinjection (pi) in severe combined immunodeficiency disease (SCID) mice bearing PC-3 xenografts without or with PA coinjection. Results: DB7, -13, and -14 displayed single-digit nanomolar affinities for GRPR. The uptake rates of [99mTc]Tc-DB7, [99mTc]Tc-DB13, and [99mTc]Tc-DB14 in PC-3 cells was comparable and consistent with a radioantagonist profile. The radiotracers were found to be ≈70% intact in mouse blood and >94% intact after coinjection of PA. Treatment of mice with PA enhanced tumor uptake. Conclusions: The present study showed that increase of PEG-spacer length in the [99mTc]Tc-DB7-[99mTc]Tc-DB13-[99mTc]Tc-DB14 series had little effect on GRPR affinity, specific uptake in PC-3 cells, in vivo stability, or tumor uptake. A significant change in in vivo stability and tumor uptake was observed only after treatment of mice with PA, without compromising the favorably low background radioactivity levels.
Collapse
|
24
|
Li R, Gao R, Wang Y, Liu Z, Xu H, Duan A, Zhang F, Ma L. Gastrin releasing peptide receptor targeted nano-graphene oxide for near-infrared fluorescence imaging of oral squamous cell carcinoma. Sci Rep 2020; 10:11434. [PMID: 32651409 PMCID: PMC7351765 DOI: 10.1038/s41598-020-68203-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant tumor that occurs in the oral mucosa. Pathological biopsy is still the current gold standard for OSCC diagnosis; however, some drawbacks need to be overcome. Therefore, it is urgently needed to find a non-invasive targeted technology for OSCC early diagnosis. Fluorescent optical imaging using near infrared (NIR) dyes tagged to tumor specific target will benefit such developments. Gastrin releasing peptide receptor (GRPR) is an attractive target for OSCC imaging and therapy. In this study, we synthesized nano-graphene oxide (NGO) nanoparticles with GRPR-specific peptides AF750-6Ahx-Sta-BBN via hydrogen bond and π–π bonds (NGO-BBN-AF750), and investigated their receptor binding, cell uptake and internalization in HSC-3 cells. NGO-BBN-AF750 and AF750-6Ahx-Sta-BBN showed a similar binding affinity to GRPR on HSC-3 cells. In contrast to AF750-6Ahx-Sta-BBN antagonist peptide, NGO-BBN-AF750 showed cellular internalization property. Overall, this study proposes a NGO nanoclusters-based nanoprobe for GRPR targeted near-infrared fluorescence imaging for OSCC. Nanoparticle-based delivery systems have shown highly significant potential in the delivery of a wide range of therapeutic agents.
Collapse
Affiliation(s)
- Ran Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Ruifang Gao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Yimei Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Zhuanzhuan Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Hang Xu
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA.,Department of Radiology, University of Missouri, Columbia, MO, 65212, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Ailin Duan
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
| | - Fang Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
| | - Lixin Ma
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, USA. .,Department of Radiology, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
25
|
Lee L, Ramos-Alvarez I, Moody TW, Mantey SA, Jensen RT. Neuropeptide bombesin receptor activation stimulates growth of lung cancer cells through HER3 with a MAPK-dependent mechanism. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118625. [PMID: 31862538 PMCID: PMC7008087 DOI: 10.1016/j.bbamcr.2019.118625] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/15/2019] [Accepted: 12/14/2019] [Indexed: 01/28/2023]
Abstract
Despite recent advances in treatment of non-small cell lung cancer (NSCLC), prognosis still remains poor and new therapeutic approaches are needed. Studies demonstrate the importance of the EGFR/HER-receptor family in NSCLC growth, as well as that of other tumors. Recently, HER3 is receiving increased attention because of its role in drug resistance and aggressive growth. Activation of overexpressed G-protein-coupled receptors (GPCR) can also initiate growth by transactivating EGFR/HER-family members. GPCR transactivation of EGFR has been extensively studied, but little is known of its ability to transactivate other EGFR/HER-members, especially HER3. To address this, we studied the ability of bombesin receptor (BnR) activation to transactivate all EGFR/HER-family members and their principal downstream signaling cascades, the PI3K/Akt- and MAPK/ERK-pathways, in human NSCLC cell-lines. In all three cell-lines studied, which possessed EGFR, HER2 and HER3, Bn rapidly transactivated EGFR, HER2 and HER3, as well as Akt and ERK. Immunoprecipitation studies revealed Bn-induced formation of both HER3/EGFR- and HER3/HER2-heterodimers. Specific EGFR/HER3 antibodies or siRNA-knockdown of EGFR and HER3, demonstrated Bn-stimulated activation of EGFR/HER members is initially through HER3, not EGFR. In addition, specific inhibition of HER3, HER2 or MAPK, abolished Bn-stimulated cell-growth, while neither EGFR nor Akt inhibition had an effect. These results show HER3 transactivation mediates all growth effects of BnR activation through MAPK. These results raise the possibility that targeting HER3 alone or with GPCR activation and its signal cascades, may be a novel therapeutic approach in NSCLC. This is especially relevant with the recent development of HER3-blocking antibodies.
Collapse
Affiliation(s)
- Lingaku Lee
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Irene Ramos-Alvarez
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Terry W Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Research, Office of the Director, Bethesda, MD 20892, USA
| | - Samuel A Mantey
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert T Jensen
- Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
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
|
27
|
Hoppenz P, Els-Heindl S, Kellert M, Kuhnert R, Saretz S, Lerchen HG, Köbberling J, Riedl B, Hey-Hawkins E, Beck-Sickinger AG. A Selective Carborane-Functionalized Gastrin-Releasing Peptide Receptor Agonist as Boron Delivery Agent for Boron Neutron Capture Therapy. J Org Chem 2019; 85:1446-1457. [PMID: 31813224 DOI: 10.1021/acs.joc.9b02406] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Boron neutron capture therapy (BNCT) allows the selective elimination of malignant tumor cells without affecting healthy tissue. Although this binary radiotherapy approach has been known for decades, BNCT failed to reach the daily clinics to date. One of the reasons is the lack of selective boron delivery agents. Using boron loaded peptide conjugates, which address G protein-coupled receptors overexpressed on tumor cells allow the intracellular accumulation of boron. The gastrin-releasing peptide receptor (GRPR) is a well-known target in cancer diagnosis and can potentially be used for BNCT. Here, we present the successful introduction of multiple bis-deoxygalactosyl-carborane building blocks to the GRPR-selective ligand [d-Phe6, β-Ala11, Ala13, Nle14]Bn(6-14) (sBB2L) generating peptide conjugates with up to 80 boron atoms per molecule. Receptor activation was retained, metabolic stability was increased, and uptake into PC3 cells was proven without showing any intrinsic cytotoxicity. Furthermore, undesired uptake into liver cells was suppressed by using l-deoxygalactosyl modified carborane building blocks. Due to its high boron loading and excellent GRPR selectivity, this conjugate can be considered as a promising boron delivery agent for BNCT.
Collapse
Affiliation(s)
- Paul Hoppenz
- Institute of Biochemistry, Faculty of Life Sciences , Leipzig University , Brüderstrasse 34 , 04103 Leipzig , Germany
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Faculty of Life Sciences , Leipzig University , Brüderstrasse 34 , 04103 Leipzig , Germany
| | - Martin Kellert
- Institute of Inorganic Chemistry , Leipzig University , Johannisallee 29 , 04103 Leipzig , Germany
| | - Robert Kuhnert
- Institute of Inorganic Chemistry , Leipzig University , Johannisallee 29 , 04103 Leipzig , Germany
| | - Stefan Saretz
- Institute of Inorganic Chemistry , Leipzig University , Johannisallee 29 , 04103 Leipzig , Germany
| | | | | | - Bernd Riedl
- Bayer AG , Aprather Weg 18A , Wuppertal , Germany
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry , Leipzig University , Johannisallee 29 , 04103 Leipzig , Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences , Leipzig University , Brüderstrasse 34 , 04103 Leipzig , Germany
| |
Collapse
|
28
|
Kellert M, Hoppenz P, Lönnecke P, Worm DJ, Riedl B, Koebberling J, Beck-Sickinger AG, Hey-Hawkins E. Tuning a modular system - synthesis and characterisation of a boron-rich s-triazine-based carboxylic acid and amine bearing a galactopyranosyl moiety. Dalton Trans 2019; 49:57-69. [PMID: 31808482 DOI: 10.1039/c9dt04031e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Introduction of a bis(isopropylidene)-protected galactopyranosyl moiety in s-triazine-based boron-rich carboxylic acids and amines results in soluble and suitable coupling partners for tumour-selective biomolecules with applications as selective agents for boron neutron capture therapy (BNCT). Bearing either a carboxylic acid or primary amine as a functional group, these compounds are highly versatile and thus largely extend the possible coupling strategies with suitable biomolecules. Modification of the gastrin-releasing peptide receptor (GRPR) selective agonist [d-Phe6, β-Ala11, Ala13, Nle14]Bn(6-14) with the carboxylic acid derivative yielded a bioconjugate with an optimal receptor activation and internalisation profile. This demonstrates the great potential of this approach for the development of novel boron delivery agents.
Collapse
Affiliation(s)
- Martin Kellert
- Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Hoppenz P, Els‐Heindl S, Beck‐Sickinger AG. Identification and stabilization of a highly selective gastrin‐releasing peptide receptor agonist. J Pept Sci 2019; 25:e3224. [DOI: 10.1002/psc.3224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Paul Hoppenz
- Institute of BiochemistryLeipzig University Leipzig Germany
| | | | | |
Collapse
|
30
|
Baratto L, Duan H, Laudicella R, Toriihara A, Hatami N, Ferri V, Iagaru A. Physiological 68Ga-RM2 uptake in patients with biochemically recurrent prostate cancer: an atlas of semi-quantitative measurements. Eur J Nucl Med Mol Imaging 2019; 47:115-122. [DOI: 10.1007/s00259-019-04503-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/22/2019] [Indexed: 12/20/2022]
|
31
|
Proniewicz E, Tąta A, Starowicz M, Szkudlarek A, Pacek J, Molenda M, Kuśtrowski P. Ions-free electrochemically synthetized in aqueous media flake-like CuO nanostructures as SERS reproducible substrates for the detection of neurotransmitters. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 215:24-33. [PMID: 30825867 DOI: 10.1016/j.saa.2019.02.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/28/2019] [Accepted: 02/16/2019] [Indexed: 06/09/2023]
Abstract
The process of catalytic destruction of tumor cells can be strengthened by introducing copper(II) oxide nanostructures (CuONSs) with receptor's agonists/antagonists immobilized on their surface. Here we show a simple and reliable electrochemical method for the fabrication ions-free flake-like CuO nanostructures in a surfactant/ions free aqueous environment. For the determination of the metal surface plasmon, size, rheology, and structure of the fabricated nanostructures ultraviolet-visible (UV-Vis), Fourier-transform infrared (FT-IR), Raman, and X-ray photoelectron (XPS) spectroscopies as well as scanning electron microscope (SEM), high-resolution transmission electron microscopy with energy dispersive X-ray (HDTEM-EDS), X-ray powder diffraction (XRD), and dynamic light scattering (DLS) analysis were used. The fabricated nanostructures were used as highly sensitive, uniform, and reproducible sensors of a natural ligand (bombesin) of some types of metabotropic seven transmembrane G protein-coupled superfamily receptors (GPCRs), which are over-express on the surface of many malignant tumors. Surface-enhanced Raman scattering (SERS) was used to monitor the geometry of adsorbate, separate, enrich, and detect various bombesin C-terminal fragments. It has been shown that the type of used substrate, surface development, and ions present in the solution have little effect on the mode of adsorption.
Collapse
Affiliation(s)
- Edyta Proniewicz
- Faculty of Foundry Engineering, AGH University of Science and Technology, 30-059 Krakow, Poland.
| | - Agnieszka Tąta
- Faculty of Foundry Engineering, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Maria Starowicz
- Faculty of Foundry Engineering, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Aleksandra Szkudlarek
- Academic Centre for Materials and Nanotechnology, AGH University of Science and Technology, 30-055 Krakow, Poland
| | - Joanna Pacek
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Marcin Molenda
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Piotr Kuśtrowski
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| |
Collapse
|
32
|
Kaloudi A, Lymperis E, Kanellopoulos P, Waser B, de Jong M, Krenning EP, Reubi JC, Nock BA, Maina T. Localization of 99mTc-GRP Analogs in GRPR-Expressing Tumors: Effects of Peptide Length and Neprilysin Inhibition on Biological Responses. Pharmaceuticals (Basel) 2019; 12:ph12010042. [PMID: 30897789 PMCID: PMC6469168 DOI: 10.3390/ph12010042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
The overexpression of gastrin-releasing peptide receptors (GRPRs) in frequently occurring human tumors has provided the opportunity to use bombesin (BBN) analogs as radionuclide carriers to cancer sites for diagnostic and therapeutic purposes. We have been alternatively exploring human GRP motifs of higher GRPR selectivity compared to frog BBN sequences aiming to improve pharmacokinetic profiles. In the present study, we compared two differently truncated human endogenous GRP motifs: GRP(14–27) and GRP(18–27). An acyclic tetraamine was coupled at the N-terminus to allow for stable binding of the SPECT radionuclide 99mTc. Their biological profiles were compared in PC-3 cells and in mice without or with coinjection of phosphoramidon (PA) to induce transient neprilysin (NEP) inhibition in vivo. The two 99mTc-N4-GRP(14/18–27) radioligands displayed similar biological behavior in mice. Coinjection of PA exerted a profound effect on in vivo stability and translated into notably improved radiolabel localization in PC-3 experimental tumors. Hence, this study has shown that promising 99mTc-radiotracers for SPECT imaging may indeed derive from human GRP sequences. Radiotracer bioavailability was found to be of major significance. It could be improved during in situ NEP inhibition resulting in drastically enhanced uptake in GRPR-expressing lesions.
Collapse
Affiliation(s)
- Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Emmanouil Lymperis
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | | | - Beatrice Waser
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, CH-3010 Berne, Switzerland.
| | - Marion de Jong
- Department of Radiology & Nuclear Medicine Erasmus MC, 3015 CN Rotterdam, The Netherlands.
| | - Eric P Krenning
- Cytrotron Rotterdam BV, Erasmus MC, 3015 CN Rotterdam, The Netherlands.
| | - Jean Claude Reubi
- Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, CH-3010 Berne, Switzerland.
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| |
Collapse
|
33
|
Lymperis E, Kaloudi A, Kanellopoulos P, de Jong M, Krenning EP, Nock BA, Maina T. Comparing Gly 11/dAla 11-Replacement vs. the in-Situ Neprilysin-Inhibition Approach on the Tumor-targeting Efficacy of the 111In-SB3/ 111In-SB4 Radiotracer Pair. Molecules 2019; 24:molecules24061015. [PMID: 30871262 PMCID: PMC6471467 DOI: 10.3390/molecules24061015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 02/08/2023] Open
Abstract
Background: The GRPR-antagonist 68Ga-SB3 visualized prostate cancer lesions in animal models and in patients. Switching radiometal from 68Ga to 111In impaired tumor targeting in mice, but coinjection of the neprilysin (NEP)-inhibitor phosphoramidon (PA) stabilized 111In-SB3 in circulation and remarkably increased tumor uptake. We herein report on the biological profile of 111In-SB4: 111In-[dAla11]SB3. Methods: The biological responses of 111In-SB3/SB4 were compared in PC-3 cells and animal models. Results: Gly11/dAla11-replacement deteriorated GRPR-affinity (SB4 IC50: 10.7 ± 0.9 nM vs. SB3 IC50: 4.6 ± 0.3 nM) and uptake in PC-3 cells (111In-SB4: 1.3 ± 0.4% vs. 111In-SB3 16.2 ± 0.8% at 1 h). 111In-SB4 was more stable than 111In-SB3, but PA-coinjection stabilized both radiotracers in peripheral mice blood. Unmodified 111In-SB3 showed higher uptake in PC-3 xenografts (8.8 ± 3.0%ID/g) vs. 111In-SB4 (3.1 ± 1.1%ID/g) at 4 h pi. PA-coinjection improved tumor uptake, with 111In-SB3 still showing superior tumor targeting (38.3 ± 7.9%ID/g vs. 7.4 ± 0.3%ID/g for 111In-SB4). Conclusions: Replacement of Gly11 by dAla11 improved in vivo stability, however, at the cost of GRPR-affinity and cell uptake, eventually translating into inferior tumor uptake of 111In-SB4 vs. unmodified 111In-SB3. On the other hand, in-situ NEP-inhibition turned out to be a more efficient and direct strategy to optimize the in vivo profile of 111In-SB3, and potentially other peptide radiotracers.
Collapse
Affiliation(s)
- Emmanouil Lymperis
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | | | - Marion de Jong
- Department of Radiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | - Eric P Krenning
- Cytrotron Rotterdam BV, Erasmus MC, 3015 GD Rotterdam, The Netherlands.
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES, NCSR "Demokritos", 15310 Athens, Greece.
| |
Collapse
|
34
|
Xu H, Bandari RP, Lee L, Li R, Yu P, Smith CJ, Ma L. Design, Synthesis, and in Vitro and in Vivo Evaluation of High Affinity and Specificity Near-Infrared Fluorescent Bombesin Antagonists for Tumor Imaging. J Med Chem 2018; 61:7657-7670. [DOI: 10.1021/acs.jmedchem.8b00614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hang Xu
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Chemical Engineering, University of Missouri, Columbia, Missouri 65211, United States
| | - Rajendra P. Bandari
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| | - Li Lee
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Ran Li
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- Department of Stomatology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Ping Yu
- Department of Physics and Astronomy, University of Missouri, Columbia, Missouri 65211, United States
| | - Charles J. Smith
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
- University of Missouri Research Reactor Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Lixin Ma
- Research Division/Biomolecular Imaging Center, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri 65201, United States
- Departments of Radiology, University of Missouri, Columbia, Missouri 65212, United States
| |
Collapse
|
35
|
Nock BA, Charalambidis D, Sallegger W, Waser B, Mansi R, Nicolas GP, Ketani E, Nikolopoulou A, Fani M, Reubi JC, Maina T. New Gastrin Releasing Peptide Receptor-Directed [ 99mTc]Demobesin 1 Mimics: Synthesis and Comparative Evaluation. J Med Chem 2018. [PMID: 29517903 DOI: 10.1021/acs.jmedchem.8b00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We have previously reported on the gastrin releasing peptide receptor (GRPR) antagonist [99mTc]1, ([99mTc]demobesin 1, 99mTc-[N4'-diglycolate-dPhe6,Leu-NHEt13]BBN(6-13)). [99mTc]1 has shown superior biological profile compared to analogous agonist-based 99mTc-radioligands. We herein present a small library of [99mTc]1 mimics generated after structural modifications in (a) the linker ([99mTc]2, [99mTc]3, [99mTc]4), (b) the peptide chain ([99mTc]5, [99mTc]6), and (c) the C-terminus ([99mTc]7 or [99mTc]8). The effects of above modifications on the biological properties of analogs were studied in PC-3 cells and tumor-bearing SCID mice. All analogs showed subnanomolar affinity for the human GRPR, while most receptor-affine 4 and 8 behaved as potent GRPR antagonists in a functional internalization assay. In mice bearing PC-3 tumors, [99mTc]1-[99mTc]6 exhibited GRPR-specific tumor uptake, rapidly clearing from normal tissues. [99mTc]4 displayed the highest tumor uptake (28.8 ± 4.1%ID/g at 1 h pi), which remained high even after 24 h pi (16.3 ± 1.8%ID/g), well surpassing that of [99mTc]1 (5.4 ± 0.7%ID/g at 24 h pi).
Collapse
Affiliation(s)
- Berthold A Nock
- Molecular Radiopharmacy, INRASTES , National Center for Scientific Research "Demokritos" , GR-153 10 Athens , Greece
| | - David Charalambidis
- Molecular Radiopharmacy, INRASTES , National Center for Scientific Research "Demokritos" , GR-153 10 Athens , Greece
| | | | - Beatrice Waser
- Cell Biology and Experimental Cancer Research, Institute of Pathology , University of Berne , CH-3010 Berne , Switzerland
| | | | | | - Eleni Ketani
- Molecular Radiopharmacy, INRASTES , National Center for Scientific Research "Demokritos" , GR-153 10 Athens , Greece
| | - Anastasia Nikolopoulou
- Molecular Radiopharmacy, INRASTES , National Center for Scientific Research "Demokritos" , GR-153 10 Athens , Greece
| | | | - Jean-Claude Reubi
- Cell Biology and Experimental Cancer Research, Institute of Pathology , University of Berne , CH-3010 Berne , Switzerland
| | - Theodosia Maina
- Molecular Radiopharmacy, INRASTES , National Center for Scientific Research "Demokritos" , GR-153 10 Athens , Greece
| |
Collapse
|
36
|
Moreno P, Mantey SA, Lee SH, Ramos-Álvarez I, Moody TW, Jensen RT. A possible new target in lung-cancer cells: The orphan receptor, bombesin receptor subtype-3. Peptides 2018; 101:213-226. [PMID: 29410320 PMCID: PMC6159918 DOI: 10.1016/j.peptides.2018.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/27/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
Human bombesin receptors, GRPR and NMBR, are two of the most frequently overexpressed G-protein-coupled-receptors by lung-cancers. Recently, GRPR/NMBR are receiving considerable attention because they act as growth factor receptors often in an autocrine manner in different lung-cancers, affect tumor angiogenesis, their inhibition increases the cytotoxic potency of tyrosine-kinase inhibitors reducing lung-cancer cellular resistance/survival and their overexpression can be used for sensitive tumor localization as well as to target cytotoxic agents to the cancer. The orphan BRS-3-receptor, because of homology is classified as a bombesin receptor but has received little attention, despite the fact that it is also reported in a number of studies in lung-cancer cells and has growth effects in these cells. To address its potential importance, in this study, we examined the frequency/relative quantitative expression of human BRS-3 compared to GRPR/NMBR and the effects of its activation on cell-signaling/growth in 13 different human lung-cancer cell-lines. Our results showed that BRS-3 receptor is expressed in 92% of the cell-lines and that it is functional in these cells, because its activation stimulates phospholipase-C with breakdown of phosphoinositides and changes in cytosolic calcium, stimulates ERK/MAPK and stimulates cell growth by EGFR transactivation in some, but not all, the lung-cancer cell-lines. These results suggest that human BRS-3, similar to GRPR/NMBR, is frequently ectopically-expressed by lung-cancer cells in which, it is functional, affecting cell signaling/growth. These results suggest that similar to GRPR/NMBR, BRS-3 should receive increased attention as possible approach for the development of novel treatments and/or diagnosis in lung-cancer.
Collapse
Affiliation(s)
- Paola Moreno
- Department of Health and Human Services, Digestive Diseases Branch, NIDDK, United States
| | - Samuel A Mantey
- Department of Health and Human Services, Digestive Diseases Branch, NIDDK, United States
| | - Suk H Lee
- Department of Health and Human Services, Digestive Diseases Branch, NIDDK, United States
| | - Irene Ramos-Álvarez
- Department of Health and Human Services, Digestive Diseases Branch, NIDDK, United States
| | - Terry W Moody
- Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Robert T Jensen
- Department of Health and Human Services, Digestive Diseases Branch, NIDDK, United States.
| |
Collapse
|
37
|
Ma Z, Zhang Y, Su J, Li X, Yang S, Qiao W, Suo C, Lei Z. Distribution of the pig gastrin-releasing peptide receptor and the effect of GRP on porcine Leydig cells. Peptides 2018; 99:142-152. [PMID: 28966141 DOI: 10.1016/j.peptides.2017.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/23/2017] [Accepted: 09/25/2017] [Indexed: 01/22/2023]
Abstract
Gastrin-releasing peptide (GRP) is a mammalian bombesin (BN)-like peptide which plays a role in a number of important physiological functions via its receptor (gastrin-releasing peptide receptor, GRPR) in most animals. However, little is known about the gene encoding GRPR and its functions (especially reproduction) in pigs. In this study, we first cloned and analyzed the pig GRPR cDNA. Then we systematically investigated the expression levels of GRPR mRNA by relative real-time PCR (RT-PCR), and analyzed the distribution of the GRPR protein in pig tissues via immunohistochemistry (IHC). Finally, we studied the effect of GRP on testosterone secretion and GRPR (mRNA and protein) expression in Leydig cells. Results showed that the pig GRPR cDNA cloned at 1487bp, including one open reading frame (ORF) of 1155bp and encodes 384 amino acids. Significantly, compared with other species, the cDNA sequence and amino acid sequence of the pig GRPR were highly homologous and conservative. The RT-PCR results showed that: in the central nervous system (CNS) and the pituitary, GRPR mRNA was found in the cerebellum, hypophysis, spinal cord and hypothalamus; in the peripheral tissues, GRPR mRNA was mainly expressed in the pancreas, esophagus, ovary, testis, spleen, thymus, jejunum lymph node, muscle and fat. Moreover, the IHC results showed that GRPR immunoreactivity was widely distributed in the pig tissues and organs, such as the pancreas, esophagus, testis, ovary, spleen, pituitary gland and adrenal gland. In addition, we found that GRP promotes testosterone secretion, and increases GRPR mRNA and protein expression in cultured Leydig cells in vitro. These molecular and morphological data not only describe the anatomical locations of GRPR in pigs, but also provide the theoretical foundation for further research into its possible physiological functions in pigs. These results suggest that the GRP/GRPR system may play an important role in regulating the reproductive system of the boar.
Collapse
Affiliation(s)
- Zhiyu Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Ying Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Juan Su
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Xiang Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Sheng Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Wenna Qiao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Chuan Suo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Zhihai Lei
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China.
| |
Collapse
|
38
|
Pendharkar SA, Drury M, Walia M, Korc M, Petrov MS. Gastrin-Releasing Peptide and Glucose Metabolism Following Pancreatitis. Gastroenterology Res 2017; 10:224-234. [PMID: 28912908 PMCID: PMC5593441 DOI: 10.14740/gr890w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Background Gastrin-releasing peptide (GRP) is a pluripotent peptide that has been implicated in both gastrointestinal inflammatory states and classical chronic metabolic diseases such as diabetes. Abnormal glucose metabolism (AGM) after pancreatitis, an exemplar inflammatory disease involving the gastrointestinal tract, is associated with persistent low-grade inflammation and altered secretion of pancreatic and gut hormones as well as cytokines. While GRP is involved in secretion of many of them, it is not known whether GRP has a role in AGM. Therefore, we aimed to investigate the association between GRP and AGM following pancreatitis. Methods Fasting blood samples were collected to measure GRP, blood glucose, insulin, amylin, glucagon, pancreatic polypeptide (PP), somatostatin, cholecystokinin, gastric-inhibitory peptide (GIP), gastrin, ghrelin, glicentin, glucagon-like peptide-1 and 2, oxyntomodulin, peptide YY (PYY), secretin, vasoactive intestinal peptide, tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein (MCP)-1, and interleukin-6. Modified Poisson regression analysis and linear regression analyses were conducted. Four statistical models were used to adjust for demographic, metabolic, and pancreatitis-related risk factors. Results A total of 83 individuals after an episode of pancreatitis were recruited. GRP was significantly associated with AGM, consistently in all four models (P -trend < 0.05), and fasting blood glucose contributed 17% to the variance of GRP. Further, GRP was significantly associated with glucagon (P < 0.003), MCP-1 (P < 0.025), and TNF-α (P < 0.025) - consistently in all four models. GRP was also significantly associated with PP and PYY in three models (P < 0.030 for both), and with GIP and glicentin in one model (P = 0.001 and 0.024, respectively). Associations between GRP and other pancreatic and gut hormones were not significant. Conclusion GRP is significantly increased in patients with AGM after pancreatitis and is associated with increased levels of pro-inflammatory cytokines, as well as certain pancreatic and gut hormones. Detailed mechanistic studies are now warranted to investigate the exact role of GRP in derangements of glucose homeostasis following pancreatitis.
Collapse
Affiliation(s)
| | - Marie Drury
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Monika Walia
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Murray Korc
- Department of Medicine, Biochemistry and Molecular Biology, Division of Endocrinology, Indiana University School of Medicine, the Melvin and Bren Simon Cancer Center and the Pancreatic Cancer Signature Centre, Indianapolis, IN, USA
| | - Maxim S Petrov
- Department of Surgery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Hallasch S, Frick S, Jung M, Hilger I. How gastrin-releasing peptide receptor (GRPR) and α vβ 3 integrin expression reflect reorganization features of tumors after hyperthermia treatments. Sci Rep 2017; 7:6916. [PMID: 28761146 PMCID: PMC5537297 DOI: 10.1038/s41598-017-06100-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/07/2017] [Indexed: 12/19/2022] Open
Abstract
The outcome of tumor treatment via hyperthermia in the clinic has been reported to be heterogeneous. Here, we assessed how the presence of gastrin-releasing peptide receptor (GRPR) and αvβ3 integrin together with the morphology of the vascularization reflects the growth behavior of tumors after hyperthermia treatment. MDA-MB-231 tumor bearing mice were treated either with high (46 °C) or low dose (42 °C) water hyperthermia for 60 min. Changes of GRPR and αvβ3 integrin expression were assessed via multiplexed optical imaging. Vascularization was reconstructed and quantified by µCT imaging after contrast agent injection. We found that high dose hyperthermia is capable of increasing the expression of GRPR, αvβ3 integrin, CD31, and Ki67 in tumors. Also the morphology of tumor vasculature changed (increased relative blood volume and small-diameter vessel density, decreased expression of α-SMA). Low dose hyperthermia induced comparatively moderate effects on the investigated protein expression pattern and vascular remodeling. We conclude that under defined circumstances, specific temperature doses affect the reorganization of tumor regrowth, which is triggered by residual "dormant" cells even though tumor volumes are transiently decreasing. Further on, GRPR, αvβ3 integrin expression are versatile tools to surveil potential tumor regrow during therapy, beyond the conventional determination of tumor volumes.
Collapse
Affiliation(s)
- Sandra Hallasch
- Department of Experimental Radiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Am Klinikum 1, D-07747, Jena, Germany
| | - Sindy Frick
- Department of Experimental Radiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Am Klinikum 1, D-07747, Jena, Germany
| | - Maximilian Jung
- Department of Experimental Radiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Am Klinikum 1, D-07747, Jena, Germany
- Department of Medical Engineering and Biotechnology, University of Applied Science Jena, Carl-Zeiss Promenade 2, 07745, Jena, Germany
| | - Ingrid Hilger
- Department of Experimental Radiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital - Friedrich Schiller University Jena, Am Klinikum 1, D-07747, Jena, Germany.
| |
Collapse
|
40
|
Maina T, Nock BA, Kulkarni H, Singh A, Baum RP. Theranostic Prospects of Gastrin-Releasing Peptide Receptor–Radioantagonists in Oncology. PET Clin 2017; 12:297-309. [PMID: 28576168 DOI: 10.1016/j.cpet.2017.02.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Maina T, Kaloudi A, Valverde IE, Mindt TL, Nock BA. Amide-to-triazole switch vs. in vivo NEP-inhibition approaches to promote radiopeptide targeting of GRPR-positive tumors. Nucl Med Biol 2017. [PMID: 28636973 DOI: 10.1016/j.nucmedbio.2017.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Radiolabeled bombesin (BBN)-analogs have been proposed for diagnosis and therapy of gastrin-releasing peptide receptor (GRPR)-expressing tumors, such as prostate, breast and lung cancer. Metabolic stability represents a crucial factor for the success of this approach by ensuring sufficient delivery of circulating radioligand to tumor sites. The amide-to-triazole switch on the backbone of DOTA-PEG4-[Nle14]BBN(7-14) (1) was reported to improve the in vitro stability of resulting 177Lu-radioligands. On the other hand, in-situ inhibition of neutral endopeptidase (NEP) by coinjection of phosphoramidon (PA) was shown to significantly improve the in vivo stability and tumor uptake of biodegradable radiopeptides. We herein compare the impact of the two methods on the bioavailability and localization of 177Lu-DOTA-PEG4-[Nle14]BBN(7-14) analogs in GRPR-positive tumors in mice. METHODS The 1,4-disubstituted [1-3]-triazole was used to replace one (2: Gly11-His12; 3: Ala9-Val10) or two (4: Ala9-Val10 and Gly11-His12) peptide bonds in 1 (reference) and all compounds were labeled with 177Lu. Each of [177Lu]1-[177Lu]4 was injected without (control) or with PA in healthy mice. Blood samples collected 5min post-injection (pi) were analyzed by HPLC. Biodistribution of [177Lu]1-[177Lu]4 was conducted in SCID mice bearing human prostate adenocarcinoma PC-3 xenografts at 4h pi. Groups of 4 animals were injected with radioligand, alone (controls), or with coinjection of PA, or of a mixture of PA and excess and [Tyr4]BBN to determine GRPR-specificity of uptake (Block). RESULTS The in vivo stability of the radioligands was: [177Lu]1 (25% intact), [177Lu]2 (45% intact), [177Lu]3 (30% intact) and [177Lu]4 (40% intact). By PA-coinjection these values notably increased to 90%-93%. Moreover, treatment with PA induced an impressive and GRPR-specific uptake of all radioligands in the PC-3 xenografts at 4h pi: [177Lu]1: 4.7±0.4 to 24.8±4.9%ID/g; [177Lu]2: 8.3±1.2 to 26.0±1.1%ID/g; [177Lu]3: 6.6±0.4 to 21.3±4.4%ID/g; and [177Lu]4: 4.8±1.6 to 13.7±3.8%ID/g. CONCLUSIONS This study has shown that amide-to-triazole substitutions in 177Lu-DOTA-PEG4-[Nle14]BBN(7-14) induced minor effects on bioavailability and tumor uptake in mice models, whereas in-situ NEP-inhibition(s) by PA impressively improved in vivo profiles.
Collapse
Affiliation(s)
- Theodosia Maina
- Molecular Radiopharmacy, INRASTES, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Aikaterini Kaloudi
- Molecular Radiopharmacy, INRASTES, National Center for Scientific Research "Demokritos", Athens, Greece
| | - Ibai E Valverde
- Institut de Chimie Moléculaire de l'Université de Bourgogne (ICMUB), UMR 6302 CNRS-UBFC, Université de Bourgogne Franche-Comté, Dijon, France
| | - Thomas L Mindt
- Department of Biomedical Imaging and Image Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics, General Hospital of Vienna, Vienna, Austria.
| | - Berthold A Nock
- Molecular Radiopharmacy, INRASTES, National Center for Scientific Research "Demokritos", Athens, Greece.
| |
Collapse
|
42
|
Epigallocatechin gallate protects BEAS-2B cells from lipopolysaccharide-induced apoptosis through upregulation of gastrin-releasing peptide. Mol Cell Biochem 2017; 434:105-111. [DOI: 10.1007/s11010-017-3040-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/12/2017] [Indexed: 01/20/2023]
|
43
|
Jaeger N, Czepielewski RS, Bagatini M, Porto BN, Bonorino C. Neuropeptide gastrin-releasing peptide induces PI3K/reactive oxygen species-dependent migration in lung adenocarcinoma cells. Tumour Biol 2017; 39:1010428317694321. [PMID: 28351312 DOI: 10.1177/1010428317694321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nerve fibers and neurotransmitters have increasingly been shown to have a role in tumor progression. Gastrin-releasing peptide is a neuropeptide linked to tumor aggressiveness, acting as an autocrine tumor growth factor by binding to its receptor, gastrin-releasing peptide receptor, expressed by many tumors. Although neuropeptides have been previously linked to tumor cell proliferation, more recent studies have uncovered roles for neuropeptides in chemotaxis and metastasis. Understanding the precise roles of such peptides in cancer is crucial to optimizing targeted therapy design. We have previously described that gastrin-releasing peptide acts directly as a chemotactic factor for neutrophils, dependent on PI3K, ERK, and p38. In this study, we investigated roles for gastrin-releasing peptide in lung adenocarcinoma. We asked if gastrin-releasing peptide would act as a proliferative and/or chemotactic stimulus for gastrin-releasing peptide receptor-expressing tumor cells. In A549 cells, a non-small cell lung carcinoma line, the treatment with gastrin-releasing peptide leads to activation of AKT and ERK1/2, and production of reactive oxygen species. Gastrin-releasing peptide induced migration of A549 cells, dependent on gastrin-releasing peptide receptor and PI3K, but not ERK. However, no proliferation was observed in these cells in response to gastrin-releasing peptide, and gastrin-releasing peptide did not promote resistance to treatment with a chemotherapy drug. Our results suggest that, similar to what happens in neutrophils, gastrin-releasing peptide is a migratory, rather than a proliferative, stimulus, for non-small cell lung carcinoma cells, indicating a putative role for gastrin-releasing peptide and gastrin-releasing peptide receptor in metastasis.
Collapse
Affiliation(s)
- Natália Jaeger
- 1 Laboratório de Imunologia Celular e Molecular, Hospital São Lucas, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,2 Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rafael Sanguinetti Czepielewski
- 1 Laboratório de Imunologia Celular e Molecular, Hospital São Lucas, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,2 Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Maira Bagatini
- 1 Laboratório de Imunologia Celular e Molecular, Hospital São Lucas, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Bárbara N Porto
- 3 Laboratório de Imunologia Clínica e Experimental, Hospital São Lucas, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Cristina Bonorino
- 1 Laboratório de Imunologia Celular e Molecular, Hospital São Lucas, Instituto de Pesquisas Biomédicas (IPB), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,2 Programa de Pós-Graduação em Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
44
|
Moreno P, Ramos-Álvarez I, Moody TW, Jensen RT. Bombesin related peptides/receptors and their promising therapeutic roles in cancer imaging, targeting and treatment. Expert Opin Ther Targets 2016; 20:1055-1073. [PMID: 26981612 PMCID: PMC5067074 DOI: 10.1517/14728222.2016.1164694] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Despite remarkable advances in tumor treatment, many patients still die from common tumors (breast, prostate, lung, CNS, colon, and pancreas), and thus, new approaches are needed. Many of these tumors synthesize bombesin (Bn)-related peptides and over-express their receptors (BnRs), hence functioning as autocrine-growth-factors. Recent studies support the conclusion that Bn-peptides/BnRs are well-positioned for numerous novel antitumor treatments, including interrupting autocrine-growth and the use of over-expressed receptors for imaging and targeting cytotoxic-compounds, either by direct-coupling or combined with nanoparticle-technology. AREAS COVERED The unique ability of common neoplasms to synthesize, secrete, and show a growth/proliferative/differentiating response due to BnR over-expression, is reviewed, both in general and with regard to the most frequently investigated neoplasms (breast, prostate, lung, and CNS). Particular attention is paid to advances in the recent years. Also considered are the possible therapeutic approaches to the growth/differentiation effect of Bn-peptides, as well as the therapeutic implication of the frequent BnR over-expression for tumor-imaging and/or targeted-delivery. EXPERT OPINION Given that Bn-related-peptides/BnRs are so frequently ectopically-expressed by common tumors, which are often malignant and become refractory to conventional treatments, therapeutic interventions using novel approaches to Bn-peptides and receptors are being explored. Of particular interest is the potential of reproducing with BnRs in common tumors the recent success of utilizing overexpression of somatostatin-receptors by neuroendocrine-tumors to provide the most sensitive imaging methods and targeted delivery of cytotoxic-compounds.
Collapse
Affiliation(s)
- Paola Moreno
- Digestive Diseases Branch, Cell Biology Section, NIDDK, and Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Irene Ramos-Álvarez
- Digestive Diseases Branch, Cell Biology Section, NIDDK, and Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Terry W. Moody
- Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert T. Jensen
- Digestive Diseases Branch, Cell Biology Section, NIDDK, and Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Gastrin-Releasing Peptide Receptor Knockdown Induces Senescence in Glioblastoma Cells. Mol Neurobiol 2016; 54:888-894. [PMID: 26780458 DOI: 10.1007/s12035-016-9696-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/05/2016] [Indexed: 12/19/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of brain tumor, characterized by excessive cell proliferation, resistance to apoptosis, and invasiveness. Due to resistance to currently available treatment options, the prognosis for patients with GBM is very dismal. The activation of gastrin-releasing peptide receptors (GRPR) stimulates GBM cell proliferation, whereas GRPR antagonists induce antiproliferative effects in in vitro and in vivo experimental models of GBM. However, the role of GRPR in regulating other aspects of GBM cell function related to tumor progression remains poorly understood, and previous studies have not used RNA interference techniques as tools to examine GRPR function in GBM. Here, we found that stable GRPR knockdown by a lentiviral vector using a short hairpin interfering RNA sequence in human A172 GBM cells resulted in increased cell size and altered cell cycle dynamics consistent with cell senescence. These changes were accompanied by increases in the content of p53, p21, and p16, activation of epidermal growth factor receptors (EGFR), and a reduction in p38 content. These results increase our understanding of GRPR regulation of GBM cells and further support that GRPR may be a relevant therapeutic target in GBM.
Collapse
|
48
|
Moody TW, Moreno P, Jensen RT. Neuropeptides as lung cancer growth factors. Peptides 2015; 72:106-111. [PMID: 25836991 DOI: 10.1016/j.peptides.2015.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 03/20/2015] [Accepted: 03/20/2015] [Indexed: 12/28/2022]
Abstract
This manuscript is written in honor of the Festschrift for Abba Kastin. I met Abba at a Society for Neuroscience meeting and learned that he was Editor-in-Chief of the Journal Peptides. I submitted manuscripts to the journal on "Neuropeptides as Growth Factors in Cancer" and subsequently was named to the Editorial Advisory Board. Over the past 30 years I have published dozens of manuscripts in Peptides and reviewed hundreds of submitted manuscripts. It was always rewarding to interact with Abba, a consummate professional. When I attended meetings in New Orleans I would sometimes go out to dinner with him at the restaurant "Commanders Palace". When I chaired the Summer Neuropeptide Conference we were honored to have him receive the Fleur Strand Award one year in Israel. I think that his biggest editorial contribution has been the "Handbook of Biologically Active Peptides." I served as a Section Editor on "Cancer/Anticancer Peptides" and again found that it was a pleasure working with him. This review focuses on the mechanisms by which bombesin-like peptides, neurotensin and vasoactive intestinal peptide regulate the growth of lung cancer.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Research, Office of the Director, Bethesda, MD 20892, USA.
| | - Paola Moreno
- National Institute of Diabetes, Digestive, and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| | - Robert T Jensen
- National Institute of Diabetes, Digestive, and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| |
Collapse
|
49
|
Moody TW, Mantey SA, Moreno P, Nakamura T, Lacivita E, Leopoldo M, Jensen RT. ML-18 is a non-peptide bombesin receptor subtype-3 antagonist which inhibits lung cancer growth. Peptides 2015; 64:55-61. [PMID: 25554218 PMCID: PMC4397132 DOI: 10.1016/j.peptides.2014.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 12/28/2022]
Abstract
Bombesin receptor subtype (BRS)-3 is a G protein coupled receptor (GPCR) for the bombesin (BB)-family of peptides. BRS-3 is an orphan GPCR and little is known of its physiological role due to the lack of specific agonists and antagonists. PD168368 is a nonpeptide antagonist for the neuromedin B (NMB) receptor (R) whereas PD176252 is a nonpeptide antagonist for the gastrin releasing peptide (GRP) R and NMBR but not BRS-3. Here nonpeptide analogs of PD176252 e.g. the S-enantiomer ML-18, and the R-enantiomer, EMY-98, were investigated as BRS-3 antagonists using lung cancer cells. ML-18 and EMY-98 inhibited specific (125)I-BA1 (DTyr-Gln-Trp-Ala-Val-βAla-His-Phe-Nle-NH2)BB(6-14) binding to NCI-H1299 lung cancer cells stably transfected with BRS-3 with IC50 values of 4.8 and >100μM, respectively. In contrast, ML-18 bound with lower affinity to the GRPR and NMBR with IC50 values of 16 and >100μM, respectively. ML-18 (16μM), but not its enantiomer EMY-98, inhibited the ability of 10nM BA1 to elevate cytosolic Ca(2+) in a reversible manner using lung cancer cells loaded with FURA2-AM. ML-18 (16μM), but not EMY-98, inhibited the ability of 100nM BA1 to cause tyrosine phosphorylation of the EGFR and ERK in lung cancer cells. ML-18 but not EMY-98 inhibited the proliferation of lung cancer cells. The results indicate that ML-18 is a nonpeptide BRS-3 antagonist that should serve as a template to improve potency and selectivity.
Collapse
Affiliation(s)
- Terry W Moody
- Department of Health and Human Services, National Cancer Institute, Center for Cancer Research, Office of the Director, Bethesda, MD 20892, USA.
| | - Samuel A Mantey
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| | - Paola Moreno
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| | - Taichi Nakamura
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| | - Enza Lacivita
- Dipartimento di Farmacia, Scienze del Farmaco, Universita degli Studi di Bari "A. Moro", Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia, Scienze del Farmaco, Universita degli Studi di Bari "A. Moro", Bari, Italy
| | - Robert T Jensen
- National Institute of Diabetes, Digestive and Kidney Disease, Digestive Diseases Branch, Bethesda, MD 20892, USA
| |
Collapse
|
50
|
Guo M, Qu X, Qin XQ. Bombesin-like peptides and their receptors: recent findings in pharmacology and physiology. Curr Opin Endocrinol Diabetes Obes 2015; 22:3-8. [PMID: 25517020 DOI: 10.1097/med.0000000000000126] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW To highlight the research progress of roles of bombesin-like peptides and their receptors in pharmacology and physiology. RECENT FINDINGS Several new bombesin-derived radioactive or nonradioactive compounds were designed for the diagnosis and therapy of tumors that are overexpressing bombesin receptors. Both gastrin-releasing peptide receptor and neuromedin B receptor activation were shown to induce membrane depolarization and excite neurons in brain. Bombesin receptor subtype-3 was found to be downregulated in the muscle cells and myocytes from obese and type 2 diabetes patients, and its relevant cell signaling events in glucose homeostasis were also investigated. The molecular events triggered by bombesin receptors activation in different types of malignancies is being explored recently and new clues were provided for a better understanding of the biological roles of abnormal expression of bombesin receptors in tumors. Novel cross-talk between gastrin-releasing peptide receptor cell signaling and Sonic hedgehog pathways was identified in small-cell lung carcinoma. SUMMARY Increasing evidence shows bombesin-like peptides and their receptors play important roles in both physiological state and diseases. More specific and safe tumor targeting Bombesin derivatives are being developed for tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Man Guo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | | | | |
Collapse
|