1
|
Medvedeva A, Chernov V, Larkina M, Rybina A, Zelchan R, Bragina O, Varvashenya R, Zebzeeva O, Bezverkhniaia E, Tolmachev V, Orlova A. Single-Photon Emission Computer Tomography Imaging of Prostate-Specific Membrane Antigen (PSMA) Expression in Prostate Cancer Patients Using a Novel Peptide-Based Probe [ 99mTc]Tc-BQ0413 with Picomolar Affinity to PSMA: A Phase I/II Clinical Study. ACS Pharmacol Transl Sci 2025; 8:736-747. [PMID: 40109737 PMCID: PMC11915028 DOI: 10.1021/acsptsci.4c00637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/28/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025]
Abstract
Radionuclide imaging of prostate-specific membrane antigen (PSMA) expression can be used for staging prostate cancer. The pseudo-peptide [99mTc]Tc-BQ0413 demonstrated high affinity and specificity to PSMA in preclinical evaluation. The purpose of this study was to clinically evaluate the safety and tolerability of a single administration of [99mTc]Tc-BQ0413 as well as study its biodistribution using SPECT to estimate dosimetry. [99mTc]Tc-BQ0413 was studied in a single-center diagnostic Phase I open-label exploratory study. Whole-body planar scintigraphy and SPECT/CT imaging were performed 2, 4, and 6 h after administration of 50, 100, or 150 μg (680 ± 140 MBq) of [99mTc]Tc-BQ0413 in five PCa patients per injected mass (NCT05839990). All injections of [99mTc]Tc-BQ0413 were well tolerated. The elimination of [99mTc]Tc-BQ0413 was predominantly renal. The stable physiological uptake of [99mTc]Tc-BQ0413 was observed in the lacrimal and salivary glands, liver, spleen, and kidneys for all tested peptide-injected masses. The average effective doses were 0.007 ± 0.001, 0.0049 ± 0.0003, and 0.0062 ± 0.0008 mSv/MBq for 50, 100, and 150 μg/injection, respectively. The radionuclide-associated dose burden per patient was 4-7 mSv/study for the given activity. Uptake of [99mTc]Tc-BQ0413 in primary tumors was identified in all patients and increased with the peptide-injected mass. Uptake in lymph nodes and bone metastases was the highest at 100 μg/injected mass. The highest tumor lesion/background ratios were observed 6 h after the administration of 100 μg of [99mTc]Tc-BQ0413. The results of the Phase I study showed that injections of [99mTc]Tc-BQ0413 were well tolerated, safe, and associated with low absorbed doses. Imaging using [99mTc]Tc-BQ0413 enabled the visualization of primary prostate cancer lesions as well as metastases in lymph nodes and bones.
Collapse
Affiliation(s)
- Anna Medvedeva
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
| | - Vladimir Chernov
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Anastasiya Rybina
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | - Roman Zelchan
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
| | - Olga Bragina
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
| | - Ruslan Varvashenya
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634009 Tomsk, Russia
- Department of Pharmaceutical Analysis, Siberian State Medical University, 634050 Tomsk, Russia
| | - Olga Zebzeeva
- Department of Nuclear Therapy and Diagnostic, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634009 Tomsk, Russia
| | | | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 752 37 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, 751 23 Uppsala, Sweden
| |
Collapse
|
2
|
Satpati D. Cancer Targeting Radiopeptidomimetics in Molecular Nuclear Medicine. Mol Pharm 2025. [PMID: 40078059 DOI: 10.1021/acs.molpharmaceut.4c01180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Peptides are highly receptor-affine molecules exhibiting suitable pharmacokinetics. Additionally, low-cost production, simple protocols allowing easy modifications, and tolerance toward harsh reaction conditions make peptides ideal ligands for preparation of radiopharmaceuticals for cancer detection and treatment. However, natural peptides being substrates for enzymes are susceptible to proteolysis, which limits the in vivo lifetime and the target uptake. Therefore, the majority of peptides are not able to progress beyond preclinical research. Advancement of peptides for clinical analysis needs modification to instill improved features. Continuous increase and further expected rise in cancer cases in the next decade require development of more disease-directed and promising radiopharmaceuticals. Redesigned peptide, mimicking the original peptide with similar or improved affinity and high metabolic stability, shall have significant edge. This review outlines the design of peptidomimetics by incorporation of D-amino acids (inverso); reversal of D-amino acid sequence (retro-inverso), and reversal of L-amino acid sequence (retro). Clinically successful radiopeptidomimetics prepared using the three approaches have been elaborated to elucidate the important role of peptidomimetics in cancer management.
Collapse
Affiliation(s)
- Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Mumbai 400094, India
| |
Collapse
|
3
|
Lepareur N. An overview of current phase 3 radiopharmaceutical therapy clinical trials. Front Med (Lausanne) 2025; 12:1549676. [PMID: 40041466 PMCID: PMC11876376 DOI: 10.3389/fmed.2025.1549676] [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: 12/21/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Over the past few years, radiopharmaceutical therapy has emerged as a groundbreaking therapeutic modality, taking advantage of the unique properties of radionuclides to deliver molecularly targeted therapy with high precision and transforming the landscape of precision oncology and personalized medicine. Its development reflects decades of advances in nuclear medicine, chemistry, and cancer biology. However, until recently, definitive clinical evidence was lacking to establish it into treatment plans, with few large randomized controlled clinical studies. The last two decades witnessed a paradigm shift, with three successful phase 3 studies which shed light on radiopharmaceutical therapy. This paper offers a brief overview of currently active phase 3 studies to highlight the dynamism and promise of this clinical domain, as well as the large variety of cancers being treated.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer) – UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
4
|
Fonseca AI, Sereno J, Almeida S, Ferreira H, Hrynchak I, Falcão A, Alves F, Gomes C, Abrunhosa AJ. Unveiling the potential of copper-61 vs. gallium-68 for SSTR PET imaging. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07116-2. [PMID: 39909885 DOI: 10.1007/s00259-025-07116-2] [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: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE In recent years, copper-61 has attracted considerable attention from both physicists and radiochemists due to its favorable physical decay properties for PET imaging and its ease of production at any cyclotron center producing [18F]FDG. The aim of this study was to evaluate the potential of 61Cu-based radiopharmaceuticals for PET imaging of NETs, as an alternative to the commonly used gallium-68. METHODS Copper-61 was produced by irradiation of natural zinc liquid targets, followed by post-processing. In vitro evaluation of 61Cu- and 68Ga-labeled SST analogues was performed in SSTR positive AR42J tumor cells. PET/MRI was carried out in mice bearing AR42J subcutaneous tumors. RESULTS High molar activity [61Cu]Cu-DOTA-TATE and [61Cu]Cu-NOTA-TATE were successfully prepared with a radiochemical purity of over 95% and were shown to be stable for at least 6 h after the EOS. Both 61Cu- and 68Ga-labeled SST analogues exhibited high cellular uptake, with residual uptake when blocked with an excessive amount of peptide precursor. [61Cu]Cu-NOTA-TATE showed the highest tumor uptake at 1 h p.i. (13.25 ± 1.86%ID/g) and the tumor-to-non-tumor ratio increased from 1 h to 4 h p.i. At the later time point, tumor visualization improved compared to 1 h p.i. Moreover, preclinical PET/MR images demonstrated that [61Cu]Cu-NOTA-TATE has a more favorable biodistribution and imaging properties than [61Cu]Cu-DOTA-TATE, with the extended PET imaging window providing a clear advantage of [61Cu]Cu-NOTA-TATE over its gallium-68 analogues. CONCLUSION [61Cu]Cu-NOTA-TATE showed similar biodistribution and pharmacokinetics to [68Ga]Ga-DOTA-TATE at 1 h p.i., while demonstrating superior imaging characteristics for late PET imaging. These findings demonstrate that [61Cu]Cu-NOTA-TATE holds promising characteristics for improving the detection of NETs with increased translational potential.
Collapse
Affiliation(s)
- A I Fonseca
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
| | - J Sereno
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
| | - S Almeida
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
| | - H Ferreira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, Portugal
| | - I Hrynchak
- ICNAS Pharma, University of Coimbra, Coimbra, Portugal
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
| | - A Falcão
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - F Alves
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal
- ESTeSC - Coimbra Health School, Instituto Politécnico Coimbra, Coimbra, Portugal
| | - C Gomes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, Coimbra, Portugal
| | - A J Abrunhosa
- CIBIT/ICNAS, Institute for Nuclear Science Applied to Health, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
5
|
Omweri JM, Houson HA, Lynch SE, Tekin V, Sorace AG, Lapi SE. PET imaging of 52Mn labeled DOTATATE and DOTAJR11. Sci Rep 2025; 15:2395. [PMID: 39827143 PMCID: PMC11742957 DOI: 10.1038/s41598-025-85143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025] Open
Abstract
Manganese-52 is gaining interest as an isotope for PET imaging due to its desirable decay and chemical properties for radiopharmaceutical development. Somatostatin receptor 2 (SSTR2) is significantly overexpressed by neuroendocrine tumors (NETs) and is an important target for nuclear imaging and therapy. As an agonist, [68Ga]Ga-DOTATATE has demonstrated significant internalization upon interaction with receptor ligands, whereas [68Ga]Ga-DOTA-JR11(as an antagonist) exhibits limited internalization but better pharmacokinetics and increased tumor uptake. The goal of this study was to label both DOTATATE and DOTA-JR11 peptides with 52Mn in high radiochemical yields (RCY) and sufficient specific activity. A comparison of these two compounds was performed in in vitro and in vivo studies in animals with somatostatin receptor-positive xenografts to characterize differences in cell, tumor, and tissue uptake. Radiolabeling of DOTATATE and DOTA-JR11 was carried out by combining varying concentrations of the peptides with [52Mn]MnCl2. In vitro stability of the radiotracers was determined in mouse serum. In vitro cell uptake and internalization assays were performed in SSTR2 + AR42J cells and negative controls. In vivo biodistribution and longitudinal PET imaging was evaluated in mice bearing AR42J tumors. Both [52Mn]Mn-DOTATATE and [52Mn]Mn-DOTA-JR11 showed affinity for SSTR2 in AR42J cells. However, the uptake of [52Mn]Mn-DOTATATE was higher (11.95 ± 0.71%/ mg) compared to [52Mn]Mn-DOTA-JR11 (7.31 ± 0.38%/ mg) after 2 h incubation. After 4 h incubation, 53.13 ± 1.83% of the total accumulated activity of [52Mn]Mn-DOTATATE was internalized, whereas only 20.85 ± 0.59% of the total accumulated activity of [52Mn]Mn-DOTA-JR11 was internalized. The PET images revealed similar biodistribution results, with [52Mn]Mn-DOTATATE showing a significant tumor uptake of 11.16 ± 2.97% ID/g, while [52Mn]Mn-DOTA-JR11 exhibited a lower tumor uptake of 2.11 ± 0.30% ID/g 4 h post-injection. The synthesis of both radiotracers was accomplished with high RCY and purity. The cell uptake and internalization of [52Mn]Mn-DOTATATE showed higher levels compared to [52Mn]Mn-DOTA-JR11. PET images of the radiotracers in AR42J tumor bearing mice demonstrated similar biodistribution in all organs except the tumor, with [52Mn]Mn-DOTATATE showing higher tumor uptake compared to [52Mn]Mn-DOTA-JR11. The variations in the properties of these tracers could be used to guide further imaging and treatment studies.
Collapse
Affiliation(s)
- James M Omweri
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hailey A Houson
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
| | - Shannon E Lynch
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Volkan Tekin
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, USA.
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
6
|
Botelho L, Dezonne RS, Wildemberg LE, Miranda RL, Gadelha MR, Andreiuolo F. Somatostatin receptors in pituitary somatotroph adenomas as predictors of response to somatostatin receptor ligands: A pathologist's perspective. Brain Pathol 2025; 35:e13313. [PMID: 39473262 DOI: 10.1111/bpa.13313] [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: 07/20/2024] [Accepted: 10/04/2024] [Indexed: 12/28/2024] Open
Abstract
There are five subtypes of somatostatin receptors (SST1-5), which are expressed in several types of solid neoplasms, neuroendocrine tumors, and pituitary adenomas. Most commonly, SST2 and SST5, are of interest regarding diagnostic, treatment, and prognostic purposes. In this article the basic biological characteristics of SST are briefly reviewed, and focus given to the immunohistochemical evaluation of SST2 and SST5 in growth hormone (GH)-secreting pituitary tumors, and their quantification as predictors of response to treatment with somatostatin receptor ligands (SRL), the mainstay of the pharmacological therapy available for these tumors. Although many different scoring systems for SST2 immunohistochemistry showing correlation with SRL response have been reported, among which the immunoreactivity score (IRS) has been the most consistently used, a universally validated immunohistochemical technique and scoring scheme is lacking. Efforts should be made on collaborative multicenter studies aiming at validating homogeneous immunostaining protocols and a scoring system for SST2 and SST5 expression, to help clinicians to define the optimal therapeutic strategy for the patients with somatotroph tumors.
Collapse
Affiliation(s)
- Laura Botelho
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- Department of Pathology, Rede D'Or, Rio de Janeiro, Brazil
| | - Rômulo Sperduto Dezonne
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Luiz Eduardo Wildemberg
- Neuroendocrinology Research Center, Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroendocrinology Division, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Renan Lyra Miranda
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Mônica R Gadelha
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- Neuroendocrinology Research Center, Endocrinology Division, Medical School and Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Neuroendocrinology Division, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Felipe Andreiuolo
- Neuropathology and Molecular Genetics Laboratory, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, Brazil
- Department of Pathology, Rede D'Or, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Guccione C, Gervasoni S, Öztürk I, Bosin A, Ruggerone P, Malloci G. Exploring key features of selectivity in somatostatin receptors through molecular dynamics simulations. Comput Struct Biotechnol J 2024; 23:1311-1319. [PMID: 39660216 PMCID: PMC11630666 DOI: 10.1016/j.csbj.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 12/12/2024] Open
Abstract
Somatostatin receptors (SSTRs) are widely distributed throughout the human body and play crucial roles in various physiological processes. They are recognized as key targets for both radiotherapy and radiodiagnosis due to their overexpression in several cancer types. However, the discovery and design of selective drugs for each of the five isoforms have been significantly hindered by the lack of complete structural information. In this study, we conducted a systematic computational analysis of all five SSTRs in complex with the endogenous ligand somatostatin to elucidate their structural and dynamic features. We thoroughly characterized each isoform using available experimental structures for SSTR2 and SSTR4, as well as AlphaFold2 models for SSTR1, SSTR3, and SSTR5. By performing multi-copy μs-long molecular dynamics simulations, we examined the differences and similarities in dynamical behavior and somatostatin binding among all SSTRs. Our analysis focused on understanding the opening and closing movements of the extracellular loop 2, which are crucial for ligand binding and recognition. Interestingly, we observed a unique conformation of somatostatin within the binding pocket of SSTR5 in which the loop can partially close, as compared to the other isoforms. Fingerprint analyses provided distinct interaction patterns of somatostatin with all receptors, thus enabling precise guidelines for the discovery and development of more selective somatostatin-based pharmaceuticals tailored for precision medicine therapies.
Collapse
Affiliation(s)
- C. Guccione
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| | - S. Gervasoni
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| | - I. Öztürk
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| | - A. Bosin
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| | - P. Ruggerone
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| | - G. Malloci
- Department of Physics, University of Cagliari, Monserrato (Cagliari), 09042, Italy
| |
Collapse
|
8
|
Wang C, Shen Z, Chen Y, Wang Y, Zhou X, Chen X, Li Y, Zhang P, Zhang Q. Research Progress on Cyclic-Peptide Functionalized Nanoparticles for Tumor-Penetrating Delivery. Int J Nanomedicine 2024; 19:12633-12652. [PMID: 39624118 PMCID: PMC11609414 DOI: 10.2147/ijn.s487303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/14/2024] [Indexed: 01/03/2025] Open
Abstract
A key challenge in cancer treatment is the effective delivery of drugs into deep regions of tumor tissues, which are impermeable due to abnormal vascular network, increased interstitial fluid pressure (IFP), abundant extra cellular matrix (ECM), and heterogeneity of tumor cells. Cyclic peptides have been used for the surface engineering of nanoparticles to enhance the tumor-penetrating efficacy of drugs. Compared with other surface ligands, cyclic peptides are more easily produced by automated chemical synthesis, and they are featured by their higher binding affinity with their targets, tumor selectivity, stability against degradation, and low toxicity. In this review, different types of cyclic peptides, their physicochemical properties and their in vivo pharmacokinetics are introduced. Next, the progress of cyclic peptide-functionalized drug delivery nanodevices is updated, and the mechanism underlying the tumor-penetrating properties of cyclic peptide-functionalized drug delivery nanodevices is discussed.
Collapse
Affiliation(s)
- Chenkai Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Zefan Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yiyang Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yifan Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xuanyi Zhou
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xinyi Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yuhang Li
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Pu Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Qi Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| |
Collapse
|
9
|
Li YG, Meng XY, Yang X, Ling SL, Shi P, Tian CL, Yang F. Structural insights into somatostatin receptor 5 bound with cyclic peptides. Acta Pharmacol Sin 2024; 45:2432-2440. [PMID: 38926478 PMCID: PMC11489758 DOI: 10.1038/s41401-024-01314-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024]
Abstract
Somatostatin receptor 5 (SSTR5) is highly expressed in ACTH-secreting pituitary adenomas and is an important drug target for the treatment of Cushing's disease. Two cyclic SST analog peptides (pasireotide and octreotide) both can activate SSTR5 and SSTR2. Pasireotide is preferential binding to SSTR5 than octreotide, while octreotide is biased to SSTR2 than SSTR5. The lack of selectivity of both pasireotide and octreotide causes side effects, such as hyperglycemia, gastrointestinal disturbance, and abnormal glucose homeostasis. However, little is known about the binding and selectivity mechanisms of pasireotide and octreotide with SSTR5, limiting the development of subtype-selective SST analog drugs specifically targeting SSTR5. Here, we report two cryo-electron microscopy (cryo-EM) structures of SSTR5-Gi complexes activated by pasireotide and octreoitde at resolutions of 3.09 Å and 3.24 Å, respectively. In combination with structural analysis and functional experiments, our results reveal the molecular mechanisms of ligand recognition and receptor activation. We also demonstrate that pasireotide preferentially binds to SSTR5 through the interactions between Tyr(Bzl)/DTrp of pasireotide and SSTR5. Moreover, we find that the Q2.63, N6.55, F7.35 and ECL2 of SSTR2 play a crucial role in octreotide biased binding of SSTR2. Our results will provide structural insights and offer new opportunities for the drug discovery of better selective pharmaceuticals targeting specific SSTR subtypes.
Collapse
Affiliation(s)
- Ying-Ge Li
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Xian-Yu Meng
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Xiru Yang
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Sheng-Long Ling
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Pan Shi
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China.
| | - Chang-Lin Tian
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China.
- The Anhui Provincial Key Laboratory of High Magnetic Resonance Image, High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Fan Yang
- The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, Joint Center for Biological Analytical Chemistry, Anhui Engineering Laboratory of Peptide Drug, Anhui Laboratory of Advanced Photonic Science and Technology, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
10
|
Shah A, Dabhade A, Bharadia H, Parekh PS, Yadav MR, Chorawala MR. Navigating the landscape of theranostics in nuclear medicine: current practice and future prospects. Z NATURFORSCH C 2024; 79:235-266. [PMID: 38807355 DOI: 10.1515/znc-2024-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024]
Abstract
Theranostics refers to the combination of diagnostic biomarkers with therapeutic agents that share a specific target expressed by diseased cells and tissues. Nuclear medicine is an exciting component explored for its applicability in theranostic concepts in clinical and research investigations. Nuclear theranostics is based on the employment of radioactive compounds delivering ionizing radiation to diagnose and manage certain diseases employing binding with specifically expressed targets. In the realm of personalized medicine, nuclear theranostics stands as a beacon of potential, potentially revolutionizing disease management. Studies exploring the theranostic profile of radioactive compounds have been presented in this review along with a detailed explanation of radioactive compounds and their theranostic applicability in several diseases. It furnishes insights into their applicability across diverse diseases, elucidating the intricate interplay between these compounds and disease pathologies. Light is shed on the important milestones of nuclear theranostics beginning with radioiodine therapy in thyroid carcinomas, MIBG labelled with iodine in neuroblastoma, and several others. Our perspectives have been put forth regarding the most important theranostic agents along with emerging trends and prospects.
Collapse
Affiliation(s)
- Aayushi Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Akshada Dabhade
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Hetvi Bharadia
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Mayur R Yadav
- Department of Pharmacy Practice and Administration, Western University of Health Science, 309 E Second St, Pomona, CA, 91766, USA
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
11
|
Brookes A, Kindon N, Scurr DJ, Alexander MR, Gershkovich P, Bradshaw TD. Cannabidiol and fluorinated derivative anti-cancer properties against glioblastoma multiforme cell lines, and synergy with imidazotetrazine agents. BJC REPORTS 2024; 2:67. [PMID: 39516685 PMCID: PMC11524125 DOI: 10.1038/s44276-024-00088-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive cancer with poor prognosis, partly due to resistance to the standard chemotherapy treatment, temozolomide (TMZ). Phytocannabinoid cannabidiol (CBD) has exhibited anti-cancer effects against GBM, however, CBD's ability to overcome common resistance mechanisms to TMZ have not yet been investigated. 4'-Fluoro-cannabidiol (4'-F-CBD, or HUF-101/PECS-101) is a derivative of CBD, that exhibits increased activity compared to CBD during in vivo behavioural studies. METHODS This anti-cancer activity of cannabinoids against GBM cells sensitive to and representing major resistance mechanisms to TMZ was investigated. Cannabinoids were also studied in combination with imidazotetrazine agents, and advanced mass spectrometry with the 3D OrbiSIMS was used to investigate the mechanism of action of CBD. RESULTS CBD and 4'-F-CBD were found to overcome two major resistance mechanisms (methylguanine DNA-methyltransferase (MGMT) overexpression and DNA mismatch repair (MMR)-deficiency). Synergistic responses were observed when cells were exposed to cannabinoids and imidazotetrazine agents. Synergy increased with T25 and 4'-F-CBD. 3D OrbiSIMS analysis highlighted the presence of methylated-DNA, a previously unknown anti-cancer mechanism of action of CBD. CONCLUSIONS This work demonstrates the anti-cancer activity of 4'-F-CBD and the synergy of cannabinoids with imidazotetrazine agents for the first time and expands understanding of CBD mechanism of action.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Nicholas Kindon
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - David J Scurr
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | | | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK.
| |
Collapse
|
12
|
Kunstman JW, Nagar A, Gibson J, Kunz PL. Modern Management of Gastric Neuroendocrine Neoplasms. Curr Treat Options Oncol 2024; 25:1137-1152. [PMID: 39083164 DOI: 10.1007/s11864-024-01207-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 09/22/2024]
Abstract
OPINION STATEMENT Gastric neuroendocrine neoplasms (G-NENs) are a heterogeneous group of tumors that broadly fall into two groups. The first group, driven by oversecretion of gastrin, are generally multifocal, small, and behave indolently with a low (but non-zero) risk of progression and metastatic spread. They are conventionally categorized into type 1, with endogenous gastric-based overproduction of gastrin, and type 2 G-NEN, with overproduction of gastrin from an extra-gastric gastrin-secreting tumor. The second group, termed type 3 G-NEN, occur spontaneously and are potentially more aggressive, having a clinical course analogous to other neuroendocrine tumors of the gastrointestinal tract. Type 1 G-NEN can be managed with endoscopic surveillance and resection of visible lesions with great success, reserving surgery for the rare high-risk lesion, whereas surgical resection of the causative gastrin-secreting tumor in type 2 G-NEN is usually curative. Type 3 G-NEN is usually managed with formal surgical resection but there is growing evidence that limited surgery or even endoscopic resection in appropriately selected patients with low risk is both safe and effective. A novel subtype of G-NEN, associated with long-term proton pump inhibitor usage, is increasing in incidence. The pathophysiology seems to parallel type 1 G-NEN. In the setting of metastatic disease, which can occur in any subtype but is most common by far in type 3 G-NEN, the lack of trial data unique to G-NEN results in extrapolation of strategies and agents for treatment of non-gastric neuroendocrine disease. The rapid pace of development in this area is likely to benefit the metastatic G-NEN patient as well. As treatment is predicate on type of G-NEN, establishing the etiology of the lesion is crucial but growing knowledge of G-NEN pathophysiology and close collaboration between pathologists, gastroenterologists, radiologists, surgeons, and oncologists have enabled a growing trend towards de-escalation and less-invasive treatment paradigms.
Collapse
Affiliation(s)
- John W Kunstman
- Department of Surgery, Division of Surgical Oncology, Yale School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Anil Nagar
- Department of Medicine, Division of Gastroenterology, Yale School of Medicine, New Haven, CT, USA
| | - Joanna Gibson
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Pamela L Kunz
- Department of Medicine, Section of Medical Oncology, Yale School of Medicine, 25 York Street, New Haven, CT, 06510, USA.
| |
Collapse
|
13
|
Omweri JM, Houson HA, Lynch SE, Tekin V, Sorace AG, Lapi SE. PET imaging of [52 Mn]Mn-DOTATATE and [52 Mn]Mn-DOTA-JR11. RESEARCH SQUARE 2024:rs.3.rs-4684098. [PMID: 39149492 PMCID: PMC11326411 DOI: 10.21203/rs.3.rs-4684098/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Manganese-52 is gaining interest as an isotope for PET imaging due to its desirable decay and chemical properties for radiopharmaceutical development. Somatostatin receptor 2 (SSTR2) is significantly overexpressed by neuroendocrine tumors (NETs) and is an important target for nuclear imaging and therapy. As an agonist, [68Ga]Ga-DOTATATE has demonstrated significant internalization upon interaction with receptor ligands, whereas [68Ga]Ga-DOTA-JR11(as an antagonist) exhibits limited internalization but better pharmacokinetics and increased tumor uptake. The goal of this study was to label both DOTATATE and DOTA-JR11 peptides with 52Mn in high radiochemical yields (RCY) and sufficient specific activity. A comparison of these two compounds was performed in in vitro and in vivo studies in animals with somatostatin receptor-positive xenografts to characterize differences in cell, tumor, and tissue uptake. Radiolabeling of DOTATATE and DOTA-JR11 was carried out by combining varying concentrations of the peptides with [52Mn]MnCl2. In vitro stability of the radiotracers was determined in mouse serum. In vitro cell uptake and internalization assays were performed in SSTR2 + AR42J cells and negative controls. In vivo biodistribution and longitudinal PET imaging was evaluated in mice bearing AR42J tumors. Both [52Mn]Mn-DOTATATE and [52Mn]Mn-DOTA-JR11showed affinity for SSTR2 in AR42J cells. However, the uptake of [52Mn]Mn-DOTATATE was higher (11.95 ± 0.71%/ mg) compared to [52Mn]Mn-DOTA-JR11 (7.31 ± 0.38%/ mg) after 2 h incubation. After 4 h incubation, 53.13 ± 1.83% of the total activity of [52Mn]Mn-DOTATATE was internalized, whereas only 20.85 ± 0.59% of the total activity of [52Mn]Mn-DOTA-JR11 was internalized. The PET images revealed similar biodistribution results, with [52Mn]Mn-DOTATATE showing a significant tumor uptake of 11.16 ± 2.97% ID/g, while [52Mn]Mn-DOTA-JR11 exhibited a lower tumor uptake of 2.11 ± 0.30% ID/g 4 h post-injection. The synthesis of both radiotracers was accomplished with high RCY and purity. The cell uptake and internalization of [52Mn]Mn-DOTATATE showed higher levels compared to [52Mn]Mn-DOTA-JR11. PET images of the radiotracers in AR42J tumor bearing mice demonstrated similar biodistribution in all organs except the tumor, with [52Mn]Mn-DOTATATE showing higher tumor uptake compared to [52Mn]Mn-DOTA-JR11. The variations in properties of these tracers could be used to guide further imaging and treatment studies.
Collapse
|
14
|
Han SA, Ryu J, Song SW, Kim JS, Ryu JS, Oh M. 68Ga-DOATATOC Brain PET/CT Imaging in a case of Dural Metastasis from Synovial Sarcoma. Nucl Med Mol Imaging 2024; 58:310-316. [PMID: 39036463 PMCID: PMC11255146 DOI: 10.1007/s13139-024-00854-5] [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: 08/04/2023] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 07/23/2024] Open
Abstract
We present the case of a 70-year-old male patient who underwent a gallium-68 (68Ga)-DOTATOC brain positron emission tomography (PET)/computed tomography (CT) for the assessment of a tumorous lesion on the dura. The patient had previously undergone below-knee amputation due to a mass of synovial sarcoma on the left foot and completed adjuvant chemotherapy approximately 3 months ago. Subsequently, a well-demarcated papillary solid mass located on the dura was surgically excised. Pathological examination confirmed that the dural metastasis originated from synovial sarcoma and post-operative magnetic resonance imaging (MRI) revealed no residual tumor. We conducted a 68Ga-DOTATOC brain PET/CT suspecting a meningioma given the presence of a dural mass. The result showed lower uptake (maximum standardized uptake [SUVmax 4.9]) than the pituitary gland (SUVmax 9.3). Thus, we successfully conducted a differential diagnosis of metastasis from the preexisting malignancy rather than the meningioma. 68Ga-DOTATOC PET/CT is a valuable tool for the differential diagnosis of meningioma. However, metastasis should also be considered, especially in patients with a history of malignancy and lesions showing mild 68Ga-DOTATOC uptake.
Collapse
Affiliation(s)
- Shin Ae Han
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Jeongryul Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Sang Woo Song
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae-Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Jin-Sook Ryu
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505 Republic of Korea
| |
Collapse
|
15
|
Pfeiffer IPM, Schröder MP, Mordhorst S. Opportunities and challenges of RiPP-based therapeutics. Nat Prod Rep 2024; 41:990-1019. [PMID: 38411278 DOI: 10.1039/d3np00057e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Covering: up to 2024Ribosomally synthesised and post-translationally modified peptides (RiPPs) comprise a substantial group of peptide natural products exhibiting noteworthy bioactivities ranging from antiinfective to anticancer and analgesic effects. Furthermore, RiPP biosynthetic pathways represent promising production routes for complex peptide drugs, and the RiPP technology is well-suited for peptide engineering to produce derivatives with specific functions. Thus, RiPP natural products possess features that render them potentially ideal candidates for drug discovery and development. Nonetheless, only a small number of RiPP-derived compounds have successfully reached the market thus far. This review initially outlines the therapeutic opportunities that RiPP-based compounds can offer, whilst subsequently discussing the limitations that require resolution in order to fully exploit the potential of RiPPs towards the development of innovative drugs.
Collapse
Affiliation(s)
- Isabel P-M Pfeiffer
- University of Tübingen, Pharmaceutical Institute, Department of Pharmaceutical Biology, Auf der Morgenstelle 8, 72076 Tübingen, Germany.
| | - Maria-Paula Schröder
- University of Tübingen, Pharmaceutical Institute, Department of Pharmaceutical Biology, Auf der Morgenstelle 8, 72076 Tübingen, Germany.
| | - Silja Mordhorst
- University of Tübingen, Pharmaceutical Institute, Department of Pharmaceutical Biology, Auf der Morgenstelle 8, 72076 Tübingen, Germany.
| |
Collapse
|
16
|
Hoogenkamp DS, de Wit-van der Veen LJ, Huizing DMV, Tesselaar MET, van Leeuwaarde RS, Stokkel MPM, Lam MGEH, Braat AJAT. Advances in Radionuclide Therapies for Patients with Neuro-endocrine Tumors. Curr Oncol Rep 2024; 26:551-561. [PMID: 38598035 PMCID: PMC11062977 DOI: 10.1007/s11912-024-01521-w] [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] [Accepted: 03/21/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE OF REVIEW To provide insights into the role of peptide receptor radionuclide therapy (PRRT) in patients with advanced neuroendocrine tumors (NET) and an overview of possible strategies to combine PRRT with locoregional and systemic anticancer treatments. RECENT FINDINGS Research on combining PRRT with other treatments encompasses a wide variety or treatments, both local (transarterial radioembolization) and systemic therapies, chemotherapy (i.e., capecitabine and temozolomide), targeted therapies (i.e., olaparib, everolimus, and sunitinib), and immunotherapies (e.g., nivolumab and pembrolizumab). Furthermore, PRRT shows promising first results as a treatment prior to surgery. There is great demand to enhance the efficacy of PRRT through combination with other anticancer treatments. While research in this area is currently limited, the field is rapidly evolving with numerous ongoing clinical trials aiming to address this need and explore novel therapeutic combinations.
Collapse
Affiliation(s)
- Denise S Hoogenkamp
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Linda J de Wit-van der Veen
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Daphne M V Huizing
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Margot E T Tesselaar
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachel S van Leeuwaarde
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Endocrinology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
| | - Marnix G E H Lam
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Arthur J A T Braat
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- ENETS Center of Excellence NKI-AVL, The Netherlands Cancer Institute/UMC Utrecht, Amsterdam, The Netherlands.
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Sugiharti RJ, Maharani R, Kurniawan F, Kartasasmita RE, Tjahjono DH. Computational studies and synthesis of 131iodine-labeled nocardiotide A analogs as a peptide-based theragnostic radiopharmaceutical ligand for cancer targeting SSTR2. RSC Adv 2024; 14:10962-10968. [PMID: 38577429 PMCID: PMC10993231 DOI: 10.1039/d4ra00684d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/22/2024] [Indexed: 04/06/2024] Open
Abstract
Radiolabeled peptides belong to a highly specific group of radiotracers used in oncology, particularly for diagnostics and cancer therapy. With the notable advantages of high binding affinity and selectivity to cancer cells, they have proven to be very useful in nuclear medicine. As a result, efforts have been focused on discovering new peptide sequences for radiopeptide preparation. Nocardiotide A, a cyclic hexapeptide comprising the amino acids cyclo-Trp-Ile-Trp-Leu-Val-Ala (cWIWLVA) isolated from Nocardiopsis sp., has shown significant cytotoxicity against cancer cells, rendering it a suitable candidate for the process. Therefore, the present study aimed to design a stable and effective radiopeptide by labeling nocardiotide A with iodine-131 (131I), ensuring that its affinity to SSTR2 is not compromised. In silico study showed that structural modification of nocardiotide A labeled with 131iodine exhibited good affinity value, forming hydrogen bonds with key residues, such as Q.102 and T.194, which are essential in SSTR2. Based on the results, cyclic hexapeptides of cWIWLYA were selected for further synthesis, and its peptide product was confirmed by the presence of an ionic molecule peak m/z [M + Na]+ 855.4332 (yield, 25.60%). In vitro tests conducted on cWIWLYA showed that cWIWLYA can bind to HeLa cancer cells. Radiopeptide synthesis was initiated with radiolabeling of cWIWLYA by 131I using the chloramine-T method that showed a radiochemical yield of 93.37%. Non-radioactive iodine labeling reaction showed that iodination was successful, which detected the presence of di-iodinated peptide (I2-cWIWLYA) with m/z [M + Na]+ 1107.1138. In summary, a radiopeptide derived from nocardiotide A showed great potential for further development as a diagnostic and therapeutic agent in cancer treatment.
Collapse
Affiliation(s)
- Rizky Juwita Sugiharti
- School of Pharmacy, Bandung Institute of Technology Bandung Indonesia
- Research Center for Radioisotope, Radiopharmaceutical, and Biodosimetry Technology, National Research and Innovation Agency Indonesia
| | - Rani Maharani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran Jatinangor Indonesia
| | | | | | | |
Collapse
|
18
|
Gape PMD, Schultz MK, Stasiuk GJ, Terry SYA. Towards Effective Targeted Alpha Therapy for Neuroendocrine Tumours: A Review. Pharmaceuticals (Basel) 2024; 17:334. [PMID: 38543120 PMCID: PMC10974115 DOI: 10.3390/ph17030334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
This review article explores the evolving landscape of Molecular Radiotherapy (MRT), emphasizing Peptide Receptor Radionuclide Therapy (PRRT) for neuroendocrine tumours (NETs). The primary focus is on the transition from β-emitting radiopharmaceuticals to α-emitting agents in PRRT, offering a critical analysis of the radiobiological basis, clinical applications, and ongoing developments in Targeted Alpha Therapy (TAT). Through an extensive literature review, the article delves into the mechanisms and effectiveness of PRRT in targeting somatostatin subtype 2 receptors, highlighting both its successes and limitations. The discussion extends to the emerging paradigm of TAT, underlining its higher potency and specificity with α-particle emissions, which promise enhanced therapeutic efficacy and reduced toxicity. The review critically evaluates preclinical and clinical data, emphasizing the need for standardised dosimetry and a deeper understanding of the dose-response relationship in TAT. The review concludes by underscoring the significant potential of TAT in treating SSTR2-overexpressing cancers, especially in patients refractory to β-PRRT, while also acknowledging the current challenges and the necessity for further research to optimize treatment protocols.
Collapse
Affiliation(s)
- Paul M. D. Gape
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Michael K. Schultz
- Departments of Radiology, Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA;
- Perspective Therapeutics, Coralville, IA 52241, USA
| | - Graeme J. Stasiuk
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Samantha Y. A. Terry
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| |
Collapse
|
19
|
Zhang T, Lei H, Chen X, Dou Z, Yu B, Su W, Wang W, Jin X, Katsube T, Wang B, Zhang H, Li Q, Di C. Carrier systems of radiopharmaceuticals and the application in cancer therapy. Cell Death Discov 2024; 10:16. [PMID: 38195680 PMCID: PMC10776600 DOI: 10.1038/s41420-023-01778-3] [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: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/11/2024] Open
Abstract
Radiopharmaceuticals play a vital role in cancer therapy. The carrier of radiopharmaceuticals can precisely locate and guide radionuclides to the target, where radionuclides kill surrounding tumor cells. Effective application of radiopharmaceuticals depends on the selection of an appropriate carrier. Herein, different types of carriers of radiopharmaceuticals and the characteristics are briefly described. Subsequently, we review radiolabeled monoclonal antibodies (mAbs) and their derivatives, and novel strategies of radiolabeled mAbs and their derivatives in the treatment of lymphoma and colorectal cancer. Furthermore, this review outlines radiolabeled peptides, and novel strategies of radiolabeled peptides in the treatment of neuroendocrine neoplasms, prostate cancer, and gliomas. The emphasis is given to heterodimers, bicyclic peptides, and peptide-modified nanoparticles. Last, the latest developments and applications of radiolabeled nucleic acids and small molecules in cancer therapy are discussed. Thus, this review will contribute to a better understanding of the carrier of radiopharmaceuticals and the application in cancer therapy.
Collapse
Affiliation(s)
- Taotao Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Huiwen Lei
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Xiaohua Chen
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Zhihui Dou
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Boyi Yu
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Su
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China
| | - Wei Wang
- College of Life Science, Northwest Normal University, Lanzhou, 730000, China
| | - Xiaodong Jin
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Hong Zhang
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Qiang Li
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Cuixia Di
- Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 101408, Beijing, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, 101408, Beijing, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| |
Collapse
|
20
|
Singh NK, Hage N, Ramamourthy B, Nagaraju S, Kappagantu KM. Nuclear Imaging Modalities in the Diagnosis and Management of Thyroid Cancer. Curr Mol Med 2024; 24:1091-1096. [PMID: 37724677 DOI: 10.2174/1566524023666230915103723] [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: 05/17/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023]
Abstract
In this review we have brought forward various nuclear imaging modalities used in the diagnosis, staging, and management of thyroid cancer. Thyroid cancer is the most common endocrine malignancy, accounting for approximately 3% of all new cancer diagnoses. Nuclear imaging plays an important role in the evaluation of thyroid cancer, and the use of radioiodine imaging, FDG imaging, and somatostatin receptor imaging are all valuable tools in the management of this disease. Radioiodine imaging involves the use of Iodine-123 [I-123] or Iodine-131 [I-131] to evaluate thyroid function and detect thyroid cancer. I-123 is a gamma-emitting isotope that is used in thyroid imaging to evaluate thyroid function and detect thyroid nodules. I-131 is a beta-emitting isotope that is used for the treatment of thyroid cancer. Radioiodine imaging is used to detect the presence of thyroid nodules and evaluate thyroid function. FDG imaging is a PET imaging modality that is used to evaluate the metabolic activity of thyroid cancer cells. FDG is a glucose analogue that is taken up by cells that are metabolically active, such as cancer cells. FDG PET/CT can detect primary thyroid cancer and metastatic disease, including lymph nodes and distant metastases. FDG PET/CT is also used to monitor treatment response and detect the recurrence of thyroid cancer. Somatostatin receptor imaging involves the use of radiolabeled somatostatin analogues to detect neuroendocrine tumors, including thyroid cancer. Radiolabeled somatostatin analogues, such as Indium-111 octreotide or Gallium-68 DOTATATE, are administered to the patient, and a gamma camera is used to detect areas of uptake. Somatostatin receptor imaging is highly sensitive and specific for the detection of metastatic thyroid cancer. A comprehensive search of relevant literature was done using online databases of PubMed, Embase, and Cochrane Library using the keywords "thyroid cancer," "nuclear imaging," "radioiodine imaging," "FDG PET/CT," and "somatostatin receptor imaging" to identify relevant studies to be included in this review. Nuclear imaging plays an important role in the diagnosis, staging, and management of thyroid cancer. The use of radioiodine imaging, thyroglobulin imaging, FDG imaging, and somatostatin receptor imaging are all valuable tools in the evaluation of thyroid cancer. With further research and development, nuclear imaging techniques have the potential to improve the diagnosis and management of thyroid cancer and other endocrine malignancies.
Collapse
Affiliation(s)
- Namit Kant Singh
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Neemu Hage
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Balaji Ramamourthy
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Sushmitha Nagaraju
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Krishna Medha Kappagantu
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| |
Collapse
|
21
|
Milewska-Kranc A, Ćwikła JB, Kolasinska-Ćwikła A. The Role of Receptor-Ligand Interaction in Somatostatin Signaling Pathways: Implications for Neuroendocrine Tumors. Cancers (Basel) 2023; 16:116. [PMID: 38201544 PMCID: PMC10778465 DOI: 10.3390/cancers16010116] [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: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Neuroendocrine tumors (NETs) arise from neuroendocrine cells and manifest in diverse organs. Key players in their regulation are somatostatin and its receptors (SSTR1-SSTR5). Understanding receptor-ligand interactions and signaling pathways is vital for elucidating their role in tumor development and therapeutic potential. This review highlights SSTR characteristics, localization, and expression in tissues, impacting physiological functions. Mechanisms of somatostatin and synthetic analogue binding to SSTRs, their selectivity, and their affinity were analyzed. Upon activation, somatostatin initiates intricate intracellular signaling, involving cAMP, PLC, and MAP kinases and influencing growth, differentiation, survival, and hormone secretion in NETs. This review explores SSTR expression in different tumor types, examining receptor activation effects on cancer cells. SSTRs' significance as therapeutic targets is discussed. Additionally, somatostatin and analogues' role in hormone secretion regulation, tumor growth, and survival is emphasized, presenting relevant therapeutic examples. In conclusion, this review advances the knowledge of receptor-ligand interactions and signaling pathways in somatostatin receptors, with potential for improved neuroendocrine tumor treatments.
Collapse
Affiliation(s)
| | - Jarosław B. Ćwikła
- School of Medicine, University of Warmia and Mazury, Aleja Warszawska 30, 10-082 Olsztyn, Poland
- Diagnostic Therapeutic Center–Gammed, Lelechowska 5, 02-351 Warsaw, Poland
| | | |
Collapse
|
22
|
Wild D, Grønbæk H, Navalkissoor S, Haug A, Nicolas GP, Pais B, Ansquer C, Beauregard JM, McEwan A, Lassmann M, Pennestri D, Volteau M, Lenzo NP, Hicks RJ. A phase I/II study of the safety and efficacy of [ 177Lu]Lu-satoreotide tetraxetan in advanced somatostatin receptor-positive neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2023; 51:183-195. [PMID: 37721581 PMCID: PMC10684626 DOI: 10.1007/s00259-023-06383-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/02/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE We present the results of an open-label, phase I/II study evaluating the safety and efficacy of the novel somatostatin receptor (SSTR) antagonist [177Lu]Lu-satoreotide tetraxetan in 40 patients with previously treated, progressive neuroendocrine tumours (NETs), in which dosimetry was used to guide maximum administered activity. METHODS This study was conducted in two parts. Part A consisted of 15 patients who completed three cycles of [177Lu]Lu-satoreotide tetraxetan at a fixed administered activity and peptide amount per cycle (4.5 GBq/300 µg). Part B, which included 25 patients who received one to five cycles of [177Lu]Lu-satoreotide tetraxetan, evaluated different administered activities (4.5 or 6.0 GBq/cycle) and peptide amounts (300, 700, or 1300 μg/cycle), limited to a cumulative absorbed radiation dose of 23 Gy to the kidneys and 1.5 Gy to the bone marrow. RESULTS Median cumulative administered activity of [177Lu]Lu-satoreotide tetraxetan was 13.0 GBq over three cycles (13.1 GBq in part A and 12.9 GBq in part B). Overall, 17 (42.5%) patients experienced grade ≥ 3 treatment‑related adverse events; the most common were lymphopenia, thrombocytopenia, and neutropenia. No grade 3/4 nephrotoxicity was observed. Two patients developed myeloid neoplasms considered treatment related by the investigator. Disease control rate for part A and part B was 94.7% (95% confidence interval [CI]: 82.3-99.4), and overall response rate was 21.1% (95% CI: 9.6-37.3). CONCLUSION [177Lu]Lu-satoreotide tetraxetan, administered at a median cumulative activity of 13.0 GBq over three cycles, has an acceptable safety profile with a promising clinical response in patients with progressive, SSTR-positive NETs. A 5-year long-term follow-up study is ongoing. TRIAL REGISTRATION ClinicalTrials.gov, NCT02592707. Registered October 30, 2015.
Collapse
Affiliation(s)
- Damian Wild
- Division of Nuclear Medicine, ENETS Centre of Excellence, University Hospital Basel, Basel, Switzerland.
| | - Henning Grønbæk
- Department of Hepatology & Gastroenterology, ENETS Centre of Excellence, Aarhus University Hospital and Clinical Institute, Aarhus University, Aarhus, Denmark
| | - Shaunak Navalkissoor
- Neuroendocrine Tumour Unit, ENETS Centre of Excellence, Royal Free London NHS Foundation Trust, London, UK
| | - Alexander Haug
- Department of Radiology and Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Guillaume P Nicolas
- Division of Nuclear Medicine, ENETS Centre of Excellence, University Hospital Basel, Basel, Switzerland
| | - Ben Pais
- SRT-Biomedical B.V., Soest, Netherlands
- Ariceum Therapeutics GmbH, Berlin, Germany
| | | | | | | | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | | | - Nat P Lenzo
- GenesisCare, East Fremantle, Australia
- Department of Medicine, Curtin University, Perth, Australia
| | - Rodney J Hicks
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Melbourne, Australia
- Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Australia
| |
Collapse
|
23
|
Wang R, Guo L, Pan L, Tian R, Shen G. Effects of somatostatin analogs on uptake of radiolabeled somatostatin analogs on imaging: a systematic review and meta-analysis. Quant Imaging Med Surg 2023; 13:6814-6826. [PMID: 37869289 PMCID: PMC10585547 DOI: 10.21037/qims-23-477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 08/14/2023] [Indexed: 10/24/2023]
Abstract
Background The imaging of somatostatin receptors (SSTRs) plays a significant role in imaging neuroendocrine tumors (NETs). However, there has been no clear definition on whether it is necessary to withdraw somatostatin analogs (SSAs) before SSTRs imaging. We aimed to assess whether nonradioactive SSAs affect the uptake of radiolabeled SSAs on imaging for NETs patients. Methods The databases of PubMed, Embase, and Web of Science (WoS) were searched until March 12, 2022 to identify eligible studies. Maximum standardized uptake values (SUVmax) in tumor and normal tissues were extracted, pooled, and compared before and after SSAs treatment. The change of tumor-to-background/liver ratio was also described. The quality of each study was assessed using the revised Quality Assessment of Diagnostic Accuracy Studies-2 tool. Results A total of 9 articles involving 285 patients were included and 5 studies using Gallium-68-labeled [1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid]-D-Phe1-Tyr3-Thr8-octreotide (68Ga-DOTATATE) were used for pooled evaluation. We found a significantly decreased SUVmax in the liver (9.56±2.47 vs. 7.62±2.12, P=0.001) and spleen (25.74±7.14 vs. 20.39±6.07, P=0.006) after SSAs treatment whereas no significant differences were observed in the uptake of thyroid, adrenal, and pituitary gland. For either primary tumor sites or metastases, the SUVmax did not change significantly before and after SSAs treatment. The tumor-to-liver/background ratio increased following SSAs therapy. High heterogeneity was observed across the studies, mainly due to inherent diversity of study design, sample size, and scanning technique. Conclusions Based on current evidence, long-acting SSAs therapy before imaging has no effect on the uptake of radiolabeled SSAs at tumor primary sites and metastatic lesions, but results in a significant reduction of uptake in the liver and spleen. These findings may implicate the unnecessary discontinuation of SSAs before radiolabeled SSAs imaging.
Collapse
Affiliation(s)
- Rang Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Linlin Guo
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Pan
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Guohua Shen
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Sullivan TE, Hernandez Vargas S, Ghosh SC, AghaAmiri S, Ikoma N, Azhdarinia A. A translational blueprint for developing intraoperative imaging agents via radiopharmaceutical-guided drug design. Curr Opin Chem Biol 2023; 76:102376. [PMID: 37572489 DOI: 10.1016/j.cbpa.2023.102376] [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: 02/09/2023] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 08/14/2023]
Abstract
Cancer imaging is a rapidly evolving field due to the discovery of novel molecular targets and the availability of corresponding techniques to detect them with high precision, accuracy, and sensitivity. Nuclear medicine is the most widely used molecular imaging modality and has a growing toolkit of clinically used radiopharmaceuticals that enable whole-body tumor visualization, staging, and treatment monitoring for a variety of tumors in a non-invasive manner. The need for similar imaging capabilities in the operating room has led to the emergence of fluorescence-guided surgery (FGS) as a powerful technique that gives surgeons unprecedented ability to distinguish tumors from healthy tissues. While a variety of strategies have been used to develop contrast agents for FGS, the use of radiopharmaceuticals as models brings exceptional translational potential and has increasingly been explored. Here, we review strategies used to convert clinically used radiopharmaceuticals into fluorescent and multimodal counterparts. Unique preclinical and clinical capabilities stemming from radiopharmaceutical-based agent design are also discussed to illustrate the advantages of this approach.
Collapse
Affiliation(s)
- Teresa E Sullivan
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Sukhen C Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.
| |
Collapse
|
25
|
Gervasoni S, Öztürk I, Guccione C, Bosin A, Ruggerone P, Malloci G. Interaction of Radiopharmaceuticals with Somatostatin Receptor 2 Revealed by Molecular Dynamics Simulations. J Chem Inf Model 2023; 63:4924-4933. [PMID: 37466559 PMCID: PMC10428218 DOI: 10.1021/acs.jcim.3c00712] [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: 05/12/2023] [Indexed: 07/20/2023]
Abstract
The development of drugs targeting somatostatin receptor 2 (SSTR2), generally overexpressed in neuroendocrine tumors, is focus of intense research. A few molecules in conjugation with radionuclides are in clinical use for both diagnostic and therapeutic purposes. These radiopharmaceuticals are composed of a somatostatin analogue biovector conjugated to a chelator moiety bearing the radionuclide. To date, despite valuable efforts, a detailed molecular-level description of the interaction of radiopharmaceuticals in complex with SSTR2 has not yet been accomplished. Therefore, in this work, we carefully analyzed the key dynamical features and detailed molecular interactions of SSTR2 in complex with six radiopharmaceutical compounds selected among the few already in use (64Cu/68Ga-DOTATATE, 68Ga-DOTATOC, 64Cu-SARTATE) and some in clinical development (68Ga-DOTANOC, 64Cu-TETATATE). Through molecular dynamics simulations and exploiting recently available structures of SSTR2, we explored the influence of the different portions of the compounds (peptide, radionuclide, and chelator) in the interaction with the receptor. We identified the most stable binding modes and found distinct interaction patterns characterizing the six compounds. We thus unveiled detailed molecular interactions crucial for the recognition of this class of radiopharmaceuticals. The microscopically well-founded analysis presented in this study provides guidelines for the design of new potent ligands targeting SSTR2.
Collapse
Affiliation(s)
| | | | - Camilla Guccione
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Andrea Bosin
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Paolo Ruggerone
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| | - Giuliano Malloci
- Department of Physics, University of Cagliari, Monserrato
(Cagliari) I-09042, Italy
| |
Collapse
|
26
|
Adnan A, Basu S. Somatostatin Receptor Targeted PET-CT and Its Role in the Management and Theranostics of Gastroenteropancreatic Neuroendocrine Neoplasms. Diagnostics (Basel) 2023; 13:2154. [PMID: 37443548 DOI: 10.3390/diagnostics13132154] [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: 05/01/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Somatostatin receptor (SSTR) agonist-based Positron Emission Tomography-Computed Tomography (PET-CT) imaging is nowadays the mainstay for the assessment and diagnostic imaging of neuroendocrine neoplasms (NEN), especially in well-differentiated neuroendocrine tumors (NET) (World Health Organization (WHO) grade I and II). Major clinical indications for SSTR imaging are primary staging and metastatic workup, especially (a) before surgery, (b) detection of unknown primary in metastatic NET, (c) patient selection for theranostics and appropriate therapy, especially peptide receptor radionuclide therapy (PRRT), while less major indications include treatment response evaluation on and disease prognostication. Dual tracer PET-CT imaging using SSTR targeted PET tracers, viz. [68Ga]Ga-DOTA-Tyr3-Octreotate (DOTA-TATE) and [68Ga]Ga-DOTA-NaI3-Octreotide (DOTA-NOC), and fluorodeoxyglucose (FDG), have recently gained widespread acceptance for better assessment of whole-body tumor biology compared to single-site histopathology, in terms of being non-invasive and the ability to assess inter- and intra-tumoral heterogeneity on a global scale. FDG uptake has been identified as independent adverse risk factor in various studies. Recently, somatostatin receptor antagonists have been shown to be more sensitive and specific in detecting the disease. The aim of this review article is to summarize the clinical importance of SSTR-based imaging in the clinical management of neuroendocrine and related tumors.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
- Homi Bhabha National Institute, Mumbai 400094, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, JerbaiWadia Road, Parel, Mumbai 400012, India
| |
Collapse
|
27
|
Lepareur N, Ramée B, Mougin-Degraef M, Bourgeois M. Clinical Advances and Perspectives in Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1733. [PMID: 37376181 DOI: 10.3390/pharmaceutics15061733] [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: 05/19/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted radionuclide therapy has become increasingly prominent as a nuclear medicine subspecialty. For many decades, treatment with radionuclides has been mainly restricted to the use of iodine-131 in thyroid disorders. Currently, radiopharmaceuticals, consisting of a radionuclide coupled to a vector that binds to a desired biological target with high specificity, are being developed. The objective is to be as selective as possible at the tumor level, while limiting the dose received at the healthy tissue level. In recent years, a better understanding of molecular mechanisms of cancer, as well as the appearance of innovative targeting agents (antibodies, peptides, and small molecules) and the availability of new radioisotopes, have enabled considerable advances in the field of vectorized internal radiotherapy with a better therapeutic efficacy, radiation safety and personalized treatments. For instance, targeting the tumor microenvironment, instead of the cancer cells, now appears particularly attractive. Several radiopharmaceuticals for therapeutic targeting have shown clinical value in several types of tumors and have been or will soon be approved and authorized for clinical use. Following their clinical and commercial success, research in that domain is particularly growing, with the clinical pipeline appearing as a promising target. This review aims to provide an overview of current research on targeting radionuclide therapy.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, 35000 Rennes, France
- Inserm, INRAE, Institut NUMECAN (Nutrition, Métabolismes et Cancer)-UMR 1317, Univ Rennes, 35000 Rennes, France
| | - Barthélémy Ramée
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
| | - Marie Mougin-Degraef
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 44000 Nantes, France
- Inserm, CNRS, CRCI2NA (Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes-Angers)-UMR 1307, Université de Nantes, ERL 6001, 44000 Nantes, France
- Groupement d'Intérêt Public ARRONAX, 1 Rue Aronnax, 44817 Saint Herblain, France
| |
Collapse
|
28
|
Barta P, Nachtigal P, Maixnerova J, Zemankova L, Trejtnar F. Validation of Freshly Isolated Rat Renal Cells as a Tool for Preclinical Assessment of Radiolabeled Receptor-Specific Peptide Uptake in the Kidney. Pharmaceuticals (Basel) 2023; 16:ph16050696. [PMID: 37242479 DOI: 10.3390/ph16050696] [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: 02/27/2023] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The synthetic analogs of regulatory peptides radiolabeled with adequate radionuclides are perspective tools in nuclear medicine. However, undesirable uptake and retention in the kidney limit their application. Specific in vitro methods are used to evaluate undesirable renal accumulation. Therefore, we investigated the usefulness of freshly isolated rat renal cells for evaluating renal cellular uptake of receptor-specific peptide analogs. Special attention was given to megalin as this transport system is an important contributor to the active renal uptake of the peptides. Freshly isolated renal cells were obtained from native rat kidneys by the collagenase method. Compounds with known accumulation in renal cells were used to verify the viability of cellular transport systems. Megalin expressions in isolated rat renal cells were compared to two other potential renal cell models by Western blotting. Specific tubular cell markers were used to confirm the presence of proximal tubular cells expressing megalin in isolated rat renal cell preparations by immunohistochemistry. Colocalization experiments on isolated rat kidney cells confirmed the presence of proximal tubular cells bearing megalin in preparations. The applicability of the method was tested by an accumulation study with several analogs of somatostatin and gastrin labeled with indium-111 or lutetium-177. Therefore, isolated rat renal cells may be an effective screening tool for in vitro analyses of renal uptake and comparative renal accumulation studies of radiolabeled peptides or other radiolabeled compounds with potential nephrotoxicity.
Collapse
Affiliation(s)
- Pavel Barta
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy in Hradec Kralové, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralové, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Jana Maixnerova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralové, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| | - Lenka Zemankova
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Frantisek Trejtnar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralové, Charles University, Akademika Heyrovskeho 1203, 50005 Hradec Kralove, Czech Republic
| |
Collapse
|
29
|
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
|
30
|
Du Y, Yan B. Ocular immune privilege and retinal pigment epithelial cells. J Leukoc Biol 2023; 113:288-304. [PMID: 36805720 DOI: 10.1093/jleuko/qiac016] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Indexed: 02/04/2023] Open
Abstract
The ocular tissue microenvironment is immune-privileged and uses multiple immunosuppressive mechanisms to prevent the induction of inflammation. The retinal pigment epithelium plays an essential role in ocular immune privilege. In addition to serving as a blood barrier separating the fenestrated choriocapillaris from the retina, the retinal pigment epithelium is a source of immunosuppressive cytokines and membrane-bound negative regulators that modulate the activity of immune cells within the retina. This article reviews the current understanding of how retinal pigment epithelium cells mediate immune regulation, focusing on the changes under pathologic conditions.
Collapse
Affiliation(s)
- Yuxiang Du
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| | - Bo Yan
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| |
Collapse
|
31
|
Schwarz JL, Williams JK, Keutgen XM, Liao CY. Light It Up! The Use of DOTATATE in Diagnosis and Treatment of Neuroendocrine Neoplasms. Surg Pathol Clin 2023; 16:151-161. [PMID: 36739162 DOI: 10.1016/j.path.2022.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Radiolabeled somatostatin analogs are increasingly used in the diagnosis and treatment of neuroendocrine tumors. Diagnostic imaging with 68Ga-DOTATATE PET/CT has demonstrated the improved sensitivity in detecting primary and metastatic neuroendocrine lesions compared with conventional imaging and prior generation somatostatin receptor imaging. Peptide receptor radionuclide therapy with 177Lu-DOTATATE is now frequently included in the management of neuroendocrine neoplasms, with prospective randomized control studies demonstrating its beneficial impact on survival and quality of life. Nonetheless, peptide rector radionuclide therapy is still considered palliative rather than curative and may be accompanied by adverse effects.
Collapse
Affiliation(s)
- Jason L Schwarz
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6040, Chicago, IL 60637, USA
| | - Jelani K Williams
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 6040, Chicago, IL 60637, USA
| | - Xavier M Keutgen
- Division of General Surgery and Surgical Oncology, Department of Surgery, University of Chicago, University of Chicago Medicine, 5841 S. Maryland Avenue, MC 4052, Chicago, IL 60637, USA
| | - Chih-Yi Liao
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, University of Chicago Medicine, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA.
| |
Collapse
|
32
|
Zavvar TS, Hörmann AA, Klingler M, Summer D, Rangger C, Desrues L, Castel H, Gandolfo P, von Guggenberg E. Effects of Side Chain and Peptide Bond Modifications on the Targeting Properties of Stabilized Minigastrin Analogs. Pharmaceuticals (Basel) 2023; 16:278. [PMID: 37052226 PMCID: PMC9959130 DOI: 10.3390/ph16020278] [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: 12/21/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Different attempts have been made in the past two decades to develop radiolabeled peptide conjugates with enhanced pharmacokinetic properties in order to improve the application for tumor imaging and peptide receptor radionuclide therapy (PRRT), which targets the cholecystokinin-2 receptor (CCK2R). In this paper, the influence of different side chain and peptide bond modifications has been explored for the minigastrin analog DOTA-DGlu-Ala-Tyr-Gly-Trp-(N-Me)Nle-Asp-1Nal-NH2 (DOTA-MGS5). Based on this lead structure, five new derivatives were synthesized for radiolabeling with trivalent radiometals. Different chemical and biological properties of the new derivatives were analyzed. Receptor interaction of the peptide derivatives and cell internalization of the radiolabeled peptides were studied in A431-CCK2R cells. The stability of the radiolabeled peptides in vivo was investigated using BALB/c mice. Tumor targeting of all 111In-labeled peptide conjugates, and of a selected compound radiolabeled with gallium-68 and lutetium-177, was evaluated in BALB/c nude mice xenografted with A431-CCK2R and A431-mock cells. All 111In-labeled conjugates, except [111In]In-DOTA-[Phe8]MGS5, showed a high resistance against enzymatic degradation. A high receptor affinity with IC50 values in the low nanomolar range was confirmed for most of the peptide derivatives. The specific cell internalization over time was 35.3-47.3% for all radiopeptides 4 h after incubation. Only [111In]In-DOTA-MGS5[NHCH3] exhibited a lower cell internalization of 6.6 ± 2.8%. An overall improved resistance against enzymatic degradation was confirmed in vivo. Of the radiopeptides studied, [111In]In-DOTA-[(N-Me)1Nal8]MGS5 showed the most promising targeting properties, with significantly increased accumulation of radioactivity in A431-CCK2R xenografts (48.1 ± 9.2% IA/g) and reduced accumulation of radioactivity in stomach (4.2 ± 0.5% IA/g). However, in comparison with DOTA-MGS5, a higher influence on the targeting properties was observed for the change of radiometal, resulting in a tumor uptake of 15.67 ± 2.21% IA/g for [68Ga]Ga-DOTA-[(N-Me)1Nal8]MGS5 and 35.13 ± 6.32% IA/g for [177Lu]Lu-DOTA-[(N-Me)1Nal8]MGS5.
Collapse
Affiliation(s)
- Taraneh Sadat Zavvar
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anton Amadeus Hörmann
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Maximilian Klingler
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Laurence Desrues
- Inserm U1245, University Rouen Normandie, 76000 Rouen, France
- Institute of Research and Biomedical Innovation (IRIB), 76000 Rouen, France
| | - Hélène Castel
- Inserm U1245, University Rouen Normandie, 76000 Rouen, France
- Institute of Research and Biomedical Innovation (IRIB), 76000 Rouen, France
| | - Pierrick Gandolfo
- Inserm U1245, University Rouen Normandie, 76000 Rouen, France
- Institute of Research and Biomedical Innovation (IRIB), 76000 Rouen, France
| | | |
Collapse
|
33
|
Makris G, Li Y, Gallazzi F, Kuchuk M, Wang J, Lewis MR, Jurisson SS, Hennkens HM. Evaluation of Re/ 99mTc-labeled somatostatin receptor-targeting peptide complexes synthesized via direct metal cyclization. RADIOCHIM ACTA 2023. [DOI: 10.1515/ract-2022-0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Abstract
With interest in the development of somatostatin receptor (SSTR) targeting agents for potential application in diagnostic SPECT imaging (99mTc) or Peptide Radionuclide Receptor Therapy (PRRT, 186Re or 188Re) of neuroendocrine tumors, we present herein 99mTc/Re (radio)complexes synthesized by the integrated (radio)labeling approach of peptide cyclization via metal complexation. In particular, we utilized the potent SSTR2 peptide antagonist sequence DOTA-4-NO2-Phe-c(DCys-Tyr-DTrp-Lys-Thr-Cys)-DTyr-NH2 (DOTA-sst2-ANT) and report the syntheses and in vitro evaluations of its respective [99mTc]Tc/Re-cyclized peptides ([99mTc]Tc/Re-cyc-DOTA-sst2-ANT). The Re-cyc-DOTA-sst2-ANT complex was synthesized via an on-resin Re(V)-cyclization reaction using the ReOCl3(PPh3)2 precursor and consisted of three isomers characterized by LC–ESI-MS. The [99mTc]Tc-cyclized analogue was prepared via a ligand exchange reaction of the [99mTc][TcO]3+ core through a [99mTc]Tc-glucoheptonate intermediate with linear DOTA-sst2-ANT and was characterized by comparative HPLC studies against Re-cyc-DOTA-sst2-ANT. Good in vitro binding affinity was demonstrated in SSTR-expressing cells (AR42J) by the Re-cyc-DOTA-sst2-ANT major isomer, similar to the potent binder Lu-DOTA-sst2-ANT, in which the Lu metal was complexed by the bifunctional chelator DOTA versus via peptide cyclization. [99mTc]Tc-cyc-DOTA-sst2-ANT was obtained in high radiochemical yield, also with an elution pattern of three isomers observed by HPLC analysis, which were comparable yet not identical to those of Re-cyc-DOTA-sst2-ANT. The [99mTc]Tc-tracer complex was shown to be hydrophilic, and stability studies at 4 h demonstrated that it remained intact in both PBS and in rat serum, with low non-specific rat serum protein binding, while exhibiting more moderate stability in 1 mM cysteine. These findings demonstrate that direct Re/[99mTc]Tc-cyclization of DOTA-sst2-ANT is feasible and may be used as an alternative approach to the bifunctional chelate labeling strategy. However, given that the non-radioactive (Re) and radiotracer (99mTc) analogues are not identical and both form isomeric products in equilibrium, additional design modifications will be necessary prior to in vivo application of [99mTc]Tc/Re-cyc-DOTA-sst2-ANT.
Collapse
Affiliation(s)
- George Makris
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
| | - Yawen Li
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| | - Fabio Gallazzi
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
- Molecular Interactions Core, University of Missouri , Columbia , MO 65211 , USA
| | - Marina Kuchuk
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
| | - Jing Wang
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
- Institute of Nuclear Physics and Chemistry, China Academy of Engineering Physics , Mianyang , Sichuan 621900 , P. R. China
| | - Michael R. Lewis
- Department of Veterinary Medicine and Surgery , University of Missouri , Columbia , MO 65211 , USA
- Research Service, Harry S. Truman Memorial Veterans’ Hospital , Columbia , MO 65201 , USA
| | - Silvia S. Jurisson
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| | - Heather M. Hennkens
- Research Reactor Center, University of Missouri , Columbia , MO 65211 , USA
- Department of Chemistry , University of Missouri , Columbia , MO 65211 , USA
| |
Collapse
|
34
|
Same-day comparative protocol PET/CT-PET/MRI [ 68 Ga]Ga-DOTA-TOC in paragangliomas and pheochromocytomas: an approach to personalized medicine. Cancer Imaging 2023; 23:4. [PMID: 36627700 PMCID: PMC9832675 DOI: 10.1186/s40644-023-00521-6] [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: 10/10/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND PET/MRI is an emerging imaging modality which enables the evaluation and quantification of biochemical processes in tissues, complemented with accurate anatomical information and low radiation exposure. In the framework of theragnosis, PET/MRI is of special interest due to its ability to delineate small lesions, adequately quantify them, and therefore to plan targeted therapies. The aim of this study was to validate the diagnostic performance of [68 Ga]Ga-DOTA-TOC PET/MRI compared to PET/CT in advanced disease paragangliomas and pheochromocytomas (PGGLs) to assess in which clinical settings, PET/MRI may have a greater diagnostic yield. METHODS We performed a same-day protocol with consecutive acquisition of a PET/CT and a PET/MRI after a single [68 Ga]Ga-DOTA-TOC injection in 25 patients. Intermodality agreement, Krenning Score (KS), SUVmax (Standard Uptake Value), target-to-liver-ratio (TLR), clinical setting, location, and size were assessed. RESULTS The diagnostic accuracy with PET/MRI increased by 14.6% compared to PET/CT especially in bone and liver locations (mean size of new lesions was 3.73 mm). PET/MRI revealed a higher overall lesion uptake than PET/CT (TLR 4.12 vs 2.44) and implied an upward elevation of the KS in up to 60% of patients. The KS changed in 30.4% of the evaluated lesions (mean size 11.89 mm), in 18.4% of the lesions it increased from KS 2 on PET/CT to a KS ≥ 3 on PET/MRI and 24.96% of the lesions per patient with multifocal disease displayed a KS ≥ 3 on PET/MR, that were not detected or showed lower KS on PET/CT. In 12% of patients, PET/MRI modified clinical management. CONCLUSIONS PET/MRI showed minor advantages over conventional PET/CT in the detection of new lesions but increased the intensity of SSRs expression in a significant number of them, opening the door to select which patients and clinical settings can benefit from performing PET/MRI.
Collapse
|
35
|
Management of Small Nonfunctioning Pancreatic Neuroendocrine Neoplasms: Current Opinion and Controversies. J Clin Med 2022; 12:jcm12010251. [PMID: 36615051 PMCID: PMC9821009 DOI: 10.3390/jcm12010251] [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: 12/07/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
The incidence of small and asymptomatic pancreatic neuroendocrine neoplasms (PNENs) has increased due to the widespread use of high-resolution diagnostic imaging in screening programs. Most PNENs are slow-growing indolent neoplasms. However, a local invasion or metastasis can sometimes occur with PNENs, leading to a poor prognosis. The management of small, nonfunctioning PNENs remains under debate. The National Comprehensive Cancer Network guidelines recommend observation in selected cases of small PNENs less than 2 cm. Pancreatic surgery remains a high-risk operation with a 28-30% morbidity and 1% mortality. Therefore, the decision on how to manage small PNENs is challenging. This review focuses on the management of small nonfunctioning PNENs. We also highlight the malignant potential of small PNENs according to tumor size, tumor grade, and tumor biomarker. Endoscopic-ultrasound-guided biopsy is recommended to evaluate the potential risk of malignancy. Furthermore, we discuss the current guidelines and future directions for the management of small PNENs.
Collapse
|
36
|
Khachatryan DS, Kolotaev AV, Malyutina ER, Osipov VN. Synthesis of precursors for obtaining targeted radiopharmaceuticals based on short peptides, analogs of the hormone somatostatin. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3700-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
37
|
Prosperi D, Gentiloni Silveri G, Panzuto F, Faggiano A, Russo VM, Caruso D, Polici M, Lauri C, Filice A, Laghi A, Signore A. Nuclear Medicine and Radiological Imaging of Pancreatic Neuroendocrine Neoplasms: A Multidisciplinary Update. J Clin Med 2022; 11:jcm11226836. [PMID: 36431313 PMCID: PMC9694730 DOI: 10.3390/jcm11226836] [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: 10/03/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Pancreatic neuroendocrine neoplasms (panNENs) are part of a large family of tumors arising from the neuroendocrine system. PanNENs show low-intermediate tumor grade and generally high somatostatin receptor (SSTR) expression. Therefore, panNENs benefit from functional imaging with 68Ga-somatostatin analogues (SSA) for diagnosis, staging, and treatment choice in parallel with morphological imaging. This narrative review aims to present conventional imaging techniques and new perspectives in the management of panNENs, providing the clinicians with useful insight for clinical practice. The 68Ga-SSA PET/CT is the most widely used in panNENs, not only fr diagnosis and staging purpose but also to characterize the biology of the tumor and its responsiveness to SSAs. On the contrary, the 18F-Fluordeoxiglucose (FDG) PET/CT is not employed systematically in all panNEN patients, being generally preferred in G2-G3, to predict aggressiveness and progression rate. The combination of 68Ga-SSA PET/CT and 18F-FDG PET/CT can finally suggest the best therapeutic strategy. Other radiopharmaceuticals are 68Ga-exendin-4 in case of insulinomas and 18F-dopamine (DOPA), which can be helpful in SSTR-negative tumors. New promising but still-under-investigation radiopharmaceuticals include radiolabeled SSTR antagonists and 18F-SSAs. Conventional imaging includes contrast enhanced CT and multiparametric MRI. There are now enriched by radiomics, a new non-invasive imaging approach, very promising to early predict tumor response or progression.
Collapse
Affiliation(s)
- Daniela Prosperi
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Guido Gentiloni Silveri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Francesco Panzuto
- Digestive Disease Unit, Department of Medical-Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Antongiulio Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant’Andrea University Hospital, ENETS Center of Excellence, Sapienza University of Rome, 00189 Roma, Italy
| | - Vincenzo Marcello Russo
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Damiano Caruso
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Michela Polici
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Chiara Lauri
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
- Correspondence:
| | - Angelina Filice
- Nucler Medicine Unit, AUSL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Andrea Laghi
- Radiology Unit, Department of Medical Surgical Sciences and Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| | - Alberto Signore
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Sant’Andrea University Hospital, Sapienza University of Rome, 00189 Roma, Italy
| |
Collapse
|
38
|
Glucose Metabolism Modification Induced by Radioligand Therapy with [ 177Lu]Lu/[ 90Y]Y-DOTATOC in Advanced Neuroendocrine Neoplasms: A Prospective Pilot Study within FENET-2016 Trial. Pharmaceutics 2022; 14:pharmaceutics14102009. [PMID: 36297443 PMCID: PMC9612170 DOI: 10.3390/pharmaceutics14102009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/13/2022] Open
Abstract
[18F]F-FDG (FDG) PET is emerging as a relevant diagnostic and prognostic tool in neuroendocrine neoplasms (NENs), as a simultaneous decrease in [68Ga]Ga-DOTA peptides and increase in FDG uptake (the “flip-flop” phenomenon) occurs during the natural history of these tumors. The aim of this study was to evaluate the variations on FDG PET in NEN patients treated with two different schemes of radioligand therapy (RLT) and to correlate them with clinical−pathologic variables. A prospective evaluation of 108 lesions in 56 patients (33 males and 23 females; median age, 64.5 years) affected by NENs of various primary origins (28 pancreatic, 13 gastrointestinal, 9 bronchial, 6 unknown primary (CUP-NENs) and 1 pheochromocytoma) and grades (median Ki-67 = 9%) was performed. The patients were treated with RLT within the phase II clinical trial FENET-2016 (CTID: NCT04790708). RLT was offered for 32 patients with the MONO scheme (five cycles of [177Lu]Lu-DOTATOC) and for 24 with the DUO scheme (three cycles of [177Lu]Lu-DOTATOC alternated with two cycles of [90Y]Y-DOTATOC). Variations in terms of the ΔSUVmax of a maximum of three target lesions per patient (58 for MONO and 50 for DUO RLT) were assessed between baseline and 3 months post-RLT FDG PET. In patients with negative baseline FDG PET, the three most relevant lesions on [68Ga]Ga-DOTA-peptide PET were assessed and matched on post-RLT FDG PET, to check for any possible changes in FDG avidity. Thirty-five patients (62.5%) had at least one pathological FDG uptake at the baseline scans, but the number was reduced to 29 (52%) after RLT. In the patients treated with DUO-scheme RLT, 20 out of 50 lesions were FDG positive before therapy, whereas only 14 were confirmed after RLT (p = 0.03). Moreover, none of the 30 FDG-negative lesions showed an increased FDG uptake after RLT. The lesions of patients with pancreatic and CUP-NENs treated with the DUO scheme demonstrated a significant reduction in ΔSUVmax in comparison to those treated with MONO RLT (p = 0.03 and p = 0.04, respectively). Moreover, we found a mild positive correlation between the grading and ΔSUVmax in patients treated with the MONO scheme (r = 0.39, p < 0.02), while no evidence was detected for patients treated with the DUO scheme. Our results suggest that RLT, mostly with the DUO scheme, could be effective in changing NEN lesions’ glycometabolism, in particular, in patients affected by pancreatic and CUP-NENs, regardless of their Ki-67 index. Probably, associating [90Y]Y-labelled peptides, which have high energy emission and a crossfire effect, and [177Lu]Lu ones, characterized by a longer half-life and a safer profile for organs at risk, might represent a valid option in FDG-positive NENs addressed to RLT. Further studies are needed to validate our preliminary findings. In our opinion, FDG PET/CT should represent a potent tool for fully assessing a patient’s disease characteristics, both before and after RLT.
Collapse
|
39
|
Wu Y, Berisha A, Borniger JC. Neuropeptides in Cancer: Friend and Foe? Adv Biol (Weinh) 2022; 6:e2200111. [PMID: 35775608 DOI: 10.1002/adbi.202200111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/31/2022] [Indexed: 01/28/2023]
Abstract
Neuropeptides are small regulatory molecules found throughout the body, most notably in the nervous, cardiovascular, and gastrointestinal systems. They serve as neurotransmitters or hormones in the regulation of diverse physiological processes. Cancer cells escape normal growth control mechanisms by altering their expression of growth factors, receptors, or intracellular signals, and neuropeptides have recently been recognized as mitogens in cancer growth and development. Many neuropeptides and their receptors exist in multiple subtypes, coupling with different downstream signaling pathways and playing distinct roles in cancer progression. The consideration of neuropeptide/receptor systems as anticancer targets is already leading to new biological and diagnostic knowledge that has the potential to enhance the understanding and treatment of cancer. In this review, recent discoveries regarding neuropeptides in a wide range of cancers, emphasizing their mechanisms of action, signaling cascades, regulation, and therapeutic potential, are discussed. Current technologies used to manipulate and analyze neuropeptides/receptors are described. Applications of neuropeptide analogs and their receptor inhibitors in translational studies and radio-oncology are rapidly increasing, and the possibility for their integration into therapeutic trials and clinical treatment appears promising.
Collapse
Affiliation(s)
- Yue Wu
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| | - Adrian Berisha
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| | - Jeremy C Borniger
- Cold Spring Harbor Laboratory, One Bungtown Rd, Cold Spring Harbor, NY, 11724, USA
| |
Collapse
|
40
|
Zhao W, Han S, Qiu N, Feng W, Lu M, Zhang W, Wang M, Zhou Q, Chen S, Xu W, Du J, Chu X, Yi C, Dai A, Hu L, Shen MY, Sun Y, Zhang Q, Ma Y, Zhong W, Yang D, Wang MW, Wu B, Zhao Q. Structural insights into ligand recognition and selectivity of somatostatin receptors. Cell Res 2022; 32:761-772. [PMID: 35739238 PMCID: PMC9343605 DOI: 10.1038/s41422-022-00679-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Somatostatin receptors (SSTRs) play versatile roles in inhibiting the secretion of multiple hormones such as growth hormone and thyroid-stimulating hormone, and thus are considered as targets for treating multiple tumors. Despite great progress made in therapeutic development against this diverse receptor family, drugs that target SSTRs still show limited efficacy with preferential binding affinity and conspicuous side-effects. Here, we report five structures of SSTR2 and SSTR4 in different states, including two crystal structures of SSTR2 in complex with a selective peptide antagonist and a non-peptide agonist, respectively, a cryo-electron microscopy (cryo-EM) structure of Gi1-bound SSTR2 in the presence of the endogenous ligand SST-14, as well as two cryo-EM structures of Gi1-bound SSTR4 in complex with SST-14 and a small-molecule agonist J-2156, respectively. By comparison of the SSTR structures in different states, molecular mechanisms of agonism and antagonism were illustrated. Together with computational and functional analyses, the key determinants responsible for ligand recognition and selectivity of different SSTR subtypes and multiform binding modes of peptide and non-peptide ligands were identified. Insights gained in this study will help uncover ligand selectivity of various SSTRs and accelerate the development of new molecules with better efficacy by targeting SSTRs.
Collapse
Affiliation(s)
- Wenli Zhao
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Han
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang, China
| | - Na Qiu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenbo Feng
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mengjie Lu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenru Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mu Wang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qingtong Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shutian Chen
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wei Xu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Juan Du
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang, China
| | - Xiaojing Chu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Cuiying Yi
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Antao Dai
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | - Yingli Ma
- Amgen Asia R&D Center, Shanghai, China
| | - Wenge Zhong
- Amgen Asia R&D Center, Shanghai, China.,Regor Therapeutics, Shanghai, China
| | - Dehua Yang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Ming-Wei Wang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Beili Wu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, Zhejiang, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Qiang Zhao
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China. .,Zhongshan Institute of Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, China.
| |
Collapse
|
41
|
Vahidfar N, Farzanehfar S, Abbasi M, Mirzaei S, Delpassand ES, Abbaspour F, Salehi Y, Biersack HJ, Ahmadzadehfar H. Diagnostic Value of Radiolabelled Somatostatin Analogues for Neuroendocrine Tumour Diagnosis: The Benefits and Drawbacks of [ 64Cu]Cu-DOTA-TOC. Cancers (Basel) 2022; 14:1914. [PMID: 35454822 PMCID: PMC9027354 DOI: 10.3390/cancers14081914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroendocrine tumours (NETs) arise from secondary epithelial cell lines in the gastrointestinal or respiratory system organs. The rate of development of these tumours varies from an indolent to an aggressive course, typically being initially asymptomatic. The identification of these tumours is difficult, particularly because the primary tumour is often small and undetectable by conventional anatomical imaging. Consequently, diagnosis of NETs is complicated and has been a significant challenge until recently. In the last 30 years, the advent of novel nuclear medicine diagnostic procedures has led to a substantial increase in NET detection. Great varieties of exclusive single photon emission computed tomography (SPECT) and positron emission tomography (PET) radiopharmaceuticals for detecting NETs are being applied successfully in clinical settings, including [111In]In-pentetreotide, [99mTc]Tc-HYNIC-TOC/TATE, [68Ga]Ga-DOTA-TATE, and [64Cu]Cu-DOTA-TOC/TATE. Among these tracers for functional imaging, PET radiopharmaceuticals are clearly and substantially superior to planar or SPECT imaging radiopharmaceuticals. The main advantages include higher resolution, better sensitivity and increased lesion-to-background uptake. An advantage of diagnosis with a radiopharmaceutical is the capacity of theranostics to provide concomitant diagnosis and treatment with particulate radionuclides, such as beta and alpha emitters including Lutetium-177 (177Lu) and Actinium-225 (225Ac). Due to these unique challenges involved with diagnosing NETs, various PET tracers have been developed. This review compares the clinical characteristics of radiolabelled somatostatin analogues for NET diagnosis, focusing on the most recently FDA-approved [64Cu]Cu-DOTA-TATE as a state-of-the art NET-PET/CT radiopharmaceutical.
Collapse
Affiliation(s)
- Nasim Vahidfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.); (M.A.); (Y.S.)
| | - Saeed Farzanehfar
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.); (M.A.); (Y.S.)
| | - Mehrshad Abbasi
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.); (M.A.); (Y.S.)
| | - Siroos Mirzaei
- Clinic Ottakring, Institute of Nuclear Medicine with PET-Center, 1220 Vienna, Austria;
| | - Ebrahim S. Delpassand
- RadioMedix, Inc., Houston, TX 77041, USA;
- Excel Diagnostics and Nuclear Oncology Center, Houston, TX 77042, USA
| | - Farzad Abbaspour
- Division of Nuclear Medicine, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, ON K1H 8L6, Canada;
| | - Yalda Salehi
- Department of Nuclear Medicine, Vali-Asr Hospital, Tehran University of Medical Sciences, Tehran 1419733133, Iran; (N.V.); (S.F.); (M.A.); (Y.S.)
| | - Hans Jürgen Biersack
- Department of Nuclear Medicine, University Hospital Bonn, 53127 Bonn, Germany;
- Betaklinik Bonn, 53227 Bonn, Germany
| | | |
Collapse
|
42
|
Vène E, Jarnouen K, Ribault C, Vlach M, Verres Y, Bourgeois M, Lepareur N, Cammas-Marion S, Loyer P. Circumsporozoite Protein of Plasmodium berghei- and George Baker Virus A-Derived Peptides Trigger Efficient Cell Internalization of Bioconjugates and Functionalized Poly(ethylene glycol)- b-poly(benzyl malate)-Based Nanoparticles in Human Hepatoma Cells. Pharmaceutics 2022; 14:804. [PMID: 35456637 PMCID: PMC9028075 DOI: 10.3390/pharmaceutics14040804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
In order to identify the peptides, selected from the literature, that exhibit the strongest tropism towards human hepatoma cells, cell uptake assays were performed using biotinylated synthetic peptides bound to fluorescent streptavidin or engrafted onto nanoparticles (NPs), prepared from biotin-poly(ethylene glycol)-block-poly(benzyl malate) (Biot-PEG-b-PMLABe) via streptavidin bridging. Two peptides, derived from the circumsporozoite protein of Plasmodium berghei- (CPB) and George Baker (GB) Virus A (GBVA10-9), strongly enhanced the endocytosis of both streptavidin conjugates and NPs in hepatoma cells, compared to primary human hepatocytes and non-hepatic cells. Unexpectedly, the uptake of CPB- and GBVA10-9 functionalized PEG-b-PMLABe-based NPs by hepatoma cells involved, at least in part, the peptide binding to apolipoproteins, which would promote NP's interactions with cell membrane receptors of HDL particles. In addition, CPB and GBVA10-9 peptide-streptavidin conjugates favored the uptake by hepatoma cells over that of the human macrophages, known to strongly internalize nanoparticles by phagocytosis. These two peptides are promising candidate ligands for targeting hepatocellular carcinomas.
Collapse
Affiliation(s)
- Elise Vène
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU Rennes, F-35033 Rennes, France
| | - Kathleen Jarnouen
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Catherine Ribault
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- INRAE, Institut AGRO, PEGASE UMR 1348, F-35590 Saint-Gilles, France
| | - Yann Verres
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Mickaël Bourgeois
- CRCINA, Inserm, CNRS, Université de Nantes, F-44000 Nantes, France;
- ARRONAX Cyclotron, F-44817 Saint Herblain, France
| | - Nicolas Lepareur
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Comprehensive Cancer Center Eugène Marquis, F-35000 Rennes, France
| | - Sandrine Cammas-Marion
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Institut des Sciences Chimiques de Rennes (ISCR), Ecole Nationale Supérieure de Chimie de Rennes, CNRS UMR 6226, University of Rennes, F-35042 Rennes, France
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| |
Collapse
|
43
|
Du Y, Chen Z, Duan X, Yan P, Zhang C, Kang L, Wang R. 99mTc-labeled peptide targeting interleukin 13 receptor α 2 for tumor imaging in a cervical cancer mouse model. Ann Nucl Med 2022; 36:360-372. [PMID: 35032308 DOI: 10.1007/s12149-022-01715-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 01/06/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Pep-1 (CGEMGWVRC) can potently bind to interleukin 13 receptor α 2 (IL-13Rα2), a tumor-restricted receptor found to be expressed in various malignancies. In this study, we intended to prepare a 99mTc-labeled probe and evaluate its in vivo tumor accumulation properties in a cervical cancer xenograft model. METHODS The Pep-1 was designed and radiolabeled with 99mTc by conjugation with mercaptoacetyl-triglycine (MAG3). The labeling yield, radiochemical purity and stability were characterized in vitro. Cell uptake assays and fluorescence imaging were conducted for qualitative and quantitative evaluation of the specificity and affinity of Pep-1. Flow cytometry and tissue immunofluorescence were used to confirm the IL-13Rα2 expression in cervical cancer. Biodistribution and in vivo imaging were performed periodically to evaluate the imaging value of 99mTc-MAG3-Pep-1 in cervical cancer xenograft model. RESULTS 99mTc-MAG3-Pep-1 was successfully prepared with a high labeling yield and radiochemical purity (> 95%). Specific cell uptake was demonstrated by scramble control and unlabeled MAG3-Pep-1 blockade. Flow cytometry and tissue immunofluorescence also confirmed the mild IL-13Rα2 expression of HeLa. In the gamma imaging study and biodistribution, the tumors were imaged clearly at 2-6 h after injection of 99mTc-MAG3-Pep-1 and the accumulation of 99mTc-MAG3-Pep-1 in tumor was significantly higher than that in the blocking and scramble controls, demonstrating ligand-receptor binding specificity. CONCLUSIONS This work demonstrated that 99mTc-MAG3-Pep-1 can bind to cervical cancer with high affinity and specificity. MAG3-Pep-1 may be a prospective precursor for IL-13Rα2-expressing cancer therapy.
Collapse
Affiliation(s)
- Yujing Du
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China.
| | - Ping Yan
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Chunli Zhang
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, No.8, Xishiku Street, Xicheng District, Beijing, 100034, China.
- Department of Nuclear Medicine, Peking University International Hospital, 1 life Garden Road, Zhongguancun Life Science Park, Changping District, Beijing, 102206, China.
| |
Collapse
|
44
|
Ichikawa Y, Kobayashi N, Takano S, Kato I, Endo K, Inoue T. Net theranostics. Cancer Sci 2022; 113:1930-1938. [PMID: 35271754 PMCID: PMC9207370 DOI: 10.1111/cas.15327] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/17/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022] Open
Abstract
Theranostics is a term coined by combining the words “therapeutics” and “diagnostics,” referring to single chemical entities developed to deliver therapy and diagnosis simultaneously. Neuroendocrine tumors are rare cancers that occur in various organs of the body, and they express neuroendocrine factors such as chromogranin A and somatostatin receptor. Somatostatin analogs bind to somatostatin receptor, and when combined with diagnostic radionuclides, such as gamma‐emitters, are utilized for diagnosis of neuroendocrine tumor. Somatostatin receptor scintigraphy when combined with therapeutic radionuclides, such as beta‐emitters, are effective in treating neuroendocrine tumor as peptide receptor radionuclide therapy. Somatostatin receptor scintigraphy and peptide receptor radionuclide therapy are some of the most frequently used and successful theranostics for neuroendocrine tumor. In Japan, radiopharmaceuticals are regulated under a complex law system, creating a significant drug lag, which is a major public concern. It took nearly 10 years to obtain the approval for somatostatin receptor scintigraphy and peptide receptor radionuclide therapy use by the Japanese government. In 2021, 111Lu‐DOTATATE (Lutathera), a drug for peptide receptor radionuclide therapy, was covered by insurance in Japan. In this review, we summarize the history of the development of neuroendocrine tumor theranostics and theranostics in general, as therapeutic treatment for cancer in the future. Furthermore, we briefly address the Japanese point of view regarding the development of new radiopharmaceuticals.
Collapse
Affiliation(s)
- Yasushi Ichikawa
- Department of Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Shoko Takano
- Department of Radiation Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keigo Endo
- Kyoto College of Medical Science, Kyoto, Japan
| | - Tomio Inoue
- Department of Radiation Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Shonan Kamakura General Hospital, Kamakura, Japan
| |
Collapse
|
45
|
Fani M, Mansi R, Nicolas GP, Wild D. Radiolabeled Somatostatin Analogs-A Continuously Evolving Class of Radiopharmaceuticals. Cancers (Basel) 2022; 14:cancers14051172. [PMID: 35267479 PMCID: PMC8909681 DOI: 10.3390/cancers14051172] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Somatostatin receptors (SSTs) are recognized as favorable molecular targets in neuroendocrine tumors (NETs) and neuroendocrine neoplasms (NENs), with subtype 2 (SST2) being the predominantly and most frequently expressed. PET/CT imaging with 68Ga-labeled SST agonists, e.g., 68Ga-DOTA-TOC (SomaKit TOC®) or 68Ga-DOTA-TATE (NETSPOT®), plays an important role in staging and restaging these tumors and can identify patients who qualify and would potentially benefit from peptide receptor radionuclide therapy (PRRT) with the therapeutic counterparts 177Lu-DOTA-TOC or 177Lu-DOTA-TATE (Lutathera®). This is an important feature of SST targeting, as it allows a personalized treatment approach (theranostic approach). Today, new developments hold promise for enhancing diagnostic accuracy and therapeutic efficacy. Among them, the use of SST2 antagonists, such as JR11 and LM3, has shown certain advantages in improving image sensitivity and tumor radiation dose, and there is evidence that they may find application in other oncological indications beyond NETs and NENs. In addition, PRRT performed with more cytotoxic α-emitters, such as 225Ac, or β- and Auger electrons, such as 161Tb, presents higher efficacy. It remains to be seen if any of these new developments will overpower the established radiolabeled SST analogs and PRRT with β--emitters.
Collapse
Affiliation(s)
- Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, 4031 Basel, Switzerland;
- Correspondence:
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, 4031 Basel, Switzerland;
| | - Guillaume P. Nicolas
- Division of Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (G.P.N.); (D.W.)
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, 4031 Basel, Switzerland
| | - Damian Wild
- Division of Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (G.P.N.); (D.W.)
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
46
|
Ambrosini V, Zanoni L, Filice A, Lamberti G, Argalia G, Fortunati E, Campana D, Versari A, Fanti S. Radiolabeled Somatostatin Analogues for Diagnosis and Treatment of Neuroendocrine Tumors. Cancers (Basel) 2022; 14:1055. [PMID: 35205805 PMCID: PMC8870358 DOI: 10.3390/cancers14041055] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are rare and heterogeneous tumors that require multidisciplinary discussion for optimal care. The theranostic approach (DOTA peptides labelled with 68Ga for diagnosis and with 90Y or 177Lu for therapy) plays a crucial role in the management of NENs to assess disease extension and as a criteria for peptide receptor radionuclide therapy (PRRT) eligibility based on somatostatin receptor (SSTR) expression. On the diagnostic side, [68Ga]Ga-DOTA peptides PET/CT (SSTR PET/CT) is the gold standard for imaging well-differentiated SSTR-expressing neuroendocrine tumors (NETs). [18F]FDG PET/CT is useful in higher grade NENs (NET G2 with Ki-67 > 10% and NET G3; NEC) for more accurate disease characterization and prognostication. Promising emerging radiopharmaceuticals include somatostatin analogues labelled with 18F (to overcome the limits imposed by 68Ga), and SSTR antagonists (for both diagnosis and therapy). On the therapeutic side, the evidence gathered over the past two decades indicates that PRRT is to be considered as an effective and safe treatment option for SSTR-expressing NETs, and is currently included in the therapeutic algorithms of the main scientific societies. The positioning of PRRT in the treatment sequence, as well as treatment personalization (e.g., tailored dosimetry, re-treatment, selection criteria, and combination with other alternative treatment options), is warranted in order to improve its efficacy while reducing toxicity. Although very preliminary (being mostly hampered by lack of methodological standardization, especially regarding feature selection/extraction) and often including small patient cohorts, radiomic studies in NETs are also presented. To date, the implementation of radiomics in clinical practice is still unclear. The purpose of this review is to offer an overview of radiolabeled SSTR analogues for theranostic use in NENs.
Collapse
Affiliation(s)
- Valentina Ambrosini
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Giuseppe Lamberti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giulia Argalia
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Emilia Fortunati
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
| | - Davide Campana
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (A.V.)
| | - Stefano Fanti
- Department of Experimental Diagnostic and Specialized Medicine, University of Bologna, 40138 Bologna, Italy; (V.A.); (G.L.); (G.A.); (E.F.); (D.C.); (S.F.)
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
47
|
Fersing C, Masurier N, Rubira L, Deshayes E, Lisowski V. AAZTA-Derived Chelators for the Design of Innovative Radiopharmaceuticals with Theranostic Applications. Pharmaceuticals (Basel) 2022; 15:234. [PMID: 35215346 PMCID: PMC8879111 DOI: 10.3390/ph15020234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
With the development of 68Ga and 177Lu radiochemistry, theranostic approaches in modern nuclear medicine enabling patient-centered personalized medicine applications have been growing in the last decade. In conjunction with the search for new relevant molecular targets, the design of innovative chelating agents to easily form stable complexes with various radiometals for theranostic applications has gained evident momentum. Initially conceived for magnetic resonance imaging applications, the chelating agent AAZTA features a mesocyclic seven-membered diazepane ring, conferring some of the properties of both acyclic and macrocyclic chelating agents. Described in the early 2000s, AAZTA and its derivatives exhibited interesting properties once complexed with metals and radiometals, combining a fast kinetic of formation with a slow kinetic of dissociation. Importantly, the extremely short coordination reaction times allowed by AAZTA derivatives were particularly suitable for short half-life radioelements (i.e., 68Ga). In view of these particular characteristics, the scope of this review is to provide a survey on the design, synthesis, and applications in the nuclear medicine/radiopharmacy field of AAZTA-derived chelators.
Collapse
Affiliation(s)
- Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
| | - Nicolas Masurier
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
| | - Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
| | - Emmanuel Deshayes
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
| | - Vincent Lisowski
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
- Department of Pharmacy, Lapeyronie Hospital, CHU Montpellier, 191 Av. du Doyen Gaston Giraud, 34295 Montpellier, France
| |
Collapse
|
48
|
Lepareur N. Cold Kit Labeling: The Future of 68Ga Radiopharmaceuticals? Front Med (Lausanne) 2022; 9:812050. [PMID: 35223907 PMCID: PMC8869247 DOI: 10.3389/fmed.2022.812050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last couple of decades, gallium-68 (68Ga) has gained a formidable interest for PET molecular imaging of various conditions, from cancer to infection, through cardiac pathologies or neuropathies. It has gained routine use, with successful radiopharmaceuticals such as somatostatin analogs ([68Ga]Ga-DOTATOC and [68Ga]GaDOTATATE) for neuroendocrine tumors, and PSMA ligands for prostate cancer. It represents a major clinical impact, particularly in the context of theranostics, coupled with their 177Lu-labeled counterparts. Beside those, a bunch of new 68Ga-labeled molecules are in the preclinical and clinical pipelines, with some of them showing great promise for patient care. Increasing clinical demand and regulatory issues have led to the development of automated procedures for the production of 68Ga radiopharmaceuticals. However, the widespread use of these radiopharmaceuticals may rely on simple and efficient radiolabeling methods, undemanding in terms of equipment and infrastructure. To make them technically and economically accessible to the medical community and its patients, it appears mandatory to develop a procedure similar to the well-established kit-based 99mTc chemistry. Already available commercial kits for the production of 68Ga radiopharmaceuticals have demonstrated the feasibility of using such an approach, thus paving the way for more kit-based 68Ga radiopharmaceuticals to be developed. This article discusses the development of 68Ga cold kit radiopharmacy, including technical issues, and regulatory aspects.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer), UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
49
|
Durmo R, Filice A, Fioroni F, Cervati V, Finocchiaro D, Coruzzi C, Besutti G, Fanello S, Frasoldati A, Versari A. Predictive and Prognostic Role of Pre-Therapy and Interim 68Ga-DOTATOC PET/CT Parameters in Metastatic Advanced Neuroendocrine Tumor Patients Treated with PRRT. Cancers (Basel) 2022; 14:cancers14030592. [PMID: 35158862 PMCID: PMC8833820 DOI: 10.3390/cancers14030592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Although a significant improvement has been achieved in the management of metastatic neuroendocrine tumor (NET), disease progression is observed in 20–30% of patients treated with peptide receptor radionuclide therapy (PRRT). Therefore, the early identification of patients who are at high risk of treatment failure is important to avoid futile therapy toxicities. The aim of this study was to identify biomarkers derived from baseline and interim 68Ga-DOTATOC PET/CT in patients undergoing PRRT. In 46 metastatic NET patients with available baseline and interim PET, only baseline total tumor volume (bTV) was able to discriminate responders to PRRT (partial response or stable disease) vs. non-responders. Patients with high bTV had also the worst overall survival. bTV, an imaging biomarker, integrated in the initial workup of NET patients could improve risk stratification and contribute to a tailored therapy approach. Abstract Peptide receptor radionuclide therapy (PRRT) is an effective therapeutic option in patients with metastatic neuroendocrine tumor (NET). However, PRRT fails in about 15–30% of cases. Identification of biomarkers predicting the response to PRRT is essential for treatment tailoring. We aimed to evaluate the predictive and prognostic role of semiquantitative and volumetric parameters obtained from the 68Ga-DOTATOC PET/CT before therapy (bPET) and after two cycles of PRRT (iPET). A total of 46 patients were included in this retrospective analysis. The primary tumor was 78% gastroenteropancreatic (GEP), 13% broncho-pulmonary and 9% of unknown origin. 35 patients (76.1%) with stable disease or partial response after PRRT were classified as responders and 11 (23.9%) as non-responders. Logistic regression analysis identified that baseline total volume (bTV) was associated with therapy outcome (OR 1.17; 95%CI 1.02–1.32; p = 0.02). No significant association with PRRT response was observed for other variables. High bTV was confirmed as the only variable independently associated with OS (HR 12.76, 95%CI 1.53–107, p = 0.01). In conclusion, high bTV is a negative predictor for PRRT response and is associated with worse OS rates. Early iPET during PRRT apparently does not provide information useful to change the management of NET patients.
Collapse
Affiliation(s)
- Rexhep Durmo
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
- PhD Program in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-0522296284
| | - Angelina Filice
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| | - Federica Fioroni
- Medical Physics Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (F.F.); (D.F.)
| | - Veronica Cervati
- Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy;
| | - Domenico Finocchiaro
- Medical Physics Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (F.F.); (D.F.)
| | - Chiara Coruzzi
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| | - Giulia Besutti
- Radiology Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Silvia Fanello
- Medical Oncology Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Andrea Frasoldati
- Department of Endocrinology and Metabolism, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Annibale Versari
- Nuclear Medicine Unit, Azienda USL-IRCCS of Reggio Emilia, 42123 Reggio Emilia, Italy; (A.F.); (C.C.); (A.V.)
| |
Collapse
|
50
|
Peptide Receptor Radionuclide Therapy with [ 177Lu]Lu-DOTA-TATE in Patients with Advanced GEP NENS: Present and Future Directions. Cancers (Basel) 2022; 14:cancers14030584. [PMID: 35158852 PMCID: PMC8833790 DOI: 10.3390/cancers14030584] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Neuroendocrine neoplasms have been usually described as infrequent tumors, but their incidence has been rising over time. [177Lu]Lu-DOTA-TATE (PRRT-Lu) was approved by the European Medicines Agency and by the Food and Drug Administration as the first radiopharmaceutical for peptide receptor radionuclide therapy in progressive gastroenteropancreatic NET. PRRT-Lu is considered a therapeutic option in progressive SSTR-positive NETs with homogenous SSTR expression. The NETTER-1 study demonstrated that PRRT-Lu yielded a statistically and clinically significant improvement in PFS as a primary endpoint (HR: 0.18, p < 0.0001), as well as a clinical trend towards improvement in OS. These results made scientific societies incorporate PRRT-Lu into their clinical guidelines; however, some questions still remain unanswered. Abstract This review article summarizes findings published in the last years on peptide receptor radionuclide therapy in GEP NENs, as well as potential future developments and directions. Unanswered questions remain, such as the following: Which is the correct dose and individual dosimetry? Which is the place for salvage PRRT-Lu? Whicht is the role of PRRT-Lu in the pediatric population? Which is the optimal sequencing of PRRT-Lu in advanced GEP NETs? Which is the place of PRRT-Lu in G3 NENs? These, and future developments such as inclusion new radiopharmaceuticals and combination therapy with different agents, such as radiosensitizers, will be discussed.
Collapse
|