1
|
Sunny SS, Hephzibah J, Chacko RT, Kodiatte TA. Therapeutic Response to PRRNT in a Rare Case of Metastatic Renal Neuroendocrine Carcinoma. World J Nucl Med 2024; 23:141-146. [PMID: 38933067 PMCID: PMC11199026 DOI: 10.1055/s-0044-1785461] [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] [Indexed: 06/28/2024] Open
Abstract
Neuroendocrine tumors (NETs) are a rare spectrum of neoplasms that are characterized by neuroendocrine and neural differentiation. The treatment can be challenging in view of the heterogeneity in differentiation and behavior. Primary renal origin NETs are rare and only a few cases have been reported in the literature. There is limited knowledge on their presentation and response to various lines of treatment. We report a case of a patient with a metastatic renal NET from a rare histological subtype of large cell neuroendocrine carcinoma, known to cause aggressive disease with poor prognosis. A multimodality treatment approach was followed. In spite of surgical management and second-line chemotherapy, the disease progressed. The patient subsequently received peptide receptor radionuclide therapy (PRRNT) using lutetium-177 DOTATATE, following which the patient demonstrated a remarkable clinical and radiological response and is stable to date. In a rare tumor with poor prognosis, the relevance of theranostics and the efficacy of targeted therapies like PRRNT are noteworthy.
Collapse
Affiliation(s)
- Saumya Sara Sunny
- Department of Nuclear Medicine, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Julie Hephzibah
- Department of Medical Oncology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Raju Titus Chacko
- Department of Medical Oncology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Thomas Alex Kodiatte
- Department of Pathology, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| |
Collapse
|
2
|
Ali MU, Chaudhary BN, Panja S, Gendelman HE. Theranostic Diagnostics. Results Probl Cell Differ 2024; 73:551-578. [PMID: 39242393 DOI: 10.1007/978-3-031-62036-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Diagnosing and then treating disease defines theranostics. The approach holds promise by facilitating targeted disease outcomes. The simultaneous analysis of finding the presence of disease pathophysiology while providing a parallel in treatment is a novel and effective strategy for seeking improved medical care. We discuss how theranostics improves disease outcomes is discussed. The chapter reviews the delivery of targeted therapies. Bioimaging techniques are highlighted as early detection and tracking systems for microbial infections, degenerative diseases, and cancers.
Collapse
Affiliation(s)
- Mohammad Uzair Ali
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bharat N Chaudhary
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
3
|
Murce E, de Blois E, van den Berg S, de Jong M, Seimbille Y. Synthesis and radiolabelling of PSMA-targeted derivatives containing GYK/MVK cleavable linkers. ROYAL SOCIETY OPEN SCIENCE 2023; 10:220950. [PMID: 36908985 PMCID: PMC9993039 DOI: 10.1098/rsos.220950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Targeted radionuclide therapy (TRT) is a promising strategy to treat different types of cancer. TRT relies on a targeting vector used to deliver a therapeutic radionuclide specifically to the tumour site. Several low molecular weight ligands targeting the prostate-specific membrane antigen (PSMA) have been synthesized, but their pharmacokinetic properties still need to be optimized. Hereby, we describe the synthesis of new conjugates, featuring the cleavable linkers Gly-Tyr-Lys (GYK) and Met-Val-Lys (MVK), to reduce the dose delivered to the kidneys. Compounds were synthesized by solid-phase peptide synthesis (SPPS) and obtained in greater than 95% chemical purity. Radiolabelling was performed with both In-111 and Lu-177 to validate potential use of the compounds as both imaging and therapeutic agents. Radiochemical purity greater than 80% was obtained for both nuclides, but significant radiolysis was observed for the methionine-containing analogue. The results obtained thus far with the GYK-PSMA conjugate could warrant further biological investigations.
Collapse
Affiliation(s)
- Erika Murce
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sophie van den Berg
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Marion de Jong
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- TRIUMF, Life Sciences Division, Vancouver, Canada
| |
Collapse
|
4
|
Zhao F, Huang L, Wang Z, Wei F, Xiao T, Liu Q. Epidemiological trends and novel prognostic evaluation approaches of patients with stage II-IV colorectal neuroendocrine neoplasms: A population-based study with external validation. Front Endocrinol (Lausanne) 2023; 14:1061187. [PMID: 36817582 PMCID: PMC9928741 DOI: 10.3389/fendo.2023.1061187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE This study aimed to clarify the incidence trend of all-stage colorectal neuroendocrine neoplasms (CRNENs), overall survival (OS), and disease-specific survival (DSS) of patients with stage II-IV CRNENs, and to establish relevant nomograms for risk stratification. METHODS Among all patients diagnosed with CRNENs in the Surveillance, Epidemiology, and End Results (SEER) database from 1975 to 2019, temporal trends in incidence were assessed. Clinical data of 668 patients with stage II-IV CRNENs from 2010 to 2016 were extracted for survival analysis. Patients were randomly divided into a training cohort and a validation cohort at a ratio of 7:3. Univariate and multivariate cox regression analyses were utilized to identify independent prognostic factors affecting OS outcomes. Competing risk analysis was applied to investigate risk factors related to the DSS of CRNENs. Two nomograms specifically for OS and DSS were developed for patients with stage II-IV CRNENs, their prognostic capabilities were evaluated using calibration curves, receiver operating characteristic (ROC) curves, the time-dependent area under the curve (AUC), and decision-curve analysis (DCA). Our hospital's independent cohort of 62 patients with CRNENs was used as the external validation cohort. RESULTS In the period of 1975-2019, the incidence of CRNENs increased steadily with an annual percentage change (APC) of 4.50 (95% confidence interval [CI]: 3.90-5.11, P < 0.05). In total, 668 patients with stage II-IV CRNENs were included in the survival analysis from 2010 and 2016. Independent adverse prognostic factors for both OS and DSS of CRNENs prior treatment included grade III/IV (HR for OS: 4.66, 95%CI: 2.92-7.42; HR for DSS: 4.79, 95%CI: 4.27-5.31), higher TNM stage ([stage III vs stage II] HR for OS: 2.22, 95%CI: 1.25-3.94; HR for DSS: 2.69, 95%CI: 1.96-3.42. [stage IV vs stage II] HR for OS: 3.99, 95%CI: 2.03-7.83; HR for DSS: 4.96, 95%CI: 4.14-5.78), liver metastasis (HR for OS: 1.61, 95%CI: 1.03-2.51; HR for DSS: 1.86, 95%CI: 1.39-2.32), and brain metastasis (HR for OS: 4.57, 95%CI: 1.66-12.58; HR for DSS: 5.01, 95%CI: 4.15-5.87). Advanced age was also identified as a risk factor for OS (HR: 2.03, 95%CI: 1.5-2.76) but not DSS. In terms of treatment, surgery can significantly prolong OS (HR: 0.62, 95%CI: 0.44-0.86) and DSS (HR: 0.67, 95%CI: 0.29-1.05), but chemotherapy and radiation failed to show significance. The respective nomograms for OS and DSS for stage II-IV CRNENs demonstrated high accuracy and robust prediction value in predicting 1-year, 3-year, and 5-year OS and DSS outcomes in training, internal validation, and external validation cohorts. Besides, two online tools regarding OS and DSS prediction were established, facilitating nomogram score calculation, risk group determination, as well as survival prediction for each individual patient. CONCLUSION Over the past 40 years, the incidence of CRNENs presented increased steadily, along with improved survival outcomes. Grade III-IV, higher TNM stage, liver metastasis, brain metastasis, and without receiving surgery were found to be associated with worse OS and DSS. Advanced age was a risk factor for OS but not DSS. Nomograms for patients with stage II-IV stage CRNENs are capable of predicting the 1-, 3-, and 5-year OS and DSS rates with high accuracy, and realize risk stratification.
Collapse
Affiliation(s)
- Fuqiang Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liling Huang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Zhijie Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangze Wei
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tixian Xiao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Qian Liu,
| |
Collapse
|
5
|
Grzmil M, Wiesmann F, Schibli R, Behe M. Targeting mTORC1 Activity to Improve Efficacy of Radioligand Therapy in Cancer. Cancers (Basel) 2022; 15:cancers15010017. [PMID: 36612012 PMCID: PMC9817840 DOI: 10.3390/cancers15010017] [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: 11/16/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Radioligand therapy (RLT) represents an effective strategy to treat malignancy by cancer-selective delivery of radioactivity following systemic application. Despite recent therapeutic successes, cancer radioresistance and insufficient delivery of the radioactive ligands, as well as cytotoxicity to healthy organs, significantly impairs clinical efficacy. To improve disease management while minimizing toxicity, in recent years, the combination of RLT with molecular targeted therapies against cancer signaling networks showed encouraging outcomes. Characterization of the key deregulated oncogenic signaling pathways revealed their convergence to activate the mammalian target of rapamycin (mTOR), in which signaling plays an essential role in the regulation of cancer growth and survival. Therapeutic interference with hyperactivated mTOR pathways was extensively studied and led to the development of mTOR inhibitors for clinical applications. In this review, we outline the regulation and oncogenic role of mTOR signaling, as well as recapitulate and discuss mTOR complex 1 (mTORC1) inhibition to improve the efficacy of RLT in cancer.
Collapse
Affiliation(s)
- Michal Grzmil
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen, Switzerland
- Correspondence:
| | - Fabius Wiesmann
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Martin Behe
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen, Switzerland
| |
Collapse
|
6
|
Dietze MMA, de Jong HWAM. Progress in large field-of-view interventional planar scintigraphy and SPECT imaging. Expert Rev Med Devices 2022; 19:393-403. [PMID: 35695477 DOI: 10.1080/17434440.2022.2088355] [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: 11/04/2022]
Abstract
INTRODUCTION Handheld gamma cameras and gamma probes have been successfully implemented for enabling nuclear image or radio-guidance in minimally-invasive procedures. There is an opportunity for large field-of-view interventional planar scintigraphy and SPECT imaging to complement these small field-of-view devices for two reasons. First, a large field-of-view camera enables imaging of relatively larger organs and activity accumulations that are not close to the patient's skin. And second, more precise corrections can be implemented in the SPECT reconstruction algorithm, improving its quality. AREAS COVERED This review article discusses the progress that has been made in the field of large field-of-view interventional planar scintigraphy and SPECT imaging. First, an overview of planar scintigraphy and SPECT is provided. Second, an exploration is given of the potential applications where large field-of-view interventional planar scintigraphy and SPECT imaging may be employed. And third, the requirements for scanner hardware are discussed and an overview of the possible system configurations is provided. EXPERT OPINION We believe that there is an opportunity for large field-of-view interventional planar scintigraphy and SPECT imaging to assist clinical workflows. A major effort is now required to evaluate the prototype systems in clinical studies so that valuable practical experience can be obtained.
Collapse
Affiliation(s)
- Martijn M A Dietze
- Radiology and Nuclear Medicine, Utrecht University and University Medical Center, Utrecht, Netherlands
| | - Hugo W A M de Jong
- Radiology and Nuclear Medicine, Utrecht University and University Medical Center, Utrecht, Netherlands
| |
Collapse
|
7
|
Desai P, Rimal R, Sahnoun SEM, Mottaghy FM, Möller M, Morgenroth A, Singh S. Radiolabeled Nanocarriers as Theranostics-Advancement from Peptides to Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200673. [PMID: 35527333 DOI: 10.1002/smll.202200673] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/15/2022] [Indexed: 06/14/2023]
Abstract
Endogenous targeted radiotherapy is emerging as an integral modality to treat a variety of cancer entities. Nevertheless, despite the positive clinical outcome of the treatment using radiolabeled peptides, small molecules, antibodies, and nanobodies, a high degree of hepatotoxicity and nephrotoxicity still persist. This limits the amount of dose that can be injected. In an attempt to mitigate these side effects, the use of nanocarriers such as nanoparticles (NPs), dendrimers, micelles, liposomes, and nanogels (NGs) is currently being explored. Nanocarriers can prolong circulation time and tumor retention, maximize radiation dosage, and offer multifunctionality for different targeting strategies. In this review, the authors first provide a summary of radiation therapy and imaging and discuss the new radiotracers that are used preclinically and clinically. They then highlight and identify the advantages of radio-nanomedicine and its potential in overcoming the limitations of endogenous radiotherapy. Finally, the review points to the ongoing efforts to maximize the use of radio-nanomedicine for efficient clinical translation.
Collapse
Affiliation(s)
- Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Sabri E M Sahnoun
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
- Department of Radiology and Nuclear Medicine, School for Cardiovascular Diseases (CARIM) and School of oncology (GROW), Maastricht University, Maastricht, 6229 HX, The Netherlands
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University hospital RWTH Aachen, Pauwelstraße 30, 52074, Aachen, Germany
| | - Smriti Singh
- DWI Leibniz Institute for Interactive Materials e.V, RWTH Aachen University, Forckenbeckstrasse 50, 52074, Aachen, Germany
- Max-Planck-Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| |
Collapse
|
8
|
Khatami A, Sistani G, Sutherland DEK, DeBrabandere S, Reid RH, Laidley DT. Toxicity and Tolerability of 177Lu-DOTA-TATE PRRT with a Modified Administered Activity Protocol in NETs of Variable Origin - A Phase 2 Registry Study. Curr Radiopharm 2022; 15:123-133. [PMID: 35135467 PMCID: PMC9900697 DOI: 10.2174/1874471014666210810100435] [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: 03/04/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Peptide receptor radionuclide therapy (PRRT) has been recently approved for advanced, metastatic, or progressive neuroendocrine tumors (NETs). OBJECTIVE This study reports the adverse events (AEs) observed with patient-tailored administered activity. METHODS Fifty-two PRRT naive patients were treated with 177Lu-DOTATATE. The administered activity ranges between 2.78 and 5.55 GBq/cycle using the patient's unique characteristics (age, symptoms, blood work, and biomarkers). RESULTS The protocol was well tolerated with the overwhelming majority of participants being forty- six (88%), completing all 4 induction therapy cycles. The median cumulative administered activity was 19.6 GBq (ranged 3.8-22.3 GBq). A total of 42/52 (81%) reported at least one symptom, and 43/52 (83%) had evidence of biochemical abnormality at enrollment that would meet grade 1 or 2 criteria for AEs. These symptoms only slightly increase with treatment to 50/52 (96%) and 51/52 (98%), respectively. The most common symptoms were mild fatigue (62%), shortness of breath (50%), nausea (44%), abdominal pain (38%), and musculoskeletal pain (37%). The most common biomarker abnormalities were mild anemia (81%), reduced estimated glomerular filtration rate (eGFR) (58%), increased alkaline phosphatase (ALP) (50%), and leukopenia (37%). Of critical importance, no 177Lu-DOTATATE related grade 3 or 4 AEs were observed. CONCLUSION Tailoring the administered activity of 177Lu-DOTATATE to the individual patient with a variety of NETs is both safe and well-tolerated. No patient developed severe grade 3 or 4 AEs. Most patients exhibit symptoms or biochemical abnormality before treatment and this only slightly worsens following induction therapy.
Collapse
Affiliation(s)
- Alireza Khatami
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada; ,Address correspondence to this author at the Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada; E-mail:
| | - Golmehr Sistani
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada; ,Division of Radiology, Department of Medical Imaging, Western University, London, ON, Canada
| | - Duncan E. K. Sutherland
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada;
| | - Sarah DeBrabandere
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada;
| | - Robert H. Reid
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada;
| | - David T. Laidley
- Division of Nuclear Medicine, Department of Medical Imaging, Western University, London, ON N645C1, Canada;
| |
Collapse
|
9
|
Maina T, Nock BA. Gamma camera imaging by radiolabeled gastrin/cholecystokinin analogs. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00183-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
10
|
Maina T, Nock BA. Peptide radiopharmaceuticals for targeted diagnosis & therapy of human tumors. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
11
|
Emanuel O, Liu J, Schartinger VH, Nei WL, Chan YY, Tsang CM, Riechelmann H, Masterson L, Haybaeck J, Oppermann U, Willems SM, Ooft ML, Wollmann G, Howard D, Vanhaesebroeck B, Lund VJ, Royle G, Chua MLK, Lo KW, Busson P, Lechner M. SSTR2 in Nasopharyngeal Carcinoma: Relationship with Latent EBV Infection and Potential as a Therapeutic Target. Cancers (Basel) 2021; 13:4944. [PMID: 34638429 PMCID: PMC8508244 DOI: 10.3390/cancers13194944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 01/04/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant epithelial tumor, most commonly located in the pharyngeal recess and endemic to parts of Asia. It is often detected at a late stage which is associated with poor prognosis (5-year survival rate of 63%). Treatment for this malignancy relies predominantly on radiotherapy and/or systemic chemotherapy, which can be associated with significant morbidity and impaired quality of life. In endemic regions NPC is associated with infection by Epstein-Barr virus (EBV) which was shown to upregulate the somatostatin receptor 2 (SSTR2) cell surface receptor. With recent advances in molecular techniques allowing for an improved understanding of the molecular aetiology of this disease and its relation to SSTR2 expression, we provide a comprehensive and up-to-date overview of this disease and highlight the emergence of SSTR2 as a key tumor biomarker and promising target for imaging and therapy.
Collapse
Affiliation(s)
- Oscar Emanuel
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
| | - Jacklyn Liu
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
| | - Volker H. Schartinger
- Department of Otorhinolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (V.H.S.); (H.R.)
| | - Wen Long Nei
- National Cancer Centre, Divisions of Radiation Oncology and Medical Sciences, Singapore 169610, Singapore; (W.L.N.); (M.L.K.C.)
- Oncology Academic Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Yuk Yu Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China; (Y.Y.C.); (C.M.T.); (K.W.L.)
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China; (Y.Y.C.); (C.M.T.); (K.W.L.)
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Herbert Riechelmann
- Department of Otorhinolaryngology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (V.H.S.); (H.R.)
| | - Liam Masterson
- Department of Otolaryngology, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Udo Oppermann
- Botnar Research Centre, University of Oxford, Oxford OX1 2JD, UK;
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79085 Freiburg, Germany
| | - Stefan M. Willems
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (S.M.W.); (M.L.O.)
- Department of Pathology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Marc L. Ooft
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (S.M.W.); (M.L.O.)
- King’s College Hospitals, NHS Foundation Trust, London SE5 9RS, UK
| | - Guido Wollmann
- Institute of Virology and Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - David Howard
- ENT Department, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London W6 9EP, UK;
- Royal National Throat, Nose and Ear Hospital, University College London Hospitals NHS Trust, London WC1E 6DG, UK
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
| | - Valerie J. Lund
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
- Royal National Throat, Nose and Ear Hospital, University College London Hospitals NHS Trust, London WC1E 6DG, UK
| | - Gary Royle
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
| | - Melvin L. K. Chua
- National Cancer Centre, Divisions of Radiation Oncology and Medical Sciences, Singapore 169610, Singapore; (W.L.N.); (M.L.K.C.)
- Oncology Academic Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong 999077, China; (Y.Y.C.); (C.M.T.); (K.W.L.)
- State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Pierre Busson
- CNRS-UMR 9018-Metsy, Gustave Roussy and Université Paris-Saclay, 94805 Villejuif, France
| | - Matt Lechner
- UCL Cancer Institute, University College London, London WC1E 6BT, UK; (O.E.); (J.L.); (B.V.); (V.J.L.); (G.R.)
- Rhinology & Endoscopic Skull Base Surgery, Department of Otolaryngology-H&N Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| |
Collapse
|
12
|
PI3K/mTOR Dual Inhibitor PF-04691502 Is a Schedule-Dependent Radiosensitizer for Gastroenteropancreatic Neuroendocrine Tumors. Cells 2021; 10:cells10051261. [PMID: 34065268 PMCID: PMC8160730 DOI: 10.3390/cells10051261] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/27/2022] Open
Abstract
Patients with advanced-stage gastroenteropancreatic neuroendocrine tumors (GEP-NETs) have a poor overall prognosis despite chemotherapy and radiotherapy (e.g., peptide receptor radionuclide therapy (PRRT)). Better treatment options are needed to improve disease regression and patient survival. The purpose of this study was to examine a new treatment strategy by combining PI3K/mTOR dual inhibition and radiotherapy. First, we assessed the efficacy of two PI3K/mTOR dual inhibitors, PF-04691502 and PKI-402, to inhibit pAkt and increase apoptosis in NET cell lines (BON and QGP-1) and patient-derived tumor spheroids as single agents or combined with radiotherapy (XRT). Treatment with PF-04691502 decreased pAkt (Ser473) expression for up to 72 h compared with the control; in contrast, decreased pAkt expression was noted for less than 24 h with PKI-402. Simultaneous treatment with PF-04691502 and XRT did not induce apoptosis in NET cells; however, the addition of PF-04691502 48 h after XRT significantly increased apoptosis compared to PF-04691502 or XRT treatment alone. Our results demonstrate that schedule-dependent administration of a PI3K/mTOR inhibitor, combined with XRT, can enhance cytotoxicity by promoting the radiosensitivity of NET cells. Moreover, our findings suggest that radiotherapy, in combination with timed PI3K/mTOR inhibition, may be a promising therapeutic regimen for patients with GEP-NET.
Collapse
|
13
|
Chemotherapy-Induced Upregulation of Somatostatin Receptor-2 Increases the Uptake and Efficacy of 177Lu-DOTA-Octreotate in Neuroendocrine Tumor Cells. Cancers (Basel) 2021; 13:cancers13020232. [PMID: 33435224 PMCID: PMC7828052 DOI: 10.3390/cancers13020232] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary The peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTA-octreotate (LuTate) is recommended for neuroendocrine tumors (NETs) which overexpress somatostatin receptors (SSTR). A combination of LuTate with chemotherapy improves its objective response in NET patients, and here we characterized chemotherapy-induced upregulation of SSTR2 receptors as a cause for this improved response to LuTate. Using multiple NET and non-NET cell lines, we examined the SSTR2 expression for up to 7 days after exposure to drugs and its effect on LuTate uptake and cell proliferation. We report that the exposure to varying doses of chemotherapeutic drugs such as temozolomide for 24 h or 5 days results in upregulation of SSTR2 receptors between 3–7 days. This effect is more pronounced in low SSTR2 expressing BON-1 cells than in high SSTR2 expressing NCI-H727 or non-NET cancer or non-cancer cells. Thus, a properly-timed pre-treatment with low doses of chemotherapy could improve therapeutic efficacy of LuTate in NET patients. Abstract The peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTA-octreotate (LuTate) is recommended for different types of neuroendocrine tumors (NETs) which overexpress somatostatin receptors (SSTR). A combination with chemotherapy improves objective response to LuTate in NET patients and here we characterized chemotherapy-induced upregulation of SSTR2 receptors as a cause for this improved response to LuTate. The NET cell lines with low (BON-1) or relatively high (NCI-H727) SSTR2-expression levels, and non-NET cancer and normal cells were treated with chemotherapeutic drugs and assessed for upregulation of SSTR2. We report that an exposure to low or high doses of drugs, such as temozolomide for 24 h or 5 day results in upregulation of SSTR2 between 3–7 days, increased LuTate uptake and decreased rate of cell proliferation. This effect is at the level of SSTR2-mRNA and is more pronounced in low SSTR2 expressing BON-1 than in high SSTR2 expressing NCI-H727 or non-NET cancer or normal cells. Thus, a properly timed pre-treatment with low-dose chemotherapy could not only improve therapeutic efficacy of LuTate in NET patients who are presently eligible for PRRT, but also allow PRRT to be administered to patients with low SSTR-expressing NETs, who would otherwise not respond to this modality because of insufficient radiation delivery.
Collapse
|
14
|
Efficacy of 177Lu-Dotatate Induction and Maintenance Therapy of Various Types of Neuroendocrine Tumors: A Phase II Registry Study. ACTA ACUST UNITED AC 2020; 28:115-127. [PMID: 33622997 PMCID: PMC7816182 DOI: 10.3390/curroncol28010015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/22/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) has been recently established as a treatment option for progressive gastro-entero-pancreatic neuroendocrine tumors (NETs) including four 200 mCi induction cycles. The purpose of this phase 2 trial is to expand use of PRRT to different types of NETs with the application of dose adjustment and evaluate value of maintenance therapy in patients who had disease control on induction therapy. Forty-seven PRRT naïve NET patients with different primary origin received 177Lu-DOTATATE induction therapy, ranging from 75 to 150 mCi per cycle, based on patients’ clinical status such as liver and renal function, extent of metastases, and previous therapies. Thirty-four patients underwent additional maintenance therapy (50–100 mCi per cycle) following induction course until they developed disease progression. The estimated median progression-free survival (PFS) was 36.1 months. The median PFS in our MNET subgroup was 47.7 months, which is markedly longer than NETTER-1 trial with median PFS of 28.4 months. The median PFS was significantly longer in patients who received PRRT as first-line treatment after disease progression on somatostatin analogs compared to patients who received other therapies first (p-value = 0.04). The total disease response rate (DRR) and disease control rate (DCR) was 32% and 85% based on RECIST 1.1 and 45% and 83% based on Choi criteria. This trial demonstrates longer PFS with the addition of low dose maintenance therapy to induction therapy compared to NETTER-1 trial that only included induction therapy. Also, we observed considerable efficacy of PRRT in various types of advanced NETs.
Collapse
|
15
|
Abstract
Neuroendocrine neoplasms are a heterogeneous group of tumors arising from cells distributed throughout the body. Local and regional disease is managed with surgical resection; however, treatment of higher-grade neuroendocrine tumors (NETs), unresectable or metastatic disease is complex involving a combination of systemic targeted agents, transarterial embolization, and peptide receptor targeted therapies and is discussed in detail. The most important concept in modern NET workup is that an optimal diagnostic strategy requires combination of both anatomic and functional imaging modalities. NETs often present with unknown primary site of disease, and 68Ga-DOTATATE PET can now diagnose these lesions with great sensitivity.
Collapse
Affiliation(s)
- Agata E Migut
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Harmeet Kaur
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Rony Avritscher
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA; Department of Interventional Radiology, 1400 Pressler Street, Unit 1471, Houston, TX 77030, USA.
| |
Collapse
|
16
|
Abbasi Gharibkandi N, Conlon JM, Hosseinimehr SJ. Strategies for improving stability and pharmacokinetic characteristics of radiolabeled peptides for imaging and therapy. Peptides 2020; 133:170385. [PMID: 32822772 DOI: 10.1016/j.peptides.2020.170385] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Tumor cells overexpress a variety of receptors that are emerging targets in cancer chemotherapy. Radiolabeled peptides with high affinity and selectivity for these overexpressed receptors have been designed for both imaging and therapy purposes. Such peptides display advantages such as high selectivity for tumor cells, rapid tumor tissue penetration, and rapid clearance from non-target tissues and the circulation. However, the very short in vivo half-life of radiolabeled peptides, arising from enzymatic degradation and/or efficient clearance by the kidney, limits their accumulation in tumors. This review presents various strategies that have been applied to extend the half-life extension and improve the pharmacokinetic characteristics of radiolabeled peptides. These include amino acid substitution, modification of the peptide termini, dimerization and multimerization of the peptide, cyclization, conjugation with polymers, sugars and albumin and use of peptidase inhibitors.
Collapse
Affiliation(s)
- Nasrin Abbasi Gharibkandi
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - J Michael Conlon
- Diabetes Research Group, School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine, Northern Ireland, BT52 1SA, UK
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
17
|
Albrecht J, Exner S, Grötzinger C, Prasad S, Konietschke F, Beindorff N, Kühl AA, Prasad V, Brenner W, Koziolek EJ. Multimodal Imaging of 2-Cycle PRRT with 177Lu-DOTA-JR11 and 177Lu-DOTATOC in an Orthotopic Neuroendocrine Xenograft Tumor Mouse Model. J Nucl Med 2020; 62:393-398. [PMID: 32859703 DOI: 10.2967/jnumed.120.250274] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using radiolabeled somatostatin receptor (SSTR) analogs is a common approach in advanced neuroendocrine neoplasms. Recently, SSTR antagonists have shown promising results for imaging and therapy due to a higher number of binding sites than in commonly used agonists. We evaluated PRRT with SSTR agonist 177Lu-DOTATOC and antagonist 177Lu-DOTA-JR11 longitudinally in an orthotopic murine pancreatic neuroendocrine neoplasm model expressing human SSTR2. Morphologic and metabolic changes during treatment were assessed using multimodal imaging, including hybrid PET/MRI and SPECT/CT. Methods: In vitro radioligand binding and internalization assays and cell-cycle analysis were performed. SSTR2-transfected BON cells (BON-SSTR2) were used for in vivo experiments. Tumor-bearing mice received 2 intravenous injections of 100 μL of saline, 30 MBq of 177Lu-DOTATOC, or 20 MBq of 177Lu-DOTA-JR11 with an interval of 3 wk. Weekly T2-weighted MRI was performed for tumor monitoring. Viability of the tumor tissue was assessed by 18F-FDG PET/MRI once after PRRT. Tumor and kidney uptake of the respective radiopharmaceuticals was measured 24 h after injection by SPECT/CT. Results: Compared with 177Lu-DOTATOC, 177Lu-DOTA-JR11 treatment resulted in an increased accumulation of cells in G2/M phase. Animals treated with the SSTR antagonist showed a significant reduction in tumor size (P < 0.001) and an increased median survival (207 d; interquartile range [IQR], 132-228) compared with 177Lu-DOTATOC (126 d; IQR, 118-129). SPECT/CT revealed a 4-fold higher median tumor uptake for the antagonist and a 3-fold higher tumor-to-kidney ratio in the first treatment cycle. During the second therapy cycle, tumor uptake of 177Lu-DOTATOC was significantly lower (P = 0.01) whereas 177Lu-DOTA-JR11 uptake remained stable. Imaging of tumor morphology indicated comparatively larger necrotic fractions for 177Lu-DOTA-JR11 despite further tumor growth. These results were confirmed by 18F-FDG PET, revealing the least amount of viable tumor tissue in 177Lu-DOTA-JR11-treated animals, at 6.2% (IQR, 2%-23%). Conclusion: 177Lu-DOTA-JR11 showed a higher tumor-to-kidney ratio and a more pronounced cytotoxic effect than did 177Lu-DOTATOC. Additionally, tumor uptake was more stable over the course of 2 treatment cycles.
Collapse
Affiliation(s)
- Jakob Albrecht
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Samantha Exner
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Grötzinger
- German Cancer Consortium, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sonal Prasad
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Konietschke
- Institute of Biometry and Clinical Epidemiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin-Immunopathology for Experimental Models, Charité-Universitätsmedizin Berlin; and
| | - Vikas Prasad
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Nuclear Medicine, University Hospital Ulm, Ulm, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany .,German Cancer Consortium, Berlin, Germany.,Berlin Experimental Radionuclide Imaging Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Eva J Koziolek
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
18
|
Hoppenz P, Els-Heindl S, Beck-Sickinger AG. Peptide-Drug Conjugates and Their Targets in Advanced Cancer Therapies. Front Chem 2020; 8:571. [PMID: 32733853 PMCID: PMC7359416 DOI: 10.3389/fchem.2020.00571] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer became recently the leading cause of death in industrialized countries. Even though standard treatments achieve significant effects in growth inhibition and tumor elimination, they cause severe side effects as most of the applied drugs exhibit only minor selectivity for the malignant tissue. Hence, specific addressing of tumor cells without affecting healthy tissue is currently a major desire in cancer therapy. Cell surface receptors, which bind peptides are frequently overexpressed on cancer cells and can therefore be considered as promising targets for selective tumor therapy. In this review, the benefits of peptides as tumor homing agents are presented and an overview of the most commonly addressed peptide receptors is given. A special focus was set on the bombesin receptor family and the neuropeptide Y receptor family. In the second part, the specific requirements of peptide-drug conjugates (PDC) and intelligent linker structures as an essential component of PDC are outlined. Furthermore, different drug cargos are presented including classical and recent toxic agents as well as radionuclides for diagnostic and therapeutic approaches. In the last part, boron neutron capture therapy as advanced targeted cancer therapy is introduced and past and recent developments are reviewed.
Collapse
Affiliation(s)
- Paul Hoppenz
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | - Sylvia Els-Heindl
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
19
|
Molecular imaging of inflammation - Current and emerging technologies for diagnosis and treatment. Pharmacol Ther 2020; 211:107550. [PMID: 32325067 DOI: 10.1016/j.pharmthera.2020.107550] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
Inflammation is a key factor in multiple diseases including primary immune-mediated inflammatory diseases e.g. rheumatoid arthritis but also, less obviously, in many other common conditions, e.g. cardiovascular disease and diabetes. Together, chronic inflammatory diseases contribute to the majority of global morbidity and mortality. However, our understanding of the underlying processes by which the immune response is activated and sustained is limited by a lack of cellular and molecular information obtained in situ. Molecular imaging is the visualization, detection and quantification of molecules in the body. The ability to reveal information on inflammatory biomarkers, pathways and cells can improve disease diagnosis, guide and monitor therapeutic intervention and identify new targets for research. The optimum molecular imaging modality will possess high sensitivity and high resolution and be capable of non-invasive quantitative imaging of multiple disease biomarkers while maintaining an acceptable safety profile. The mainstays of current clinical imaging are computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US) and nuclear imaging such as positron emission tomography (PET). However, none of these have yet progressed to routine clinical use in the molecular imaging of inflammation, therefore new approaches are required to meet this goal. This review sets out the respective merits and limitations of both established and emerging imaging modalities as clinically useful molecular imaging tools in addition to potential theranostic applications.
Collapse
|
20
|
Agnew HD, Coppock MB, Idso MN, Lai BT, Liang J, McCarthy-Torrens AM, Warren CM, Heath JR. Protein-Catalyzed Capture Agents. Chem Rev 2019; 119:9950-9970. [PMID: 30838853 DOI: 10.1021/acs.chemrev.8b00660] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protein-catalyzed capture agents (PCCs) are synthetic and modular peptide-based affinity agents that are developed through the use of single-generation in situ click chemistry screens against large peptide libraries. In such screens, the target protein, or a synthetic epitope fragment of that protein, provides a template for selectively promoting the noncopper catalyzed azide-alkyne dipolar cycloaddition click reaction between either a library peptide and a known ligand or a library peptide and the synthetic epitope. The development of epitope-targeted PCCs was motivated by the desire to fully generalize pioneering work from the Sharpless and Finn groups in which in situ click screens were used to develop potent, divalent enzymatic inhibitors. In fact, a large degree of generality has now been achieved. Various PCCs have demonstrated utility for selective protein detection, as allosteric or direct inhibitors, as modulators of protein folding, and as tools for in vivo tumor imaging. We provide a historical context for PCCs and place them within the broader scope of biological and synthetic aptamers. The development of PCCs is presented as (i) Generation I PCCs, which are branched ligands engineered through an iterative, nonepitope-targeted process, and (ii) Generation II PCCs, which are typically developed from macrocyclic peptide libraries and are precisely epitope-targeted. We provide statistical comparisons of Generation II PCCs relative to monoclonal antibodies in which the protein target is the same. Finally, we discuss current challenges and future opportunities of PCCs.
Collapse
Affiliation(s)
- Heather D Agnew
- Indi Molecular, Inc. , 6162 Bristol Parkway , Culver City , California 90230 , United States
| | - Matthew B Coppock
- Sensors and Electron Devices Directorate , U.S. Army Research Laboratory , Adelphi , Maryland 20783 , United States
| | - Matthew N Idso
- Institute for Systems Biology , 401 Terry Avenue North , Seattle , Washington 98109-5234 , United States
| | - Bert T Lai
- Indi Molecular, Inc. , 6162 Bristol Parkway , Culver City , California 90230 , United States
| | - JingXin Liang
- Institute for Systems Biology , 401 Terry Avenue North , Seattle , Washington 98109-5234 , United States
| | - Amy M McCarthy-Torrens
- Institute for Systems Biology , 401 Terry Avenue North , Seattle , Washington 98109-5234 , United States
| | - Carmen M Warren
- Indi Molecular, Inc. , 6162 Bristol Parkway , Culver City , California 90230 , United States
| | - James R Heath
- Institute for Systems Biology , 401 Terry Avenue North , Seattle , Washington 98109-5234 , United States
| |
Collapse
|
21
|
In Vivo Biokinetics of 177Lu-OPS201 in Mice and Pigs as a Model for Predicting Human Dosimetry. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:6438196. [PMID: 30733648 PMCID: PMC6348830 DOI: 10.1155/2019/6438196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/25/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022]
Abstract
Introduction 177Lu-OPS201 is a high-affinity somatostatin receptor subtype 2 antagonist for PRRT in patients with neuroendocrine tumors. The aim is to find the optimal scaling for dosimetry and to compare the biokinetics of 177Lu-OPS201 in animals and humans. Methods Data on biokinetics of 177Lu-OPS201 were analyzed in athymic nude Foxn1nu mice (28 F, weight: 26 ± 1 g), Danish Landrace pigs (3 F-1 M, weight: 28 ± 2 kg), and patients (3 F-1 M, weight: 61 ± 17 kg) with administered activities of 0.19–0.27 MBq (mice), 97–113 MBq (pigs), and 850–1086 MBq (patients). After euthanizing mice (up to 168 h), the organ-specific activity contents (including blood) were measured. Multiple planar and SPECT/CT scans were performed until 250 h (pigs) and 72 h (patients) to quantify the uptake in the kidneys and liver. Blood samples were taken up to 23 h (patients) and 300 h (pigs). In pigs and patients, kidney protection was applied. Time-dependent uptake data sets were created for each species and organ/tissue. Biexponential fits were applied to compare the biokinetics in the kidneys, liver, and blood of each species. The time-integrated activity coefficients (TIACs) were calculated by using NUKFIT. To determine the optimal scaling, several methods (relative mass scaling, time scaling, combined mass and time scaling, and allometric scaling) were compared. Results A fast blood clearance of the compound was observed in the first phase (<56 h) for all species. In comparison with patients, pigs showed higher liver retention. Based on the direct comparison of the TIACs, an underestimation in mice (liver and kidneys) and an overestimation in pigs' kidneys compared to the patient data (kidney TIAC: mice = 1.4 h, pigs = 7.7 h, and patients = 5.8 h; liver TIAC: mice = 0.7 h, pigs = 4.1 h, and patients = 5.3 h) were observed. Most similar TIACs were obtained by applying time scaling (mice) and combined scaling (pigs) (kidney TIAC: mice = 3.9 h, pigs = 4.8 h, and patients = 5.8 h; liver TIAC: mice = 0.9 h, pigs = 4.7 h, and patients = 5.3 h). Conclusion If the organ mass ratios between the species are high, the combined mass and time scaling method is optimal to minimize the interspecies differences. The analysis of the fit functions and the TIACs shows that pigs are better mimicking human biokinetics.
Collapse
|
22
|
Gosain R, Mukherjee S, Yendamuri SS, Iyer R. Management of Typical and Atypical Pulmonary Carcinoids Based on Different Established Guidelines. Cancers (Basel) 2018; 10:E510. [PMID: 30545054 PMCID: PMC6315766 DOI: 10.3390/cancers10120510] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Neuroendocrine tumors (NETs) are a group of malignancies that originated from neuroendocrine cells, with the most common sites being lungs and the gastrointestinal tract. Lung NETs comprise 25% of all lung malignancies. Small cell lung cancer is the most common form of lung NETs, and other rare forms include well-differentiated typical carcinoids (TCs) and poorly differentiated atypical carcinoids (ACs). Given the paucity of randomized studies, rational treatment is challenging. Therefore, it is recommended that these decisions be made using a multidisciplinary collaborative approach. Surgery remains the mainstay of treatment, when feasible. Following surgery, various guidelines offer different recommendations in the adjuvant setting. In this paper, we describe the adjuvant management of lung NETs, as recommended by different guidelines, and highlight their differences. In addition to that, we also discuss the management of metastatic lung NETS, including the use of peptide receptor radionucleotide therapy.
Collapse
Affiliation(s)
- Rohit Gosain
- Division of Hematology & Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo School of Medicine, Buffalo, NY 14203, USA.
| | - Sarbajit Mukherjee
- Division of Hematology & Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo School of Medicine, Buffalo, NY 14203, USA.
- Division of Hematology & Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | - Sai S Yendamuri
- Depart of Thoracic Surgery Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo School of Medicine, Buffalo, NY 14203, USA.
| | - Renuka Iyer
- Division of Hematology & Oncology, Roswell Park Comprehensive Cancer Center, University at Buffalo School of Medicine, Buffalo, NY 14203, USA.
| |
Collapse
|
23
|
Zhang C, Lin KS, Bénard F. Molecular Imaging and Radionuclide Therapy of Melanoma Targeting the Melanocortin 1 Receptor. Mol Imaging 2018; 16:1536012117737919. [PMID: 29182034 PMCID: PMC5714078 DOI: 10.1177/1536012117737919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Melanoma is a deadly disease at late metastatic stage, and early diagnosis and accurate staging remain the key aspects for managing melanoma. The melanocortin 1 receptor (MC1 R) is overexpressed in primary and metastatic melanomas, and its endogenous ligand, the α-melanocyte-stimulating hormone (αMSH), has been extensively studied for the development of MC1 R-targeted molecular imaging and therapy of melanoma. Natural αMSH is not well suited for this purpose due to low stability in vivo. Unnatural amino acid substitutions substantially stabilized the peptide, while cyclization via lactam bridge and metal coordination further improved binding affinity and stability. In this study, we summarized the development and the in vitro and in vivo characteristics of the radiolabeled αMSH analogues, including 99mTc-, 111In-, 67 Ga-, or 125I-labeled αMSH analogues for imaging with single-photon emission computed tomography; 68Ga-, 64Cu-, or 18F-labeled αMSH analogues for imaging with positron emission tomography; and 188Re-, 177Lu-, 90Y-, or 212Pb-labeled αMSH analogues for radionuclide therapy. These radiolabeled αMSH analogues showed promising results with high tumor uptake and rapid normal tissue activity clearance in the preclinical model of B16F1 and B16F10 mouse melanomas. These results highlight the potential of using radiolabeled αMSH analogues in clinical applications for molecular imaging and radionuclide therapy of melanoma.
Collapse
Affiliation(s)
- Chengcheng Zhang
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada.,2 Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - François Bénard
- 1 Department of Molecular Oncology, BC Cancer, Vancouver, British Columbia, Canada.,2 Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
24
|
Gabriel M, Nilica B, Kaiser B, Virgolini IJ. Twelve-Year Follow-up After Peptide Receptor Radionuclide Therapy. J Nucl Med 2018; 60:524-529. [PMID: 30115690 DOI: 10.2967/jnumed.118.215376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/10/2018] [Indexed: 02/01/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) has been used for more than 20 y as a systemic treatment approach in inoperable or metastatic somatostatin receptor-positive tumors. The purpose of this study was to analyze the long-term outcome of PRRT with regard to the most commonly used radiopharmaceuticals, 90Y-DOTATOC and 177Lu-DOTATATE. Methods: This retrospective clinical study included a total of 44 consecutive patients (27 men) with advanced tumors and enhanced somatostatin receptor expression. Mean age at initial diagnosis was 60 y (SD, 11.3 y; range, 40-84 y). Median follow-up was 80 mo. For 177Lu-PRRT, the mean number of cycles administered was 5.3 ± 2.5 and the mean activity was 27.2 ± 14.9 GBq per patient. For 90Y-PRRT, the mean number of cycles administered was 5.5 ± 2.6 and the mean activity was 14.7 ± 7.3 GBq per patient. Overall, 378 cycles were administered (mean, 8.6 ± 3.4 cycles per patient), with an overall cumulative activity of 1,514.1 GBq. Results: Median overall survival was 79 mo. Twenty-one (77.8%) of the 27 men and 9 (52.9%) of the 17 women had died 12 y after commencement of PRRT. The shortest duration of illness was 8 mo and the longest 155 mo. Severe side effects (World Health Organization grades III and IV) were seen in 9 of the 14 patients still alive. Chronic kidney disease in combination with anemia was the most common finding in the 9 patients with severe side effects. A poor prognosis was found for those patients who showed progressive disease, in comparison with patients with cumulative disease control after initial PRRT (log rank, P < 0.001), whereas women and patients with no more than 2 tumor sites seemed to especially benefit from PRRT (not reaching significance levels). Conclusion: PRRT is encouraging in terms of long-term outcome. Thirty-two percent (14/44 patients) of the patients with metastatic or inoperable disease were still alive more than 12 y after the beginning of radionuclide therapy. Possible predictors for favorable outcome are having an initial response to PRRT, having a low number of affected sites, and being female.
Collapse
Affiliation(s)
- Michael Gabriel
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and .,Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Linz, Austria
| | - Bernhard Nilica
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and
| | - Bernhard Kaiser
- Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Linz, Austria
| | - Irene J Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria; and
| |
Collapse
|
25
|
Martini C, Buxbaum S, Rodrigues M, Nilica B, Scarpa L, Holzner B, Virgolini I, Gamper EM. Quality of Life in Patients with Metastatic Gastroenteropancreatic Neuroendocrine Tumors Receiving Peptide Receptor Radionuclide Therapy: Information from a Monitoring Program in Clinical Routine. J Nucl Med 2018; 59:1566-1573. [PMID: 30042164 DOI: 10.2967/jnumed.117.204834] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/31/2018] [Indexed: 12/16/2022] Open
Abstract
In patients with metastatic gastroenteropancreatic neuroendocrine tumors (NETs), we evaluated health-related quality of life (HRQoL) from the first peptide receptor radionuclide therapy (PRRT) to the first restaging and compared the scores with general-population (GP) norms. Methods: The data were from routine HRQoL monitoring using the core quality-of-life questionnaire of the European Organization for Research and Treatment of Cancer (EORTC QLQ-C30). Patients received 4-6 cycles of 177Lu-DOTATATE or 90Y-DOTATOC. To be eligible for analysis, patients had to have at least one HRQoL assessment before PRRT and at least one HRQoL assessment at the end of or after treatment completion. Linear mixed models were used to consider HRQoL changes over time. Results: In total, 61 gastroenteropancreatic NET patients (small-intestine NETs, n = 37; pancreatic NETs, n = 24) were eligible for analysis. Clear improvements from baseline to the first restaging were found for diarrhea in small-intestine NET patients, showing a decrease of 16 points, which represents a moderately large change. We observed a clinically relevant decrease in appetite loss (17 points), but for female small-intestine NET patients only. Other HRQoL changes were also restricted to sociodemographic or clinical subgroups and mainly reflected improvements, except for physical and social functioning, which showed decreasing scores in older small-intestine NET patients. Compared with HRQoL GP norms, patients had impairments consisting of diarrhea; fatigue; appetite loss; reduced physical, social, and role functioning; and reduced global HRQoL. Except for diarrhea and appetite loss, patient scores at the first restaging did not reach GP levels. Conclusion: Our analyses support previous findings of overall stable HRQoL under PRRT. Yet, significant HRQoL impairments compared with the GP and potentially specific subgroup patterns need to be considered.
Collapse
Affiliation(s)
- Caroline Martini
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatry I, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabine Buxbaum
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Margarida Rodrigues
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Nilica
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenza Scarpa
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Holzner
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatry I, Medical University of Innsbruck, Innsbruck, Austria.,Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatry II, Medical University of Innsbruck, Innsbruck, Austria; and
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Eva-Maria Gamper
- Innsbruck Institute of Patient-Centered Outcome Research (IIPCOR), Innsbruck, Austria
| |
Collapse
|
26
|
Gleeson EC, Jackson WR, Robinson AJ. Ring closing metathesis of unprotected peptides. Chem Commun (Camb) 2018; 53:9769-9772. [PMID: 28815236 DOI: 10.1039/c7cc04100d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An efficient and expedient route to the synthesis of dicarba peptides from protecting group-free sequences is reported using Ru-alkylidene catalysed olefin metathesis. A range of cyclic peptides was prepared from linear peptides containing two Z-crotyl glycine residues. Free amine groups were masked as salts with Brønsted acids preventing in situ catalyst decomposition. Excellent RCM conversion was obtained in both DMF and methanol.
Collapse
Affiliation(s)
- Ellen C Gleeson
- School of Chemistry, Monash University, Clayton 3800, Victoria, Australia.
| | | | | |
Collapse
|
27
|
Abstract
Pancreatic neuroendocrine tumours (PNETs) might occur as a non-familial isolated endocrinopathy or as part of a complex hereditary syndrome, such as multiple endocrine neoplasia type 1 (MEN1). MEN1 is an autosomal dominant disorder characterized by the combined occurrence of PNETs with tumours of the parathyroids and anterior pituitary. Treatments for primary PNETs include surgery. Treatments for non-resectable PNETs and metastases include biotherapy (for example, somatostatin analogues, inhibitors of receptors and monoclonal antibodies), chemotherapy and radiological therapy. All these treatments are effective for PNETs in patients without MEN1; however, there is a scarcity of clinical trials reporting the efficacy of the same treatments of PNETs in patients with MEN1. Treatment of PNETs in patients with MEN1 is challenging owing to the concomitant development of other tumours, which might have metastasized. In recent years, preclinical studies have identified potential new therapeutic targets for treating MEN1-associated neuroendocrine tumours (including PNETs), and these include epigenetic modification, the β-catenin-wingless (WNT) pathway, Hedgehog signalling, somatostatin receptors and MEN1 gene replacement therapy. This Review discusses these advances.
Collapse
Affiliation(s)
- Morten Frost
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
- Endocrine Research Unit, University of Southern Denmark, Odense, 5000, Denmark
| | - Kate E Lines
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, OX3 7LJ. United Kingdom
| |
Collapse
|
28
|
Katsila T, Liontos M, Patrinos GP, Bamias A, Kardamakis D. The New Age of -omics in Urothelial Cancer - Re-wording Its Diagnosis and Treatment. EBioMedicine 2018; 28:43-50. [PMID: 29428524 PMCID: PMC5835572 DOI: 10.1016/j.ebiom.2018.01.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 01/31/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023] Open
Abstract
Unmet needs in urothelial cancer management represent an important challenge in our effort to improve long-term overall and disease-free survival rates with no significant compromise in quality of life. Radical cystectomy with pelvic lymph node dissection is the standard for the management of muscle-invasive, non-metastatic cancers. In spite of a 90% local disease control, up to 50% of patients ultimately die of distant metastasis. Bladder preservation using chemo-radiation is an acceptable alternative, but optimal patient selection remains elusive. Recent research is focused on the employment of tailored-made strategies in urothelial cancer exploiting the potential of theranostics in patient selection for specific therapies. Herein, we review the current knowledge on molecular theranostics in urothelial cancer and we suggest that this is the time to move toward imaging theranostics, if tailored-made disease management and patient stratification is envisaged. Urothelial cancer management represents an important challenge. Optimum patient stratification and tailored-made theranostics remain elusive. Imaging theranostics is envisaged as a cancer roadmap.
Collapse
Affiliation(s)
- Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece; Department of Radiation Oncology, University of Patras Medical School, Patras, Greece.
| | - Michalis Liontos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras, Greece; Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Kardamakis
- Department of Radiation Oncology, University of Patras Medical School, Patras, Greece
| |
Collapse
|
29
|
Mathur A, Prashant V, Sakhare N, Chakraborty S, Vimalnath K, Mohan RK, Arjun C, Karkhanis B, Seshan R, Basu S, Korde A, Banerjee S, Dash A, Sachdev SS. Bulk Scale Formulation of Therapeutic Doses of Clinical Grade Ready-to-Use 177Lu-DOTA-TATE: The Intricate Radiochemistry Aspects. Cancer Biother Radiopharm 2017. [DOI: 10.1089/cbr.2017.2208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Anupam Mathur
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Vrinda Prashant
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Navin Sakhare
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Sudipta Chakraborty
- Department of Atomic Energy, Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - K.V. Vimalnath
- Department of Atomic Energy, Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Repaka Krishna Mohan
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Chanda Arjun
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Barkha Karkhanis
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Ravi Seshan
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| | - Sandip Basu
- Department of Atomic Energy, Radiation Medicine Centre, Mumbai, India
| | - Aruna Korde
- Department of Atomic Energy, Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Sharmila Banerjee
- Department of Atomic Energy, Radiation Medicine Centre, Mumbai, India
| | - Ashutosh Dash
- Department of Atomic Energy, Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Satbir Singh Sachdev
- Department of Atomic Energy, Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai, India
| |
Collapse
|
30
|
Maina T, Nock BA, Kulkarni H, Singh A, Baum RP. Theranostic Prospects of Gastrin-Releasing Peptide Receptor–Radioantagonists in Oncology. PET Clin 2017; 12:297-309. [DOI: 10.1016/j.cpet.2017.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Gill MR, Falzone N, Du Y, Vallis KA. Targeted radionuclide therapy in combined-modality regimens. Lancet Oncol 2017; 18:e414-e423. [PMID: 28677577 DOI: 10.1016/s1470-2045(17)30379-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/27/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Targeted radionuclide therapy (TRT) is a branch of cancer medicine concerned with the use of radioisotopes, radiolabelled molecules, nanoparticles, or microparticles that either naturally accumulate in or are designed to target tumours. TRT combines the specificity of molecular and sometimes physical targeting with the potent cytotoxicity of ionising radiation. Targeting vectors for TRT include antibodies, antibody fragments, proteins, peptides, and small molecules. The diversity of available carrier molecules, together with the large panel of suitable radioisotopes with unique physicochemical properties, allows vector-radionuclide pairings to be matched to the molecular, pathological, and physical characteristics of a tumour. Some pairings are designed for dual therapeutic and diagnostic applications. Use of TRT is increasing with the adoption into practice of radium-223 dichloride for the treatment of bone metastases and with the ongoing clinical development of, among others, 177Lu-dodecanetetraacetic acid tyrosine-3-octreotate (DOTATATE) for the treatment of neuroendocrine tumours and 90Y-microspheres for the treatment of hepatic tumours. The increasing use of TRT raises the question of how best to integrate TRT into multimodality protocols. Achievements in this area and the future prospects of TRT are evaluated in this Review.
Collapse
Affiliation(s)
- Martin R Gill
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Nadia Falzone
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Yong Du
- The Royal Marsden Hospital NHS Foundation Trust, Sutton, Surrey, UK
| | - Katherine A Vallis
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
32
|
Liu F, Zhu H, Yu J, Han X, Xie Q, Liu T, Xia C, Li N, Yang Z. 68Ga/177Lu-labeled DOTA-TATE shows similar imaging and biodistribution in neuroendocrine tumor model. Tumour Biol 2017; 39:1010428317705519. [PMID: 28618966 DOI: 10.1177/1010428317705519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Fei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiangyuan Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xuedi Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qinghua Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
- College of Chemistry, Sichuan University, Chengdu, China
| | - Teli Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chuanqin Xia
- College of Chemistry, Sichuan University, Chengdu, China
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
33
|
Clinical Response Profile of Metastatic/Advanced Pulmonary Neuroendocrine Tumors to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE. Clin Nucl Med 2017; 42:428-435. [DOI: 10.1097/rlu.0000000000001639] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
34
|
Abstract
神经内分泌瘤是一类起源于神经内分泌细胞, 发病率低且具有分子和生物学行为异质性的肿瘤. 现报道一例右半结肠神经内分泌瘤G2伴多发淋巴结转移患者, 以期对临床工作者有所帮助.
Collapse
|
35
|
Nicolas GP, Mansi R, McDougall L, Kaufmann J, Bouterfa H, Wild D, Fani M. Biodistribution, Pharmacokinetics, and Dosimetry of 177Lu-, 90Y-, and 111In-Labeled Somatostatin Receptor Antagonist OPS201 in Comparison to the Agonist 177Lu-DOTATATE: The Mass Effect. J Nucl Med 2017; 58:1435-1441. [DOI: 10.2967/jnumed.117.191684] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/11/2017] [Indexed: 01/08/2023] Open
|
36
|
Somatostatin receptor expression related to TP53 and RB1 alterations in pancreatic and extrapancreatic neuroendocrine neoplasms with a Ki67-index above 20. Mod Pathol 2017; 30:587-598. [PMID: 28059098 DOI: 10.1038/modpathol.2016.217] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/14/2022]
Abstract
Somatostatin receptor 2A expression is a feature of well-differentiated neuroendocrine neoplasms and is important for their diagnosis and therapy. Little is known about somatostatin receptor 2A expression in poorly differentiated neuroendocrine neoplasms in relation to TP53 and RB1 status and how these features may contribute to the separation of well from poorly differentiated neuroendocrine neoplasms with a proliferation index above 20%. This study investigates the expression of somatostatin receptors, p53 and Rb1, and TP53 alterations in pancreatic and extrapancreatic well and poorly differentiated neuroendocrine neoplasms (Ki67-index >20%). Thirty-seven poorly differentiated neuroendocrine neoplasms of pancreatic (n=12) and extrapancreatic origin (n=25) as well as 10 well-differentiated neuroendocrine neoplasms of the pancreas (n=9) and rectum (n=1) with a Ki67-index >20% were immunostained for synaptophysin, chromogranin A, Ki67, CD56, p53, Rb1, ATRX, DAXX, progesterone receptor, somatostatin receptor 2A, somatostatin receptor 5, and cytokeratin 20, and sequenced for TP53, exons 5-9. Somatostatin receptor 2A was positive in 6/37 of poorly differentiated and in 8/10 of well-differentiated neuroendocrine neoplasms. One well-differentiated and two poorly differentiated neuroendocrine neoplasms expressed somatostatin receptor 5. Abnormal nuclear p53 and Rb1 staining was found in 29/37 and 22/37 poorly differentiated neuroendocrine neoplasms, respectively, whereas all well-differentiated neuroendocrine neoplasms showed normal p53 and Rb1 expression. TP53 gene alterations were restricted to poorly differentiated neuroendocrine neoplasms (24/34) and correlated well with p53 expression. All cases were progesterone receptor negative. Somatostatin receptor 2A expression is not limited to well-differentiated neuroendocrine neoplasms but also occurs in 16% of poorly differentiated neuroendocrine neoplasms from various sites. Most poorly differentiated neuroendocrine neoplasms are characterized by TP53 alterations and Rb1 loss, usually in the absence of somatostatin receptor 2A expression. In the pancreas, these criteria contribute to separate well-differentiated neuroendocrine neoplasms with a Ki67-index above 20% from poorly differentiated neuroendocrine neoplasms.
Collapse
|
37
|
Fani M, Peitl PK, Velikyan I. Current Status of Radiopharmaceuticals for the Theranostics of Neuroendocrine Neoplasms. Pharmaceuticals (Basel) 2017; 10:E30. [PMID: 28295000 PMCID: PMC5374434 DOI: 10.3390/ph10010030] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
Nuclear medicine plays a pivotal role in the management of patients affected by neuroendocrine neoplasms (NENs). Radiolabeled somatostatin receptor analogs are by far the most advanced radiopharmaceuticals for diagnosis and therapy (radiotheranostics) of NENs. Their clinical success emerged receptor-targeted radiolabeled peptides as an important class of radiopharmaceuticals and it paved the way for the investigation of other radioligand-receptor systems. Besides the somatostatin receptors (sstr), other receptors have also been linked to NENs and quite a number of potential radiolabeled peptides have been derived from them. The Glucagon-Like Peptide-1 Receptor (GLP-1R) is highly expressed in benign insulinomas, the Cholecystokinin 2 (CCK2)/Gastrin receptor is expressed in different NENs, in particular medullary thyroid cancer, and the Glucose-dependent Insulinotropic Polypeptide (GIP) receptor was found to be expressed in gastrointestinal and bronchial NENs, where interestingly, it is present in most of the sstr-negative and GLP-1R-negative NENs. Also in the field of sstr targeting new discoveries brought into light an alternative approach with the use of radiolabeled somatostatin receptor antagonists, instead of the clinically used agonists. The purpose of this review is to present the current status and the most innovative strategies for the diagnosis and treatment (theranostics) of neuroendocrine neoplasms using a cadre of radiolabeled regulatory peptides targeting their receptors.
Collapse
Affiliation(s)
- Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital of Basel, 4031 Basel, Switzerland.
| | - Petra Kolenc Peitl
- Department of Nuclear Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia.
| | - Irina Velikyan
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden.
| |
Collapse
|
38
|
Peptide Receptor Radionuclide Therapy Outcomes in a North American Cohort With Metastatic Well-Differentiated Neuroendocrine Tumors. Pancreas 2017; 46:151-156. [PMID: 27759712 PMCID: PMC5595066 DOI: 10.1097/mpa.0000000000000734] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The objective of this study was to describe the outcomes of patients in the University of Iowa Neuroendocrine Tumor (NET) Database treated with peptide receptor radionuclide therapy (PRRT). METHODS One hundred thirty-five patients from the University of Iowa NET Database who received PRRT were analyzed, their characteristics were described, and survival was calculated. RESULTS The median age at diagnosis was 51 years, and 64% were men. The primary tumor was located in the small bowel (SBNET) in 37.8%, in the pancreas (PNET) in 26.0%, in the lung in 13.3%, in unknown primary in 9.6%, and in other sites in 13.3%. A radiographic response of any magnitude was observed in 65.8%, 11.1% had a mixed response, and 15.4% showed progression. The overall survival (OS) from the first PRRT was 40 months, and the median time to progression was 23.9 months. Higher pretreatment chromogranin A and pancreastatin levels predicted inferior OS. CONCLUSIONS Peptide receptor radionuclide therapy resulted in a relatively long OS and time to progression in heavily pretreated North American patients with advanced NETs. Elevated pretreatment chromogranin A and pancreastatin predicted shorter OS after therapy. Peptide receptor radionuclide therapy is a valuable treatment option in patients with advanced NETs, especially SBNETS.
Collapse
|
39
|
Zhao J, Zhou M, Li C. Synthetic nanoparticles for delivery of radioisotopes and radiosensitizers in cancer therapy. Cancer Nanotechnol 2016; 7:9. [PMID: 27909463 PMCID: PMC5112292 DOI: 10.1186/s12645-016-0022-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy has been, and will continue to be, a critical modality to treat cancer. Since the discovery of radiation-induced cytotoxicity in the late 19th century, both external and internal radiation sources have provided tremendous benefits to extend the life of cancer patients. Despite the dramatic improvement of radiation techniques, however, one challenge persists to limit the anti-tumor efficacy of radiotherapy, which is to maximize the deposited dose in tumor while sparing the rest of the healthy vital organs. Nanomedicine has stepped into the spotlight of cancer diagnosis and therapy during the past decades. Nanoparticles can potentiate radiotherapy by specifically delivering radionuclides or radiosensitizers into tumors, therefore enhancing the efficacy while alleviating the toxicity of radiotherapy. This paper reviews recent advances in synthetic nanoparticles for radiotherapy and radiosensitization, with a focus on the enhancement of in vivo anti-tumor activities. We also provide a brief discussion on radiation-associated toxicities as this is an area that, up to date, has been largely missing in the literature and should be closely examined in future studies involving nanoparticle-mediated radiosensitization.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Road, Houston, TX 77054 USA
| | - Min Zhou
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Road, Houston, TX 77054 USA
| |
Collapse
|
40
|
Sevilla I, Segura Á, Capdevila J, López C, García-Carbonero R, Grande E. Management of controversial gastroenteropancreatic neuroendocrine tumour clinical situations with somatostatin analogues: results of a Delphi questionnaire panel from the NETPraxis program. BMC Cancer 2016; 16:858. [PMID: 27821081 PMCID: PMC5100262 DOI: 10.1186/s12885-016-2901-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There are clinical situations (CS) in which the use of somatostatin analogs (SSAs) in patients with neuroendocrine tumors (NET) is controversial due to lack of evidence. A Delphi study was conducted to develop common treatment guidelines for these CS, based on clinical practice and expert opinion of Spanish oncologists. METHODS A scientific committee identified 5 CS with a common core (c-c) [non-functioning NET, not susceptible of surgery/locoregional therapy, Ki67 < 10 % (except for CS5: >10 %), ECOG ≤ 2], and controversy regarding use of SSAs, and prepared a Delphi questionnaire of 48 treatment statements. Statements were rated on a 1 (completely disagree) to 9 (completely agree) scale. Responses were grouped by tertiles: 1-3: Disagreement, 4-6: Neutral, 7-9: Agreement. Consensus was reached when the responses of ≥2/3 participants were located in the same tertile as the median value of all reported responses for that statement. RESULTS Sixty five (81.2 %) of 80 invited oncologists with experience in the management of NETs answered a first round of the questionnaire and 57 (87.7 %) of those 65 answered a second round (mean age 43.5 years; 53.8 % women; median time of experience 9 years). Consensus was obtained in 42 (36 agreement and 6 disagreement) of the 48 statements (87.5 %). Regarding CS1 (Enteropancreatic NET, c-c, non-progressive in the last 3-6 months), overall, SSA treatment is recommended (a wait and see approach is anecdotal and reserved for fragile patients or with low tumor load or ki-67 < 2 %); CS2 (Pancreatic NET, c-c), overall, SSA monotherapy is recommended, except when high tumor load or tumor progression exists, where combination therapy would be considered; CS3 [Gastroenteropancreatic (GEP)-NET, c-c, in treatment with anti-proliferative dose of SSA and progressing], overall, SSA maintenance is recommended at the time of progression, with or without adding molecular targeted drugs; CS4 (GEP-NET, c-c, and negative octreoscan®), SSA in monotherapy is only considered in low-risk patients (low tumor load and Ki-67 < 5 %); CS5 [GEP-NET, c-c (ki67 > 10 %), and positive octreoscan®], monotherapy with SSA is mainly considered in patients with comorbidities. CONCLUSION Several recommendations regarding use of SSAs in controversial NET CS were reached in consensus and might be considered as treatment guideline.
Collapse
Affiliation(s)
- Isabel Sevilla
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
| | - Ángel Segura
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
| | - Jaume Capdevila
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Carlos López
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
| | - Rocío García-Carbonero
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
| | - Enrique Grande
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| | - On behalf of GETNE (Spanish Group of NeuroEndocrine Tumors)
- Oncology Unit. Hospital Clínico y Regional de Málaga, Colonia Santa Inés s/n, Málaga, 29010 Spain
- Oncology Unit. Hospital Universitario La Fe, Avda. de Fernando Abril Martorell 106, 46026 Valencia, Spain
- Oncology Unit. Hospital Vall d’Hebron, Pg de la Vall d’Hebron 119-129, 08035 Barcelona, Spain
- Oncology Unit. Hospital Marqués de Valdecilla, Avda. Valdecilla 25, 39008 Santander, Spain
- Oncology Unit. Hospital Universitario 12 de Octubre, Avda. de Córdoba s/n, 28041 Madrid, Spain
- Oncology Unit. Hospital Universitario Ramón y Cajal, Ctra. de Colmenar Viejo km. 9.100, 28034 Madrid, Spain
| |
Collapse
|
41
|
Chang CA, Pattison DA, Tothill RW, Kong G, Akhurst TJ, Hicks RJ, Hofman MS. (68)Ga-DOTATATE and (18)F-FDG PET/CT in Paraganglioma and Pheochromocytoma: utility, patterns and heterogeneity. Cancer Imaging 2016; 16:22. [PMID: 27535829 PMCID: PMC4989291 DOI: 10.1186/s40644-016-0084-2] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022] Open
Abstract
Background Pheochromocytomas (PCC) and paragangliomas (PGL) are neuroendocrine tumours arising from pluripotent neural crest stem cells and are associated with neurons of the autonomic nervous system. PCCs/PGLs are often hereditary and multifocal, and their biologic behaviour and metabolic activity vary making imaging of these tumours challenging. The imaging gold standard has been I-123 MIBG complemented by CT or MRI. PGLs being neuroendocrine tumours express somatostatin receptors enabling imaging with Ga-68 DOTA-coupled peptides such as DOTATATE. Imaging with F-18 FDG also provides additional information regarding metabolic activity and biologic aggressiveness of these tumours, or, in some situations, reflecting metabolic reprogramming of these tumours. We report our experience using both Ga-68 DOTATATE and F-18 FDG PET/CT imaging in patients with PGLs and PCCs. Methods This was a retrospective review of 23 patients with proven PGL/PCC who underwent both DOTATATE and FDG PET/CT. Seven patients also had I-123 MIBG SPECT/CT and 1 patient had I-124 MIBG PET/CT. Lesional intensity and patterns of uptake were analysed. Results DOTATATE and FDG were positive at most sites of disease (96.2 % vs 91.4 %), although uptake intensity was significantly higher on DOTATATE with a median SUV of 21 compared to 12.5 for FDG (p < 0.001). SUVmax on F-18 FDG was significantly higher (p < 0.001) in clinically aggressive cases. I-123/I-124 MIBG detected fewer lesions (30.4 %). Conclusion Overall, Ga-68 DOTATATE PET/CT detected similar number but has significantly greater lesion-to-background contrast compared to F-18 FDG PET/CT. Combined with high specificity, patient convenience and relatively low cost, DOTATATE PET/CT should be considered the ideal first line investigation for imaging PGL/PCC. Depending on DOTATATE findings and the clinical question, FDG and MIBG remain useful and, in selected cases, may provide more accurate staging, disease characterisation and guide treatment choices.
Collapse
Affiliation(s)
- Chian A Chang
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - David A Pattison
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Endocrinology Service, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Richard W Tothill
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Grace Kong
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia
| | - Tim J Akhurst
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Rodney J Hicks
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael S Hofman
- Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
42
|
Beykan S, Dam JS, Eberlein U, Kaufmann J, Kjærgaard B, Jødal L, Bouterfa H, Bejot R, Lassmann M, Jensen SB. 177Lu-OPS201 targeting somatostatin receptors: in vivo biodistribution and dosimetry in a pig model. EJNMMI Res 2016; 6:50. [PMID: 27294582 PMCID: PMC4906090 DOI: 10.1186/s13550-016-0204-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/27/2016] [Indexed: 11/10/2022] Open
Abstract
Background 177Lu is used in peptide receptor radionuclide therapies for the treatment of neuroendocrine tumors. Based on the recent literature, SST2 antagonists are superior to agonists in tumor uptake. The compound OPS201 is the novel somatostatin antagonist showing the highest SST2 affinity. The aim of this study was to measure the in vivo biodistribution and dosimetry of 177Lu-OPS201 in five anesthetized Danish Landrace pigs as an appropriate substitute for humans to quantitatively assess the absorbed doses for future clinical applications. Results 177Lu-OPS201 was obtained with a specific activity ranging from 10 to 17 MBq/μg. Prior to administration, the radiochemical purity was measured as s > 99.7 % in all cases. After injection, fast clearance of the compound from the blood stream was observed. Less than 5 % of the injected activity was presented in blood 10 min after injection. A series of SPECT/CT and whole-body scans conducted until 10 days after intravenous injection showed uptake mostly in the liver, spine, and kidneys. There was no visible uptake in the spleen. Blood samples were taken to determine the time-activity curve in the blood. Time-activity curves and time-integrated activity coefficients were calculated for the organs showing visible uptake. Based on these data, the absorbed organ dose coefficients for a 70-kg patient were calculated with OLINDA/EXM. For humans after an injection of 5 GBq 177Lu-OPS201, the highest predicted absorbed doses are obtained for the kidneys (13.7 Gy), the osteogenic cells (3.9 Gy), the urinary bladder wall (1.8 Gy), and the liver (1.0 Gy). No metabolites of 177Lu-OPS201 were found by radio HPLC analysis. None of the absorbed doses calculated will exceed organ toxicity levels. Conclusions The 177Lu-OPS201 was well tolerated and caused no abnormal physiological or behavioral signs. In vivo distributions and absorbed doses of pigs are comparable to those observed in other publications. According to the biodistribution data in pigs, presented in this work, the expected radiation exposure in humans will be within the acceptable range. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0204-9) contains supplementary material, which is available to authorized users.
Collapse
|
43
|
Salavati A, Puranik A, Kulkarni HR, Budiawan H, Baum RP. Peptide Receptor Radionuclide Therapy (PRRT) of Medullary and Nonmedullary Thyroid Cancer Using Radiolabeled Somatostatin Analogues. Semin Nucl Med 2016; 46:215-24. [DOI: 10.1053/j.semnuclmed.2016.01.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
44
|
Ramos-Álvarez I, Moreno P, Mantey SA, Nakamura T, Nuche-Berenguer B, Moody TW, Coy DH, Jensen RT. Insights into bombesin receptors and ligands: Highlighting recent advances. Peptides 2015; 72:128-44. [PMID: 25976083 PMCID: PMC4641779 DOI: 10.1016/j.peptides.2015.04.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/29/2015] [Accepted: 04/30/2015] [Indexed: 12/22/2022]
Abstract
This following article is written for Prof. Abba Kastin's Festschrift, to add to the tribute to his important role in the advancement of the role of peptides in physiological, as well as pathophysiological processes. There have been many advances during the 35 years of his prominent role in the Peptide field, not only as editor of the journal Peptides, but also as a scientific investigator and editor of two volumes of the Handbook of Biological Active Peptides [146,147]. Similar to the advances with many different peptides, during this 35 year period, there have been much progress made in the understanding of the pharmacology, cell biology and the role of (bombesin) Bn receptors and their ligands in various disease states, since the original isolation of bombesin from skin of the European frog Bombina bombina in 1970 [76]. This paper will briefly review some of these advances over the time period of Prof. Kastin 35 years in the peptide field concentrating on the advances since 2007 when many of the results from earlier studies were summarized [128,129]. It is appropriate to do this because there have been 280 articles published in Peptides during this time on bombesin-related peptides and it accounts for almost 5% of all publications. Furthermore, 22 Bn publications we have been involved in have been published in either Peptides [14,39,55,58,81,92,93,119,152,216,225,226,231,280,302,309,355,361,362] or in Prof. Kastin's Handbook of Biological Active Peptides [137,138,331].
Collapse
Affiliation(s)
- Irene Ramos-Álvarez
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Paola Moreno
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Samuel A Mantey
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Taichi Nakamura
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Bernardo Nuche-Berenguer
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - Terry W Moody
- Center for Cancer Research, Office of the Director, NCI, National Institutes of Health, Bethesda, MD 20892-1804, United States
| | - David H Coy
- Peptide Research Laboratory, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70112-2699, United States
| | - Robert T Jensen
- Digestive Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892-1804, United States.
| |
Collapse
|
45
|
Molecular response assessed by (68)Ga-DOTANOC and survival after (90)Y microsphere therapy in patients with liver metastases from neuroendocrine tumours. Eur J Nucl Med Mol Imaging 2015; 43:432-40. [PMID: 26323577 DOI: 10.1007/s00259-015-3178-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/14/2015] [Indexed: 01/04/2023]
Abstract
PURPOSE We investigated the prognostic role of (68)Ga-DOTANOC in patients affected by hepatic metastases from neuroendocrine tumours (NET) undergoing (90)Y radioembolization ((90)Y-RE). METHODS A group of 15 consecutive patients with unresectable NET liver metastases underwent (68)Ga-DOTANOC PET at baseline and 6 weeks after (90)Y-RE. Molecular response was defined as a reduction of >50% in the tumour-to-spleen ratio (ΔT/S). The patients were divided into two groups (responders with ΔT/S >50% and nonresponders with ΔT/S <50%) Patients were followed up by imaging and laboratory tests every 3 months until death or for at least 36 months following (90)Y-RE. Statistical analysis was performed to identify factors predicting overall survival (OS) and progression-free survival (PFS). RESULTS A decrease in T/S ratio was seen in all patients on (68)Ga-DOTANOC PET scans performed after (90)Y-RE. Nine patients were classified as responders and six as nonresponders. The mean OS in all patients was 31.0 months. Responders had a significantly (p < 0.001) longer OS (mean 36.0 ± 2.5 months) and PFS (mean 29.7 ± 3.4 months) than nonresponders. In a multivariate analysis, none of the other examined variables including age, unilobar vs. bilobar locations, bilirubin levels, radiological response or the presence of extrahepatic disease significantly predicted patient outcome. CONCLUSION Molecular response assessed with (68)Ga-DOTANOC PET might be a useful predictor of survival in patients affected by NET liver metastases treated with (90)Y-RE.
Collapse
|
46
|
Royle G, Myhra S, Chakalova R, Vallis KA, Falzone N. Spatial distribution of Auger electrons emitted from internalised radionuclides in cancer cells: the photoresist autoradiography (PAR) method. RADIATION PROTECTION DOSIMETRY 2015; 166:228-32. [PMID: 25889606 DOI: 10.1093/rpd/ncv162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Microdosimetric evaluation of Auger electron-emitting radionuclides involves a detailed evaluation of energy deposition at a nanometre scale. To perform Monte Carlo modelling of such energy deposition, accurate information regarding the spatial distribution of the radionuclide is required. A recent addition to the methods for determining the spatial distribution of cellular internalised radionuclides is based on detection in a polymer photoresist (e.g. polymethyl methacralate), followed by atomic force microscopy analysis of the resultant 3D pattern. In comparison with present practice, the method offers greater spatial resolution and improved quantification. The volume of the pattern is proportional to the total dose, thereby permitting assessment of variability of accumulated activity, while the variation in depth across the pattern reflects the lateral spatial distribution in the local fluence per unit area. An added advantage is the similarity in response to ionising radiation of an organic polymer compared to that of biological material. A pattern in the resist from radiation emitted by a radionuclide treated cell gives additional spatial information about the energy deposited in the resist.
Collapse
Affiliation(s)
- G Royle
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - S Myhra
- Department of Material Science, University of Oxford, Oxford, UK
| | - R Chakalova
- Department of Material Science, University of Oxford, Oxford, UK
| | - K A Vallis
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - N Falzone
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK Department of Biomedical Science, Tshwane University of Technology, Pretoria, South Africa
| |
Collapse
|
47
|
Pre-therapeutic dosimetry of normal organs and tissues of (177)Lu-PSMA-617 prostate-specific membrane antigen (PSMA) inhibitor in patients with castration-resistant prostate cancer. Eur J Nucl Med Mol Imaging 2015; 42:1976-83. [PMID: 26227531 DOI: 10.1007/s00259-015-3125-3] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 06/29/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE (177)Lu-617-prostate-specific membrane antigen (PSMA) ligand seems to be a promising tracer for radionuclide therapy of progressive prostate cancer. However, there are no published data regarding the radiation dose given to the normal tissues. The aim of the present study was to estimate the pretreatment radiation doses in patients who will undergo radiometabolic therapy using a tracer amount of (177)Lu-labeled PSMA ligand. METHODS The study included seven patients with progressive prostate cancer with a mean age of 63.9 ± 3.9 years. All patients had prior PSMA positron emission tomography (PET) imaging and had intense tracer uptake at the lesions. The injected (177)Lu-PSMA-617 activity ranged from 185 to 210 MBq with a mean of 192.6 ± 11.0 MBq. To evaluate bone marrow absorbed dose 2-cc blood samples were withdrawn in short variable times (3, 15, 30, 60, and 180 min and 24, 48, and 120 h) after injection. Whole-body images were obtained at 4, 24, 48, and 120 h post-injection (p.i.). The geometric mean of anterior and posterior counts was determined through region of interest (ROI) analysis. Attenuation correction was applied using PSMA PET/CT images. The OLINDA/EXM dosimetry program was used for curve fitting, residence time calculation, and absorbed dose calculations. RESULTS The calculated radiation-absorbed doses for each organ showed substantial variation. The highest radiation estimated doses were calculated for parotid glands and kidneys. Calculated radiation-absorbed doses per megabecquerel were 1.17 ± 0.31 mGy for parotid glands and 0.88 ± 0.40 mGy for kidneys. The radiation dose given to the bone marrow was significantly lower than those of kidney and parotid glands (p < 0.05). The calculated radiation dose to bone marrow was 0.03 ± 0.01 mGy/MBq. CONCLUSION Our first results suggested that (177)Lu-PSMA-617 therapy seems to be a safe method. The dose-limiting organ seems to be the parotid glands rather than kidneys and bone marrow. The lesion radiation doses are within acceptable ranges; however, there is a substantial individual variance so patient dosimetry seems to be mandatory.
Collapse
|
48
|
Van Binnebeek S, Vanbilloen B, Baete K, Terwinghe C, Koole M, Mottaghy FM, Clement PM, Mortelmans L, Bogaerts K, Haustermans K, Nackaerts K, Van Cutsem E, Verslype C, Verbruggen A, Deroose CM. Comparison of diagnostic accuracy of (111)In-pentetreotide SPECT and (68)Ga-DOTATOC PET/CT: A lesion-by-lesion analysis in patients with metastatic neuroendocrine tumours. Eur Radiol 2015; 26:900-9. [PMID: 26162577 DOI: 10.1007/s00330-015-3882-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 05/04/2015] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To compare the diagnostic accuracy of (111)In-pentetreotide-scintigraphy with (68)Ga-DOTATOC-positron emission tomography (PET)/computed tomography (CT) in patients with metastatic-neuroendocrine tumour (NET) scheduled for peptide receptor radionuclide therapy (PRRT). Incremental lesions (ILs) were defined as lesions observed on only one modality. METHODS Fifty-three metastatic-NET-patients underwent (111)In-pentetreotide-scintigraphy (24 h post-injection; planar+single-photon emission CT (SPECT) abdomen) and whole-body (68)Ga-DOTATOC-PET/CT. SPECT and PET were compared in a lesion-by-lesion and organ-by-organ analysis, determining the total lesions and ILs for both modalities. RESULTS Significantly more lesions were detected on (68)Ga-DOTATOC-PET/CT versus (111)In-pentetreotide-scintigraphy. More specifically, we observed 1,098 lesions on PET/CT (range: 1-105; median: 15) versus 660 on SPECT (range: 0-73, median: 9) (p<0.0001), with 439 PET-ILs (42/53 patients) and one SPECT-IL (1/53 patients). The sensitivity for PET/CT was 99.9 % (95 % CI, 99.3-100.0), for SPECT 60.0 % (95 % CI, 48.5-70.2). The organ-by-organ analysis showed that the PET-ILs were most frequently visualized in liver and skeleton. CONCLUSION Ga-DOTATOC-PET/CT is superior for the detection of NET-metastases compared to (111)In-pentetreotide SPECT. KEY POINTS Somatostatin receptor PET is superior to SPECT in detecting NET metastases. PET is the scintigraphic method for accurate depiction of NET tumour burden. The sensitivity of PET is twofold higher than the sensitivity of SPECT.
Collapse
Affiliation(s)
- S Van Binnebeek
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - B Vanbilloen
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - K Baete
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - C Terwinghe
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - M Koole
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - F M Mottaghy
- Department of Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Nuclear Medicine, University Hospital Aachen, Aachen, Germany
| | - P M Clement
- Medical Oncology, University Hospitals Leuven and Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - L Mortelmans
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - K Bogaerts
- Department of Public Health and Primary Care (I-BioStat), KU Leuven and UHasselt, Leuven, Belgium
| | - K Haustermans
- Radiation Oncology, University Hospitals Leuven and Department of Oncology, KU Leuven, Leuven, Belgium
| | - K Nackaerts
- Pulmonology, University Hospitals Leuven, Leuven, Belgium
| | - E Van Cutsem
- Division of Digestive Oncology, University Hospitals Leuven and Department of Oncology, KU Leuven, Leuven, Belgium
| | - C Verslype
- Division of Digestive Oncology, University Hospitals Leuven and Department of Oncology, KU Leuven, Leuven, Belgium
| | - A Verbruggen
- Laboratory for Radiopharmacy, KU Leuven, Leuven, Belgium
| | - C M Deroose
- Nuclear Medicine, University Hospitals Leuven and Department of Imaging and Pathology, KU Leuven, Leuven, Belgium. .,Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
49
|
Somatostatin receptor based imaging and radionuclide therapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:917968. [PMID: 25879040 PMCID: PMC4387942 DOI: 10.1155/2015/917968] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 01/09/2023]
Abstract
Somatostatin (SST) receptors (SSTRs) belong to the typical 7-transmembrane domain family of G-protein-coupled receptors. Five distinct subtypes (termed SSTR1-5) have been identified, with SSTR2 showing the highest affinity for natural SST and synthetic SST analogs. Most neuroendocrine tumors (NETs) have high expression levels of SSTRs, which opens the possibility for tumor imaging and therapy with radiolabeled SST analogs. A number of tracers have been developed for the diagnosis, staging, and treatment of NETs with impressive results, which facilitates the applications of human SSTR subtype 2 (hSSTr2) reporter gene based imaging and therapy in SSTR negative or weakly positive tumors to provide a novel approach for the management of tumors. The hSSTr2 gene can act as not only a reporter gene for in vivo imaging, but also a therapeutic gene for local radionuclide therapy. Even a second therapeutic gene can be transfected into the same tumor cells together with hSSTr2 reporter gene to obtain a synergistic therapeutic effect. However, additional preclinical and especially translational and clinical researches are needed to confirm the value of hSSTr2 reporter gene based imaging and therapy in tumors.
Collapse
|
50
|
Medical imaging in personalised medicine: a white paper of the research committee of the European Society of Radiology (ESR). Insights Imaging 2015; 6:141-55. [PMID: 25763994 PMCID: PMC4376812 DOI: 10.1007/s13244-015-0394-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/02/2015] [Indexed: 02/06/2023] Open
Abstract
The future of medicine lies in early diagnosis and individually tailored treatments, a concept that has been designated 'personalised medicine' (PM), which aims to deliver the right treatment to the right patient at the right time. Medical imaging has always been personalised and is fundamental to almost all aspects of PM. It is instrumental in solving clinical differential diagnoses. Imaging procedures are tailored to the clinical problem and patient characteristics. Screening for preclinical disease is done with imaging. Stratification based on imaging biomarkers can help identify individuals suited for preventive intervention. Treatment decisions are based on the in vivo visualisation of the location and extent of an abnormality, as well as the loco-regional physiological, biochemical and biological processes using structural and molecular imaging. Image-guided biopsy provides relevant tissue specimens for genetic/molecular characterisation. In addition, radiogenomics relate imaging biomarkers to these genetic and molecular features. Furthermore, imaging is essential to patient-tailored therapy planning, therapy monitoring and follow-up of disease, as well as targeting non-invasive or minimally invasive treatments, especially with the rise of theranostics. Radiologists need to be prepared for this new paradigm as it will mean changes in training, clinical practice and in research. Key Points • Medical imaging is a key component in personalised medicine • Personalised prevention will rely on image-based screening programmes • Anatomical, functional and molecular imaging biomarkers affect decisions on the type and intensity of treatment • Treatment response assessment with imaging will improve personalised treatment • Image-based invasive intervention integrates personalised diagnosis and personalised treatment.
Collapse
|