1
|
Padwal MK, Nazar AK, Parghane RV, Basu S, Basu B. Evaluating the prognostic significance of the pre-treatment neutrophil-to-lymphocyte and monocyte-to-lymphocyte ratios in 177Lu-DOTATATE PRRT treated patients with advanced metastatic neuroendocrine tumors. Endocrine 2025:10.1007/s12020-025-04212-z. [PMID: 40131599 DOI: 10.1007/s12020-025-04212-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE This study aimed to investigate the role of pre-treatment neutrophil-to-lymphocyte ratio (NLR) and monocyte-to-lymphocyte ratio (MLR) in the prognosis assessment of 177Lu-DOTATATE Peptide Receptor Radionuclide Therapy (PRRT) treated patients with advanced metastatic neuroendocrine tumors (NETs). METHODS Eligible PRRT-treated patients (n = 247, 2010-2019) were included. Pre-PRRT NLR and MLR were calculated from complete blood count data. Data on tumor characteristics, response to PRRT, progression-free survival (PFS), and overall survival (OS) were evaluated using COXPH analyses. RESULTS In NET patients, median values of NLR and MLR were 2.21 (IQR = 1.66-3.00) and 0.14 (IQR = 0.06-0.24), respectively. NLR showed significant positive association with G3 tumors (median = 3.64, IQR = 2.1-4.26, p = 0.022) and high 18F-FDG avidity (SUVmax>5) (median = 2.5, IQR = 1.82-3.56, p = 0.003). MLR was significantly associated with high disease burden (median = 0.18, IQR = 0.08-0.29, p = 0.0083). MLR distinguished between the PRRT non-responders with progressive disease and responders with complete/partial response (median 0.19 versus 0.12, p = 0.043) or responders with stable disease (median 0.19 versus 0.14, p = 0.045). The ratios independently correlated with disease progression and OS. Patients in NLRhigh (>3.5) group displayed significantly shorter median PFS and OS (48 and 58 months) compared to NLRlow (≤3.5) group (108 and 96 months) (p < 0.01). Patients in MLRhigh (>0.25) group displayed significantly shorter median PFS and OS (40 and 52 months) compared to MLRlow (≤0.25) group (108 and 102 months) (p < 0.01). CONCLUSION Pre-treatment NLR and MLR had an independent prognostic impact on disease progression and OS in PRRT-treated NET patients. This routine, inexpensive CBC-based test in the standard pre-PRRT workup demonstrates the prognostic value and may aid clinicians in the risk stratification of NET patients.
Collapse
Affiliation(s)
- Mahesh K Padwal
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Amir K Nazar
- Homi Bhabha National Institute, Mumbai, India
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India
| | - Rahul V Parghane
- Homi Bhabha National Institute, Mumbai, India
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India
| | - Sandip Basu
- Homi Bhabha National Institute, Mumbai, India.
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India.
| | - Bhakti Basu
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India.
- Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
2
|
Kuiper J, Zoetelief E, Brabander T, de Herder WW, Hofland J. Current status of peptide receptor radionuclide therapy in grade 1 and 2 gastroenteropancreatic neuroendocrine tumours. J Neuroendocrinol 2025; 37:e13469. [PMID: 39563515 PMCID: PMC11919478 DOI: 10.1111/jne.13469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024]
Abstract
Peptide receptor radionuclide therapy (PRRT) using [177Lu-DOTA0,Tyr3]octreotate (177Lu-DOTATATE) represents an established treatment modality for somatostatin receptor-positive, locally advanced or metastatic gastroenteropancreatic neuroendocrine tumours (GEP NET) of grade 1 or 2. The studies have demonstrated that four cycles of PRRT with 177Lu-DOTATATE prolongs progression-free survival and preserves quality of life, in patients with grade 1 and 2 advanced GEP NET. Notably, first-line PRRT using 177Lu-DOTATATE in grade 2 and 3 GEP NET patients has also shown efficacy and safety. Furthermore, PRRT can ameliorate symptoms in patients with NET-associated functioning syndromes. Although various studies have explored alternative radionuclides for PRRT, none currently meet the criteria for routine clinical implementation. Ongoing research aims to further enhance PRRT, and the results from large clinical trials comparing PRRT with other NET treatments are anticipated, potentially leading to significant modifications in NET treatment strategies and PRRT protocols. The results of these studies are likely to help address existing knowledge gaps in the coming years. This review describes the clinical practice, recent developments and future treatment options of PRRT in patients with grade 1 and 2 GEP NET.
Collapse
Affiliation(s)
- Jelka Kuiper
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Eline Zoetelief
- Department of Radiology & Nuclear MedicineErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Tessa Brabander
- Department of Radiology & Nuclear MedicineErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Wouter W. de Herder
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Johannes Hofland
- Department of Internal Medicine, Section of EndocrinologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| |
Collapse
|
3
|
Prasad V, Koumarianou A, Denecke T, Sundin A, Deroose CM, Pavel M, Christ E, Lamarca A, Caplin M, Castaño JP, Dromain C, Falconi M, Grozinsky-Glasberg S, Hofland J, Knigge UP, Kos-Kudla B, Krishna BA, Reed NS, Scarpa A, Srirajaskanthan R, Toumpanakis C, Kjaer A, Hicks RJ, Ambrosini V. Challenges in developing response evaluation criteria for peptide receptor radionuclide therapy: A consensus report from the European Neuroendocrine Tumor Society Advisory Board Meeting 2022 and the ENETS Theranostics Task Force. J Neuroendocrinol 2025; 37:e13479. [PMID: 39653582 DOI: 10.1111/jne.13479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025]
Abstract
Assessing the response to systemic therapy in neuroendocrine tumors (NET) is challenging since morphological imaging response is often delayed and not necessarily reflective of clinical benefit. Peptide receptor radionuclide therapy (PRRT) has a complex mechanism of action, further complicating response assessment. In response to these challenges, the European Neuroendocrine Tumor Society (ENETS) Theranostics Task Force conducted a statement-based survey among experts to identify the current landscape and unmet needs in PRRT response assessment. The survey, presented at the 2022 ENETS Advisory Board (AB) meeting in Vienna, was completed by 70% of AB members, most of whom (81%) were from ENETS Centers of Excellence (CoE). It comprised a set of 13 questions with two substatements in three questions. Six (46%) of the statements achieved more than 75% agreement, while five (39%) additional statements reached over 60% consensus. Key points from the survey include: AB members agreed that lesions deemed equivocal on computed tomography (CT) or magnetic resonance imaging (MRI) should be characterized by somatostatin receptor (SST) positron emission tomography (PET)/CT before being designated as target lesions. It was agreed that interim response assessments should occur after the second or third PRRT cycle. Over half (54%) preferred using both conventional cross-sectional imaging (CT and/or MRI) and hybrid imaging (SST PET/CT) for this purpose. Almost all AB members supported further response assessment 3 months after the final PRRT cycle. A majority (62%) preferred using a combination of conventional cross-sectional imaging and SST PET/CT. For cases showing equivocal progression (ambiguous lesions or nontarget lesions) on CT and/or MRI, further confirmation using SST PET/CT was recommended. A significant majority (74%) preferred assessing pseudo-progression and delayed response by combining SST PET with diagnostic CT and/ or MRI. Though just below the 75% consensus threshold, there was substantial agreement on selecting target lesions based on SST PET/CT uptake intensity and homogeneity. Sixty-nine percent noted the importance of documenting and closely following heterogeneity in lesions in liver, lymph nodes, primary tumors, or other organs. As to the statement on parameters for new response criteria, AB members recommended exploring maximum standard unit value, tumor-to-background ratio, Hounsfield Unit (Choi Criteria), total tumor burden, and novel serum or molecular markers for future response evaluation criteria. Sixty-five percent supported the use of a single SST PET/CT for response assessment of NET lesions treated with PRRT. These findings highlight the importance of integrating advanced imaging techniques and recognizing the need for more nuanced criteria in assessing the efficacy of PRRT in NET patients. This approach aims to enhance the accuracy of treatment monitoring and improve patient outcomes.
Collapse
Affiliation(s)
- Vikas Prasad
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, USA
| | - Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Timm Denecke
- Department of Diagnostic and Interventional Radiology, University Medical Centre Leipzig, Leipzig, Germany
| | - Anders Sundin
- Radiology and Molecular Imaging, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals Leuven and Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Marianne Pavel
- Department of Medicine 1, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Emanuel Christ
- Center of Endocrine and Neuroendocrine Tumors, ENETS Center of Excellence (CoE), Division of Endocrinology, Diabetology and Metabolism, University Hospital of Basel, Basel, Switzerland
| | - Angela Lamarca
- Department of Oncology-OncoHealth Institute-Instituto de Investigaciones Sanitarias FJD, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Martyn Caplin
- Neuroendocrine Tumour Unit, ENETS Center of Excellence, Royal Free Hospital, London, UK
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain
- Reina Sofia University Hospital, Córdoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Córdoba, Spain
| | - Clarisse Dromain
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Massimo Falconi
- Pancreatic Surgery, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Simona Grozinsky-Glasberg
- Neuroendocrine Tumor Unit, ENETS Center of Excellence, Division of Medicine, Hadassah Medical Organization and Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Johannes Hofland
- Department of Internal Medicine, Section of Endocrinology, ENETS Center of Excellence, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ulrich Peter Knigge
- Department of Surgery and Transplantation, Department of Nephrology and Endocrinology, Center of Cancer and Transplantation, ENETS Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Beata Kos-Kudla
- Department of Endocrinology and Neuroendocrine Tumours, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
| | - Balkundi A Krishna
- Department of Nuclear Medicine & PET imaging, Lilavati Hospital & Research Centre, Mumbai, India
| | | | - Aldo Scarpa
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Christos Toumpanakis
- Neuroendocrine Tumour Unit, ENETS Center of Excellence, Royal Free Hospital, London, UK
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital-Rigshospitalet
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rodney J Hicks
- St Vincent's Hospital, Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, Translational Medicine, the Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
4
|
Loree JM, Chan D, Lim J, Stuart H, Fidelman N, Koea J, Posavad J, Cummins M, Doucette S, Myrehaug S, Naraev B, Bailey DL, Bellizzi A, Laidley D, Boyle V, Goodwin R, Del Rivero J, Michael M, Pasieka J, Singh S. Biomarkers to Inform Prognosis and Treatment for Unresectable or Metastatic GEP-NENs. JAMA Oncol 2024; 10:1707-1720. [PMID: 39361298 DOI: 10.1001/jamaoncol.2024.4330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Importance Evidence-based treatment decisions for advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) require individualized patient-centered decision-making that accounts for patient and cancer characteristics. Objective To create an accessible guidance document to educate clinicians and patients on biomarkers informing prognosis and treatment in unresectable or metastatic GEP-NENs. Methods A multidisciplinary panel in-person workshop was convened to define methods. English language articles published from January 2016 to January 2023 in PubMed (MEDLINE) and relevant conference abstracts were reviewed to investigate prognostic and treatment-informing features in unresectable or metastatic GEP-NENs. Data from included studies were used to form evidence-based recommendations. Quality of evidence and strength of recommendations were determined using the Grading of Recommendations, Assessment, Development and Evaluations framework. Consensus was reached via electronic survey following a modified Delphi method. Findings A total of 131 publications were identified, including 8 systematic reviews and meta-analyses, 6 randomized clinical trials, 29 prospective studies, and 88 retrospective cohort studies. After 2 rounds of surveys, 24 recommendations and 5 good clinical practice statements were developed, with full consensus among panelists. Recommendations focused on tumor and functional imaging characteristics, blood-based biomarkers, and carcinoid heart disease. A single strong recommendation was made for symptomatic carcinoid syndrome informing treatment in midgut neuroendocrine tumors. Conditional recommendations were made to use grade, morphology, primary site, and urinary 5-hydroxyindoleacetic levels to inform treatment. The guidance document was endorsed by the Commonwealth Neuroendocrine Tumour Collaboration and the North American Neuroendocrine Tumor Society. Conclusions and Relevance The study results suggest that select factors have sufficient evidence to inform care in GEP-NENs, but the evidence for most biomarkers is weak. This article may help guide management and identify gaps for future research to advance personalized medicine and improve outcomes for patients with GEP-NENs.
Collapse
Affiliation(s)
- Jonathan M Loree
- BC Cancer, Vancouver Centre, Vancouver, British Columbia, Canada
| | - David Chan
- Northern Clinical School, University of Sydney, Sydney, Australia
- ENETS Centre of Excellence, Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Jennifer Lim
- St George Hospital, Sydney, New South Wales, Australia
- University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Heather Stuart
- University of British Columbia and BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | - Jonathan Koea
- Te Whatu Ora Waitemata and the University of Auckland, Auckland, New Zealand
| | - Jason Posavad
- Canadian Neuroendocrine Tumours Society, Cornwall, Ontario, Canada
| | | | | | - Sten Myrehaug
- Odette Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Boris Naraev
- Tampa General Hospital Cancer Institute, Tampa, Florida
| | - Dale L Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | | | - David Laidley
- Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Veronica Boyle
- School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Oncology, Auckland City Hospital, Te Whatu Ora Tamaki Makaurau, Auckland, New Zealand
| | - Rachel Goodwin
- Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, Ontario, Canada
| | - Jaydi Del Rivero
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael Michael
- NET Unit and ENETS Centre of Excellence, Peter MacCallum Cancer Centre, Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Janice Pasieka
- Section of General Surgery, Division of Endocrine Surgery and Surgical Oncology, Department of Surgery and Oncology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Simron Singh
- University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Odette Cancer Center, Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Sorbye H, Hjortland GO, Vestermark LW, Sundlov A, Assmus J, Couvelard A, Perren A, Langer SW. NETest in advanced high-grade gastroenteropancreatic neuroendocrine neoplasms. J Neuroendocrinol 2024; 36:e13428. [PMID: 38937137 DOI: 10.1111/jne.13428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
Molecular blood biomarkers are lacking for high-grade (HG) gastroenteropancreatic (GEP) neuroendocrine neoplasms (NEN). To histologically distinguish between neuroendocrine carcinoma (NEC), neuroendocrine tumors G3 (NET G3), adenocarcinoma and MINEN is often challenging. The mRNA-based NETest has diagnostic, prognostic and predictive value in neuroendocrine tumors G1-2 but has not been studied in HG GEP-NEN. Patients with advanced HG GEP-NEN were prospectively included in an observational study. A blood sample was collected before the start of chemotherapy and pseudonymised before NETest was performed. NETest results are expressed as an activity index (NETest score) from 0 to 100. The normal score cut-off is 20. Histological sections were pseudonymised before centralized pathological re-evaluation. Samples from 60 patients were evaluable with the NETest. Main primary tumor sites were colon (14), rectum (12), pancreas (11) and esophagus (7). Re-classification: 30 NEC, 12 NET G3, 3 HG-NEN ambiguous morphology, 8 MiNEN, 3 adenocarcinomas with neuroendocrine differentiation (ADNE), 3 adenocarcinomas and 1 NET G2. Elevated NETest (>20) was seen in 38/45 (84%) HG GEP-NEN, all 17 large-cell NEC (100%), 11/13 (85%) small-cell NEC, all ambiguous cases and 7/12 (64%) NET G3. NETest was elevated in 5/8 (63%) MiNEN, 2/3 ADNE, however not in 3 adenocarcinomas. Median survival was 10.2 months (9.6-10.8 95%CI) for evaluable HG GEP-NEN treated with palliative chemotherapy (n = 39), and survival was significantly shorter in patients with NETest >60 with an OS of only 6.5 months. This is the first study to evaluate use of the NETest in advanced HG GEP-NEN. The NETest was almost always elevated in GEP-NEC and in all large-cell NEC. The NETest was also frequently elevated in NET G3 and MiNEN, however cases were limited. Baseline NETest was not predictive for benefit of chemotherapy, however a NETest >60 was prognostic with a shorter survival for patients receiving chemotherapy.
Collapse
Affiliation(s)
- H Sorbye
- Department of Oncology, Haukeland University Hospital and Department of Clinical Science, University of Bergen, Bergen, Norway
| | - G O Hjortland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - A Sundlov
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - J Assmus
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | - A Couvelard
- Department of Pathology, AP-HP Bichat Hospital, Université Paris Cité, Paris, France
| | - A Perren
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - S W Langer
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Mallak N, Yilmaz B, Meyer C, Winters C, Mench A, Jha AK, Prasad V, Mittra E. Theranostics in Neuroendocrine Tumors: Updates and Emerging Technologies. Curr Probl Cancer 2024; 52:101129. [PMID: 39232443 DOI: 10.1016/j.currproblcancer.2024.101129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/22/2024] [Indexed: 09/06/2024]
Abstract
Advancements in somatostatin receptor (SSTR) targeted imaging and treatment of well-differentiated neuroendocrine tumors (NETs) have revolutionized the management of these tumors. This comprehensive review delves into the current practice, discussing the use of the various FDA-approved SSTR-agonist PET tracers and the predictive imaging biomarkers, and elaborating on Lu177-DOTATATE peptide receptor radionuclide therapy (PRRT) including the evolving areas of post-therapy imaging practices, PRRT retreatment, and the potential role of dosimetry in optimizing patient treatments. The future directions sections highlight ongoing research on investigational PET imaging radiotracers, future prospects in alpha particle therapy, and combination therapy strategies.
Collapse
Affiliation(s)
- Nadine Mallak
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Burcak Yilmaz
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Catherine Meyer
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Celeste Winters
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Anna Mench
- Department of Diagnostic Radiology, Medical Physics Section, Oregon Health & Sciences University, Portland, OR, USA
| | - Abhinav K Jha
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA; Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Vikas Prasad
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University, St Louis, MO, US
| | - Erik Mittra
- Department of Diagnostic Radiology, Molecular Imaging and Therapy Section, Oregon Health & Sciences University, Portland, OR, USA.
| |
Collapse
|
7
|
Almeida C, Gervaso L, Frigè G, Spada F, Benini L, Cella CA, Mazzarella L, Fazio N. The Role of Liquid Biopsy in Gastroenteropancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2024; 16:3349. [PMID: 39409968 PMCID: PMC11475604 DOI: 10.3390/cancers16193349] [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: 07/21/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Neuroendocrine neoplasms incidence has been increasing, arising the need for precise and early diagnostic tools. Liquid biopsy (LB) offers a less invasive alternative to tissue biopsy, providing real-time molecular information from circulating tumour components in body fluids. The aim of this review is to analyse the current evidence concerning LB in NENs and its role in clinical practice. We conducted a systematic review in July 2024 focusing on LB applications in NENs, including circulating tumour cells (CTCs), circulating tumour DNA (ctDNA), micro RNA (miRNA), messenger RNA (mRNA) and extracellular vesicles. Sixty-five relevant articles were analysed. The LB showed potential in diagnosing and monitoring NENs. While CTCs face limitations due to low shedding, ctDNA provides valuable information on high-grade neoplasms. MiRNA and mRNA (e.g., the NETest) offer high sensitivity and specificity for diagnosis and prognosis, outperforming traditional markers like chromogranin A. The LB has significant potential for NEN diagnosis and monitoring but lacks widespread clinical integration due to limited prospective studies and guidelines, requiring further validation. Advances in sequencing technologies may enhance the clinical utility of LB in NENs. Future research should focus on refining LB methods, standardising protocols and exploring applications in high-grade NENs.
Collapse
Affiliation(s)
- Catarina Almeida
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
- Department of Medical Oncology, São João University Hospital Center, 4200-319 Porto, Portugal
| | - Lorenzo Gervaso
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
| | - Gianmaria Frigè
- Laboratory of Translational Oncology, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy;
| | - Francesca Spada
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
| | - Lavinia Benini
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
| | - Chiara Alessandra Cella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
| | - Luca Mazzarella
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
- Laboratory of Translational Oncology, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy;
| | - Nicola Fazio
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO IRCCS, 20141 Milan, Italy; (C.A.); (F.S.); (L.B.); (C.A.C.); (L.M.)
| |
Collapse
|
8
|
Sedlack AJH, Varghese DG, Naimian A, Yazdian Anari P, Bodei L, Hallet J, Riechelmann RP, Halfdanarson T, Capdevilla J, Del Rivero J. Update in the management of gastroenteropancreatic neuroendocrine tumors. Cancer 2024; 130:3090-3105. [PMID: 39012928 DOI: 10.1002/cncr.35463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 07/18/2024]
Abstract
Neuroendocrine neoplasms are a diverse group of neoplasms that can occur in various areas throughout the body. Well-differentiated neuroendocrine tumors (NETs) most often arise in the gastrointestinal tract, termed gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Although GEP-NETs are still uncommon, their incidence and prevalence have been steadily increasing over the past decades. The primary treatment for GEP-NETs is surgery, which offers the best chance for a cure. However, because GEP-NETs are often slow-growing and do not cause symptoms until they have spread widely, curative surgery is not always an option. Significant advances have been made in systemic and locoregional treatment options in recent years, including peptide-receptor radionuclide therapy with α and β emitters, somatostatin analogs, chemotherapy, and targeted molecular therapies.
Collapse
Affiliation(s)
- Andrew J H Sedlack
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Diana Grace Varghese
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Amirkia Naimian
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Pouria Yazdian Anari
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Julie Hallet
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, East York, Ontario, Canada
| | | | | | | | - Jaydira Del Rivero
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Zhang H, Tsuchikawa T, Takeuchi S, Hirata K, Tanaka K, Matsui A, Nakanishi Y, Asano T, Noji T, Nakamura T, Takeuchi S, Wada M, Hirano S. Initial validation of the clinical significance of the NETest in Japanese gastroenteropancreatic neuroendocrine tumor patients. Endocr J 2024; 71:873-880. [PMID: 39069495 DOI: 10.1507/endocrj.ej24-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
As novel biomarkers for gastroenteropancreatic neuroendocrine tumors (GEPNET) are in demand, we aimed to validate the clinical value of the NETest in Japanese patients. Between 2021 and 2023, blood and clinical data were collected from patients with GEPNET. Among 35 patients (median age: 59 [49-66] years), 27 cases originated from the pancreas and eight from the gastrointestinal tract. Of 69 samples sent to the laboratory, 56 (81.2%) underwent NETest. The diagnostic sensitivity was 97.1%. Among three patients who underwent R0 resection and four treated with peptide receptor radionuclide therapy, the changes in NETest scores closely correlated with disease progression. The NETest demonstrated high diagnostic efficacy and accurate therapeutic monitoring capabilities in a Japanese population.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Satoshi Takeuchi
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Kimitaka Tanaka
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Aya Matsui
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Yoshitsugu Nakanishi
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Toshimichi Asano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Takehiro Noji
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Shintaro Takeuchi
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Masataka Wada
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
| |
Collapse
|
10
|
De Jesus-Acosta A, Mohindroo C. Genomic Landscape of Pancreatic Neuroendocrine Tumors and Implications for Clinical Practice. JCO Precis Oncol 2024; 8:e2400221. [PMID: 39231376 DOI: 10.1200/po.24.00221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Pancreatic neuroendocrine tumors (pNETs) are the second most prevalent neoplasms of the pancreas with variable prognosis and clinical course. Our knowledge of the genetic alterations in patients with pNETs has expanded in the past decade with the availability of whole-genome sequencing and germline testing. This review will focus on potential clinical applications of the genetic testing in patients with pNETs. For somatic testing, we discuss the commonly prevalent somatic mutations and their impact on prognosis and treatment of patients with pNET. We also highlight the relevant genomic biomarkers that predict response to specific treatments. Previously, germline testing was only recommended for high-risk patients with syndromic features (MEN1, VHL, TSC, and NF1), we review the evolving paradigm of germline testing in pNETs as recent studies have now shown that sporadic-appearing pNETs can also harbor germline variants.
Collapse
Affiliation(s)
- Ana De Jesus-Acosta
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chirayu Mohindroo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
11
|
Bodei L, Jayaprakasam VS, Ying Wong BZ, Aparici CM. Neuroendocrine Tumors: Beta Labeled Radiopeptides. PET Clin 2024; 19:e1-e11. [PMID: 40199623 DOI: 10.1016/j.cpet.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Peptide receptor radionuclide therapy (PRRT) consists of administrating a radiolabeled octreotide derivative that targets somatostatin receptors present on the cell membrane of neuroendocrine tumor cells. Although PRRT was initially performed with 90Y-peptides, currently 177Lu-peptides represent the predominant form of treatment. PRRT results in significant tumor and symptomatic control in patients. Like with other available systemic therapies, responses are relatively short-lived. Several new peptides and strategies to improve the efficacy and tolerability of PRRT have been proposed. A critical step is individualizing treatments based on specific dosimetric estimates for the tumor and normal organs, and determining tissue radiosensitivity.
Collapse
Affiliation(s)
- Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA.
| | - Vetri Sudar Jayaprakasam
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | | | - Carina Mari Aparici
- Division of Nuclear Medicine, Department of Radiology, University of Stanford, Stanford, CA, USA
| |
Collapse
|
12
|
Kusne Y, Lasho T, Finke C, Elsabbagh Z, McCue S, Hobday T, Starr J, Bekaii-Saab T, Halfdanarson TR, Patnaik MM, Ou FS, Sonbol MB. Clonal Hematopoiesis in Patients With Neuroendocrine Tumor Treated With Lutetium-177 and the Risk of Thrombocytopenia: A Prospective Study. JCO Precis Oncol 2024; 8:e2400143. [PMID: 38976813 PMCID: PMC11371079 DOI: 10.1200/po.24.00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 07/10/2024] Open
Abstract
PURPOSE Thrombocytopenia is a relatively common dose-limiting toxicity during peptide receptor radionuclide therapy (PRRT) in patients with NET. Although uncommon, some patients develop persistent cytopenia and eventually therapy-related myeloid neoplasm (t-MN), which has a dismal prognosis. As the indications for PRRT are expanding, it is important to investigate factors that may predict cytopenias during/after PRRT. We prospectively evaluated the prevalence of clonal hematopoiesis (CH) and cytopenia in patients with NET undergoing PRRT. MATERIALS AND METHODS Patients with metastatic NET with plan to receive four cycles of lutetium-177 were enrolled. CH was evaluated before PRRT using a panel of 220 genes with a targeted depth of ≥1,000×. Patients were followed during PRRT and every 3 months thereafter. RESULTS Of 37 patients enrolled, the median age was 68 years and 51.4% were male. Previous treatment exposures included alkylating agents in 30%, platinum agents in 8%, and external radiation in 13%. CH was detected in 35.1% using a variant allele frequency (VAF) cutoff of ≥2% and 45.9% with a VAF of ≥1%. The most common mutations were in age-related genes (DNMT3A, TET2). CH was not associated with anemia or neutropenia; however, it was associated with lower platelet count at baseline and more time spent in a thrombocytopenic state during/after PRRT. Five patients had bone marrow biopsies (BMBs) because of sustained hematologic dysfunction post-PRRT, and of those, diagnoses included clonal cytopenia of undetermined significance (CCUS) in three and idiopathic cytopenia of undetermined significance (ICUS) in two. CONCLUSION CH is present in 35.1% of patients with NET and is associated with thrombocytopenia risk during PRRT. Future studies with long-term follow-up will delineate whether CH might be a predictor for higher risk of t-MN after PRRT.
Collapse
Affiliation(s)
- Yael Kusne
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ
| | - Terra Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Christy Finke
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Zaid Elsabbagh
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ
| | - Shaylene McCue
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Timothy Hobday
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jason Starr
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | | | | | - Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Fang-Shu Ou
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
13
|
Smith TAD. Gene Abnormalities and Modulated Gene Expression Associated with Radionuclide Treatment: Towards Predictive Biomarkers of Response. Genes (Basel) 2024; 15:688. [PMID: 38927624 PMCID: PMC11202453 DOI: 10.3390/genes15060688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Molecular radiotherapy (MRT), also known as radioimmunotherapy or targeted radiotherapy, is the delivery of radionuclides to tumours by targeting receptors overexpressed on the cancer cell. Currently it is used in the treatment of a few cancer types including lymphoma, neuroendocrine, and prostate cancer. Recently reported outcomes demonstrating improvements in patient survival have led to an upsurge in interest in MRT particularly for the treatment of prostate cancer. Unfortunately, between 30% and 40% of patients do not respond. Further normal tissue exposure, especially kidney and salivary gland due to receptor expression, result in toxicity, including dry mouth. Predictive biomarkers to select patients who will benefit from MRT are crucial. Whilst pre-treatment imaging with imaging versions of the therapeutic agents is useful in demonstrating tumour binding and potentially organ toxicity, they do not necessarily predict patient benefit, which is dependent on tumour radiosensitivity. Transcript-based biomarkers have proven useful in tailoring external beam radiotherapy and adjuvant treatment. However, few studies have attempted to derive signatures for MRT response prediction. Here, transcriptomic studies that have identified genes associated with clinical radionuclide exposure have been reviewed. These studies will provide potential features for seeding multi-component biomarkers of MRT response.
Collapse
Affiliation(s)
- Tim A D Smith
- Nuclear Futures Institute, School of Computer Science and Engineering, Bangor University, Dean Street, Bangor LL57 1UT, UK
| |
Collapse
|
14
|
Taunk NK, Escorcia FE, Lewis JS, Bodei L. Radiopharmaceuticals for Cancer Diagnosis and Therapy: New Targets, New Therapies-Alpha-Emitters, Novel Targets. Cancer J 2024; 30:218-223. [PMID: 38753757 PMCID: PMC11232930 DOI: 10.1097/ppo.0000000000000720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Radiopharmaceutical therapy has emerged as a promising approach for the treatment of various cancers. The exploration of novel targets such as tumor-specific antigens, overexpressed receptors, and intracellular biomolecules using antibodies, peptides, or small molecules has expanded the scope of radiopharmaceutical therapy, enabling precise and effective cancer treatment for an increasing number of tumor types. Alpha emitters, characterized by their high linear energy transfer and short path length, offer unique advantages in targeted therapy due to their potent cytotoxicity against cancer cells while sparing healthy tissues. This article reviews recent advancements in identifying novel targets for radiopharmaceutical therapy and applications in utilizing α-emitters for targeted treatment.
Collapse
Affiliation(s)
- Neil K. Taunk
- Department of Radiation Oncology and Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
15
|
Mallak N, O'Brien SR, Pryma DA, Mittra E. Theranostics in Neuroendocrine Tumors. Cancer J 2024; 30:185-193. [PMID: 38753753 DOI: 10.1097/ppo.0000000000000723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Neuroendocrine tumors (NETs) are rare tumors that develop from cells of the neuroendocrine system and can originate in multiple organs and tissues such as the bowels, pancreas, adrenal glands, ganglia, thyroid, and lungs. This review will focus on gastroenteropancreatic NETs (more commonly called NETs) characterized by frequent somatostatin receptor (SSTR) overexpression and pheochromocytomas/paragangliomas (PPGLs), which typically overexpress norepinephrine transporter. Advancements in SSTR-targeted imaging and treatment have revolutionized the management of patients with NETs. This comprehensive review delves into the current practice, discussing the use of the various Food and Drug Administration-approved SSTR-agonist positron emission tomography tracers and the predictive imaging biomarkers, and elaborating on 177Lu-DOTATATE peptide receptor radionuclide therapy including the evolving areas of posttherapy imaging practices and peptide receptor radionuclide therapy retreatment. SSTR-targeted imaging and therapy can also be used in patients with PPGL; however, this patient population has demonstrated the best outcomes from norepinephrine transporter-targeted therapy with 131I-metaiodobenzylguanidine. Metaiodobenzylguanidine theranostics for PPGL will be discussed, noting that in 2024 it became commercially unavailable in the United States. Therefore, the use and reported success of SSTR theranostics for PPGL will also be explored.
Collapse
Affiliation(s)
- Nadine Mallak
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| | - Sophia R O'Brien
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Daniel A Pryma
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Erik Mittra
- From the Department of Diagnostic Radiology, Oregon Health & Sciences University, Portland, OR
| |
Collapse
|
16
|
Strosberg JR, Al-Toubah T, El-Haddad G, Reidy Lagunes D, Bodei L. Sequencing of Somatostatin-Receptor-Based Therapies in Neuroendocrine Tumor Patients. J Nucl Med 2024; 65:340-348. [PMID: 38238038 DOI: 10.2967/jnumed.123.265706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/20/2023] [Indexed: 03/03/2024] Open
Abstract
Most well-differentiated neuroendocrine tumors (NETs) express high levels of somatostatin receptors, particularly subtypes 2 and 5. Somatostatin analogs (SSAs) bind to somatostatin receptors and are used for palliation of hormonal syndromes and control of tumor growth. The long-acting SSAs octreotide long-acting release and lanreotide are commonly used in the first-line metastatic setting because of their tolerable side effect profile. Radiolabeled SSAs are used both for imaging and for treatment of NETs. 177Lu-DOTATATE is a β-emitting radiolabeled SSA that has been proven to significantly improve progression-free survival among patients with progressive midgut NETs and is approved for treatment of metastatic gastroenteropancreatic NETs. A key question in management of patients with gastroenteropancreatic and lung NETs is the sequencing of 177Lu-DOTATATE in relation to other systemic treatments (such as everolimus) or liver-directed therapies. This question is particularly complicated given the heterogeneity of NETs and the near absence of randomized trials comparing active treatment options. This state-of-the-art review examines the evidence supporting use of somatostatin-receptor-targeted treatments within the larger landscape of NET therapy and offers insights regarding optimal patient selection, assessment of benefit versus risk, and treatment sequencing.
Collapse
Affiliation(s)
- Jonathan R Strosberg
- Department of GI Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida;
| | - Taymeyah Al-Toubah
- Department of GI Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ghassan El-Haddad
- Department of Radiology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Diane Reidy Lagunes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
17
|
Bodei L, Michael Tuttle R, Grewal RK, Mauguen A, Augensen F, Abusamra M, Mahajan S, Jayaprakasam VS, Osborne JR, Haque S, Wong BZY, Ghossein RA, Fagin J, Schӧder H, Ho A, Humm JL, Larson SM. Toward individualized dosimetry for radiopharmaceutical therapy in day-to-day clinical practice of nuclear oncology: overcoming heterogeneity of radiation-absorbed dose to tumor and critical organs. Eur J Nucl Med Mol Imaging 2024; 51:325-329. [PMID: 37712994 PMCID: PMC10774147 DOI: 10.1007/s00259-023-06420-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Affiliation(s)
- Lisa Bodei
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - R Michael Tuttle
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravinder K Grewal
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Audrey Mauguen
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Finn Augensen
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Murad Abusamra
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Sonia Mahajan
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Vetri Sudar Jayaprakasam
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Joseph R Osborne
- Division of Molecular Imaging and Therapeutics, Weill Cornell Medical College, New York, NY, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Bernadette Z Y Wong
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Ronald A Ghossein
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heiko Schӧder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA
| | - Alan Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John L Humm
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 77, New York, NY, 2C-21210065, USA.
| |
Collapse
|
18
|
Lee H, Kipnis ST, Niman R, O’Brien SR, Eads JR, Katona BW, Pryma DA. Prediction of 177Lu-DOTATATE Therapy Outcomes in Neuroendocrine Tumor Patients Using Semi-Automatic Tumor Delineation on 68Ga-DOTATATE PET/CT. Cancers (Basel) 2023; 16:200. [PMID: 38201627 PMCID: PMC10778298 DOI: 10.3390/cancers16010200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Treatment of metastatic neuroendocrine tumors (NET) with 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) results in favorable response only in a subset of patients. We investigated the prognostic value of quantitative pre-treatment semi-automatic 68Ga-DOTATATE PET/CT analysis in NET patients treated with PRRT. METHODS The medical records of 94 NET patients who received at least one cycle of PRRT at a single institution were retrospectively reviewed. On each pre-treatment 68Ga-DOTATATE PET/CT, the total tumor volume (TTV), maximum tumor standardized uptake value for the patient (SUVmax), and average uptake in the lesion with the lowest radiotracer uptake (SUVmin) were determined with a semi-automatic tumor delineation method. Progression-free survival (PFS) and overall survival (OS) among the patients were compared based on optimal cutoff values for the imaging parameters. RESULTS On Kaplan-Meier analysis and univariate Cox regression, significantly shorter PFS was observed in patients with lower SUVmax, lower SUVmin, and higher TTV. On multivariate Cox regression, lower SUVmin and higher TTV remained predictive of shorter PFS. Only higher TTV was found to be predictive of shorter OS on Kaplan-Meier and Cox regression analyses. In a post hoc Kaplan-Meier analysis, patients with at least one high-risk feature (low SUVmin or high TTV) showed shorter PFS and OS, which may be the most convenient parameter to measure in clinical practice. CONCLUSIONS The tumor volume and lowest lesion uptake on 68Ga-DOTATATE PET/CT can predict disease progression following PRRT in NET patients, with the former also predictive of overall survival. NET patients at risk for poor outcomes following PRRT can be identified with semi-automated quantitative analysis of 68Ga-DOTATATE PET/CT.
Collapse
Affiliation(s)
- Hwan Lee
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sarit T. Kipnis
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Medicine, Georgetown University, Washington, DC 20007, USA
| | - Remy Niman
- MIM Software Inc., Cleveland, OH 44122, USA
| | - Sophia R. O’Brien
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jennifer R. Eads
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Bryson W. Katona
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Daniel A. Pryma
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
19
|
Li X, Lv X, Li H, Zhang G, Long Y, Li K, Fan Y, Jin D, Zhou F, Liu H. Undifferentially Expressed CXXC5 as a Transcriptionally Regulatory Biomarker of Breast Cancer. Adv Biol (Weinh) 2023; 7:e2300189. [PMID: 37423953 DOI: 10.1002/adbi.202300189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/17/2023] [Indexed: 07/11/2023]
Abstract
This work hypothesizes that some genes undergo radically changed transcription regulations (TRs) in breast cancer (BC), but don't show differential expressions for unknown reasons. The TR of a gene is quantitatively formulated by a regression model between the expression of this gene and multiple transcription factors (TFs). The difference between the predicted and real expression levels of a gene in a query sample is defined as the mqTrans value of this gene, which quantitatively reflects its regulatory changes. This work systematically screens the undifferentially expressed genes with differentially expressed mqTrans values in 1036 samples across five datasets and three ethnic groups. This study calls the 25 genes satisfying the above hypothesis in at least four datasets as dark biomarkers, and the strong dark biomarker gene CXXC5 (CXXC Finger Protein 5) is even supported by all the five independent BC datasets. Although CXXC5 does not show differential expressions in BC, its transcription regulations show quantitative associations with BCs in diversified cohorts. The overlapping long noncoding RNAs (lncRNAs) may have contributed their transcripts to the expression miscalculations of dark biomarkers. The mqTrans analysis serves as a complementary view of the transcriptome-based detections of biomarkers that are ignored by many existing studies.
Collapse
Affiliation(s)
- Xue Li
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Xiaoying Lv
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Haijun Li
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Gongyou Zhang
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Yaohang Long
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Kewei Li
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Yusi Fan
- College of Software, Jilin University, Changchun, 130012, China
| | - Dawei Jin
- Research Institute of Guizhou Huada Life Big Data, Guiyang, Guizhou, 550025, China
| | - Fengfeng Zhou
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
- College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Hongmei Liu
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550025, China
- Engineering Research Center of Medical Biotechnology, Guizhou Medical University, Guiyang, Guizhou, 550025, China
- Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, 130012, China
| |
Collapse
|
20
|
Waldeck K, Van Zuylekom J, Cullinane C, Gulati T, Simpson KJ, Tothill RW, Blyth B, Hicks RJ. A genome-wide CRISPR/Cas9 screen identifies DNA-PK as a sensitiser to 177Lutetium-DOTA-octreotate radionuclide therapy. Theranostics 2023; 13:4745-4761. [PMID: 37771787 PMCID: PMC10526672 DOI: 10.7150/thno.84628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/17/2023] [Indexed: 09/30/2023] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) using 177Lutetium-DOTA-octreotate (LuTate) for neuroendocrine tumours (NET) is now an approved treatment available in many countries, though primary or secondary resistance continue to limit its effectiveness or durability. We hypothesised that a genome-wide CRISPR/Cas9 screen would identify key mediators of response to LuTate and gene targets that might offer opportunities for novel combination therapies for NET patients. Methods: We utilised a genome-wide CRISPR-Cas9 screen in LuTate-treated cells to identify genes that impact on the sensitivity or resistance of cells to LuTate. Hits were validated through single-gene knockout. LuTate-resistant cells were assessed to confirm LuTate uptake and retention, and persistence of somatostatin receptor 2 (SSTR2) expression. Gene knockouts conferring LuTate sensitivity were further characterised by pharmacological sensitisation using specific inhibitors and in vivo analysis of the efficacy of these inhibitors in combination with LuTate. Results: The CRISPR-Cas9 screen identified several potential targets for both resistance and sensitivity to PRRT. Two gene knockouts which conferred LuTate resistance in vitro, ARRB2 and MVP, have potential mechanisms related to LuTate binding and retention, and modulation of DNA-damage repair (DDR) pathways, respectively. The screen showed that sensitivity to LuTate treatment in vitro can be conferred by the loss of a variety of genes involved in DDR pathways, with loss of genes involved in Non-Homologous End-Joining (NHEJ) being the most lethal. Loss of the key NHEJ gene, PRKDC (DNA-PK), either by gene loss or inhibition by two different inhibitors, resulted in significantly reduced cell survival upon exposure of cells to LuTate. In SSTR2-positive xenograft-bearing mice, the combination of nedisertib (a DNA-PK specific inhibitor) and LuTate produced a more robust control of tumour growth and increased survival compared to LuTate alone. Conclusions: DDR pathways are critical for sensing and repairing radiation-induced DNA damage, and our study shows that regulation of DDR pathways may be involved in both resistance and sensitivity to PRRT. Additionally, the use of a DNA-PK inhibitor in combination with LuTate PRRT significantly improves the efficacy of the treatment in pre-clinical models, providing further evidence for the clinical efficacy of this combination.
Collapse
Affiliation(s)
- Kelly Waldeck
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Jessica Van Zuylekom
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Carleen Cullinane
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Twishi Gulati
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
| | - Kaylene J. Simpson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Richard W. Tothill
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
- Department of Clinical Pathology and University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Benjamin Blyth
- Models of Cancer Translational Research Centre, Research Division, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010
| | - Rodney J. Hicks
- St Vincent's Hospital Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia, 3010
| |
Collapse
|