1
|
Hughes DJ, Chand G, Johnson J, Tegala R, Bailey D, Adamson K, Edmonds S, Meszaros LK, Moore AEB, Manickavasagar T, Ndagire S, Gennatas S, Georgiou A, Ghosh S, Josephs D, Karapanagiotou E, McLean E, Ting HH, Spicer J, Goh V, Cook GJR. PD-L1 imaging with [ 99mTc]NM-01 SPECT/CT is associated with metabolic response to pembrolizumab with/without chemotherapy in advanced lung cancer. Br J Cancer 2025; 132:913-921. [PMID: 40188291 DOI: 10.1038/s41416-025-02991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/15/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) immunohistochemistry is a predictive biomarker for anti-PD-(L)1 therapy in non-small cell lung cancer (NSCLC). It is not a reliable predictor of clinical benefit with non-invasive imaging providing a potential solution. We present the PECan study, the aim of which to assess the relationship of [99mTc]-labeled anti-PD-L1 single-domain antibody (NM-01) single-photon emission computed tomography (SPECT)/CT with metabolic response to anti-PD-(L)1. METHODS PD-L1 tumour proportion score (TPS) measured using SP263 assay. [99mTc]NM-01 SPECT/CT and [18F]FDG PET/CT performed before and 9-weeks following pembrolizumab with/without chemotherapy in patients with advanced NSCLC. Tumor (T) to blood pool (BP) maximum region of interest (ROImax) measurements performed in primary and metastatic lesions using SPECT/CT images. RESULTS Fifteen patients were included (median age 63 years, 9 male). Intertumoural heterogeneity evident in 10(67%) patients. Mean [99mTc]NM-01 T:BP demonstrated moderate correlation with PD-L1 TPS (r = 0.45, p < 0.05). Depth of [18F]FDG PET/CT metabolic response at 9-weeks (n = 13), correlated strongly with baseline [99mTc]NM-01 T:BP (r = -0.73, p < 0.05), but only moderately with PD-L1 TPS (r = -0.46, p = 0.06). CONCLUSION [99mTc]NM-01 SPECT/CT allows non-invasive quantification of PD-L1 in primary tumour and metastases in NSCLC. [99mTc]NM-01 uptake moderately correlates with PD-L1 immunohistochemistry, determines heterogeneity, and is associated with early metabolic response to anti-PD-1 pembrolizumab. CLINICAL TRIALS REGISTRATION PD-L1 Expression in Cancer (PECan) study (NCT04436406), registered 18 June 2020 https://clinicaltrials.gov/ct2/show/NCT04436406.
Collapse
Affiliation(s)
- Daniel Johnathan Hughes
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gitasha Chand
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - Jessica Johnson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Ronan Tegala
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Damion Bailey
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Kathryn Adamson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Scott Edmonds
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | - Amelia Elizabeth Broomfield Moore
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
| | - Thubeena Manickavasagar
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Susan Ndagire
- King's Health Partners Cancer Biobank, Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Spyridon Gennatas
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Alexandros Georgiou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Debra Josephs
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Eleni Karapanagiotou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Emma McLean
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hong Hoi Ting
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - James Spicer
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK.
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
2
|
Di Franco M, Lamberti G, Campana D, Ambrosini V. Molecular Imaging for Response Assessment of Neuroendocrine Tumors (NET). Semin Nucl Med 2025:S0001-2998(25)00049-2. [PMID: 40345899 DOI: 10.1053/j.semnuclmed.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025]
Abstract
Assessing treatment response in neuroendocrine tumors (NET) remains a significant challenge due to their typically indolent growth and heterogenity, the frequent occurrence of disease stabilization rather than tumor shrinkage after therapy, and the inherent limitations of conventional imaging criteria. While molecular imaging-primarily somatostatin receptor (SST) PET/CT-has improved lesion detection, the absence of standardized response criteria limits its clinical utility and prevents its use as full replacement of conventional imaging. Emerging strategies, including revised thresholds for dimensional changes, criteria evaluating different features, such as lesions' density and functional tumor volumes, offer potential improvements in response evaluation but require further validation for routine clinical implementation. This review examines the current challenges in assessing NET treatment response, evaluates the strengths and limitations of available imaging modalities, and discusses emerging approaches and future directions for optimizing therapeutic monitoring in the heterogeneous panorama of NET.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy.
| | - Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Davide Campana
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy; Medical Oncology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy; Nuclear Medicine, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
3
|
Adnan A, Basu S. PET-based Quantitative Techniques in Assessing Efficacy of Interventional Radiology Procedures in Oncology. PET Clin 2025:S1556-8598(25)00028-8. [PMID: 40340172 DOI: 10.1016/j.cpet.2025.03.005] [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: 05/10/2025]
Abstract
Interventional radiology (IR) is a super specialised branch where imaging modalities are employed to guide disease specific diagnostic and therapeutic interventions. IR interventions have gained popularity in various oncological and non-oncological indications due to it's ability to effectively diagnose the disease and direct specific targeted treatment. Hybrid imaging using PET CT and PET MRI combines the best of morphological and functional informations and offers improved sensitivity and specificity for detection of lesion; helps in accurate mapping of tumour burden, thereby aiding in planning curative vs palliative intent intervention; more accurate response evaluation to plan redo session in cases of residual / recurrent disease or for follow up evaluation and for prognostication and predicting response. Albeit visual analysis of PET images by specialist is most commonly performed for reading PET scans, PET has a remarkable capability to provide quantitative information. The present review provides a comprehensive assessment of the role of various aspects of quantitative PET parameters in assessing the efficacy of IR interventions. The insights provided will help clinicians, researchers, and medical professionals understand the role of PET imaging in advancing patient care and enhancing the therapeutic outcomes of IR procedures.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, Maharahtra, India; Homi Bhabha National Institute, Mumbai, India; Department of Nuclear Medicine, Medica Cancer Center, Medica Superspeciality Hospital, 127, Eastern Metropolitean Bypass, Mukundapur, Kolkata, West Bengal, India
| | - Sandip Basu
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, Maharahtra, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
4
|
Leath CA, Deng W, Mell LK, Richardson DL, Walker JL, Holman LL, Lea JS, Amarnath SR, Santos-Reyes LJ, Arend RC, Mayadev J, Jegadeesh N, DiSilvestro P, Chon HS, Ghamande SA, Gao L, Albuquerque K, Chino JP, Donnelly E, Feddock JM, Lowenstein J, Quick AM, Kunos CM, MacKay H, Aghajanian C, Monk BJ. Incorporation of triapine (T) to cisplatin chemoradiation (CRT) for locally advanced cervical and vaginal cancer: Results from NRG-GY006, a phase III randomized trial. Gynecol Oncol 2025; 195:122-133. [PMID: 40101606 PMCID: PMC12009185 DOI: 10.1016/j.ygyno.2025.03.007] [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: 01/05/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Cisplatin-based chemoradiation (CRT) plus brachytherapy for locally advanced cervical cancer (LACC) is standard. Intrinsic overexpression of ribonucleotide reductase (RNR) may enhance DNA damage repair from CRT. We report on outcomes of adding RNR inhibitor, triapine (T), to CRT. METHODS NRG-GY006 is an open-label randomized phase III trial. FIGO 2009 LACC (stages IB2, II, IIIB or IVA) without para-aortic nodal involvement or stages II-IV vaginal cancer were eligible. Random assignment to CRT or in combination with thrice-weekly T (CRT + T) occurred. Radiation consisted of either 3D conformal (3DCRT) or image-guided intensity modulated RT (IG-IMRT) followed by intracavitary brachytherapy. Primary endpoint was overall survival (OS). Progression-free survival (PFS) was secondary. Exploratory endpoints included complete metabolic response rate on post treatment PET/CT imaging and comparative toxicity and outcomes for 3DCRT vs. IG-IMRT. FINDINGS Four-hundred-fifty patients were randomized including 448 eligible (224 in CRT and 224 in CRT + T). Median age was 47 (range 23-85). The majority had cervical cancer (93.3 %) with squamous histology (82 %). 52 % had FIGO stage II disease. Racial/ethnic distribution included non-Hispanic white (53.8 %), black (15.2 %) and Hispanic/Latina (22.5 %). At randomization, IG-IMRT was planned in 74.3 % and HDR brachytherapy in 98.2 %. No differences in Grade 3-5 toxicities were observed: CRT: 52 % and CRT + T: 49 %, with two G5 toxicities (cardiac arrest and acidosis) in the CRT + T arm. The median patient follow-up was 28 months (IQR 15-45). HR for death was 1.018 (95 % CI 0.634-1.635) while HR for progression was 1.021 (95 % CI 0.694-1.501). INTERPRETATION Triapine added to CRT did not improve OS.
Collapse
Affiliation(s)
- Charles A Leath
- University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, United States of America.
| | - Wei Deng
- NRG Statistical Center, Roswell Park Cancer Institute, Buffalo, NY, United States of America.
| | - Loren K Mell
- University of California San Diego, San Diego, CA, United States of America.
| | - Debra L Richardson
- University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Oklahoma City, OK, United States of America.
| | - Joan L Walker
- University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Oklahoma City, OK, United States of America.
| | - Laura L Holman
- University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Oklahoma City, OK, United States of America.
| | - Jayanthi S Lea
- University of Texas-Southwestern Medical Center, Dallas, TX, United States of America.
| | - Sudha R Amarnath
- Cleveland Clinic Foundation, Cleveland, OH, United States of America.
| | | | - Rebecca C Arend
- University of Alabama at Birmingham, O'Neal Comprehensive Cancer Center, Birmingham, AL, United States of America.
| | - Jyoti Mayadev
- University of California San Diego, San Diego, CA, United States of America.
| | - Naresh Jegadeesh
- John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, United States of America.
| | - Paul DiSilvestro
- Women & Infant's Hospital of Rhode Island, Legorreta Cancer Center at Brown University, Providence, RI, United States of America.
| | - Hye Sook Chon
- H. Lee Moffitt Cancer Center, Tampa, FL, United States of America.
| | | | - Lei Gao
- University of California San Diego, San Diego, CA, United States of America.
| | - Kevin Albuquerque
- University of Texas-Southwestern Medical Center, Dallas, TX, United States of America.
| | - Junzo P Chino
- Duke Cancer Center, Department of Radiation Oncology, Durham, NC, United States of America.
| | - Eric Donnelly
- Northwestern University, Chicago, IL, United States of America.
| | | | - Jessica Lowenstein
- MD Anderson Cancer Center, Imaging and Radiation Oncology Core (IROC), Houston, TX, United States of America.
| | - Allison M Quick
- The Ohio State University Medial Center, Columbus, OH, United States of America.
| | - Charles M Kunos
- University of Miami Miller School of Medicine, Miami, FL, United States of America.
| | - Helen MacKay
- Sunnybrook Odette Cancer Centre/University of Toronto, Toronto, CA, United States of America.
| | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America.
| | - Bradley J Monk
- Florida Cancer Specialists and Research Institute, West Palm Beach, FL, United States of America.
| |
Collapse
|
5
|
Vera P, Giraud P, Hapdey S, Gouel P, Jan O, Le Roux P, Langlais A, Lévêque E, Le Tinier F, Olivier A, Martin E, Berriolo-Riedinger A, Pourel N, Broglia JM, Boisselier P, Guillemard S, Salem N, Brenot-Rossi I, Garcia C, Berthold C, Giroux-Leprieur E, Moreau D, Guillerm S, Benali K, Tessonnier L, Audigier-Valette C, Lerouge D, Quak E, Massabeau C, Courbon F, Loo M, Larrouy A, Ghazzar N, Chaumet-Riffaud P, Amour E, Zalcman G, Modzelewski R, Thureau S. Prognostic Value of FDG PET Metabolic Parameters Before and After 42 Gy of Radiochemotherapy in Patients with Inoperable Stage III Nonsmall Cell Lung Cancer. J Nucl Med 2025; 66:516-524. [PMID: 40015915 DOI: 10.2967/jnumed.124.268499] [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: 08/29/2024] [Accepted: 01/24/2025] [Indexed: 03/01/2025] Open
Abstract
The purpose of this study was to assess the prognostic value of 18F-FDG PET parameter variation between baseline and 42 Gy (PET2) of radiochemotherapy at 6 mo and 1 y of evaluation in patients with stage III inoperable nonsmall cell lung cancer based on RECIST 1.1. Methods: In total, 158 patients in a prospective multicenter phase II/III study were analyzed. Patients were randomized into 2 groups: an experimental arm (group A) and a standard arm (group B). Patients from group A with residual metabolism on PET2 (group A+) at 42 Gy received a radiation boost (74 Gy). Patients without residual uptake on 18F-FDG PET at 42 Gy (group A-) and patients in group B received a standard radiotherapy dose (66 Gy). We compared group A with group B. The 18F-FDG PET parameters SUVmax, SUVmean, SUVpeak, peak SUV normalized on lean body mass, mean SUV normalized on lean body mass, total lesion glycolysis, total metabolic tumor volume (MTV) (tumor and nodes), and tumor MTV were measured. All patients were evaluated with RECIST 1.1 using CT at 6 mo and 1 y after radiochemotherapy. Progression-free survival and overall survival were evaluated. Results: Except for the radiotherapy dose (P < 0.001), patient demographic characteristics were similar between the 2 groups (A vs. B). All 18F-FDG PET uptake and volume parameter measurements were correlated. Therefore, only the change in SUVmax (ΔSUVmax) and total MTV were selected for the analysis. There was no significant difference in any variable between the 2 groups. In the multivariate analysis, ΔSUVmax appeared to be the most important prognostic factor for overall survival, and SUVmax of PET2 appeared to be the most important prognostic factor for progression-free survival. Conclusion: 18F-FDG PET at 42 Gy can be used to identify good responders to radiochemotherapy in patients with inoperable stage III nonsmall cell lung cancer. The SUVmax of PET2 and ΔSUVmax are independent prognostic factors.
Collapse
Affiliation(s)
- Pierre Vera
- Nuclear Medicine, QuantIF-LITIS (EA4108), Centre Henri Becquerel, Rouen, France;
| | - Philippe Giraud
- Radiotherapy, Université Paris Cité, European Hospital Georges-Pompidou, AP-HP, Paris, France
| | | | | | - Orianne Jan
- Nuclear Medicine, Centre Henri Becquerel, Rouen, France
| | - Paul Le Roux
- Nuclear Medicine, Centre Henri Becquerel, Rouen, France
| | | | - Emilie Lévêque
- Clinical Research, Centre Henri Becquerel, Rouen, France
| | | | - Anaïs Olivier
- Nuclear Medicine, Centre Oscar Lambret, Lille, France
| | - Etienne Martin
- Radiotherapy, Centre Georges-Françsois Leclerc, Dijon, France
| | | | - Nicolas Pourel
- Oncologie-thoracique, Institut du Cancer, Avignon, France
| | | | - Pierre Boisselier
- Radiotherapy, Institut du Cancer de Montpellier, Montpellier, France
| | - Sophie Guillemard
- Nuclear Medicine, Institut du Cancer de Montpellier, Montpellier, France
| | - Naji Salem
- Radiotherapy, Institut Paoli-Calmettes, Marseille, France
| | | | - Camilo Garcia
- Nuclear Medicine Department, Gustave Roussy, Villejuif, France
| | - Céline Berthold
- Radiation Oncology Department, Gustave Roussy, Villejuif, France
| | - Etienne Giroux-Leprieur
- Department of Thoracic Oncology, AP-HP, Ambroise Paré Hospital, Boulogne-Billancourt, France
| | - Damien Moreau
- Radiotherapy, European Hospital Georges-Pompidou, AP-HP, Paris, France
| | | | - Khadija Benali
- Nuclear Medicine, Bichat-Claude Bernard, AP-HP Nord, Paris, France
| | | | | | | | - Elske Quak
- Nuclear Medicine, Centre Francois Baclesse, Caen, France
| | | | | | - Maxime Loo
- Radiotherapy, Hopital René Huguenin, Institut Curie, Saint-Cloud, France
| | - Anne Larrouy
- Radiotherapy, Centre de Cancerologie Paris Nord, Sarcelles, France
| | - Nadia Ghazzar
- Nuclear Medicine, Université Paris Cité, European Hospital Georges-Pompidou, AP-HP, Paris, France
| | | | | | - Gérard Zalcman
- Thoracic Oncology Department, Université Paris Cité, CIC INSERM 1425, Hôpital Bichat-Claude Bernard, AP-HP Nord, Paris, France; and
| | | | | |
Collapse
|
6
|
Zhou L, Barros E Silva MJ, Hsiao E, Eroglu Z, Sandhu S, Samoylenko I, Lo SN, Carlino MS, Au-Yeung G, Gonzalez M, Spillane AJ, Pennington TE, Shannon KF, Kapoor R, Burton EM, Tawbi HA, Amaria RN, Blank CU, Duprat JP, Brito de Paula R, Gyorki DE, Saw RPM, Ch'Ng S, Rawson RV, Scolyer RA, Pires da Silva I, Akkooi ACJV, Long GV, Menzies AM. FDG-PET associations with pathological response and survival with neoadjuvant immunotherapy for melanoma. J Immunother Cancer 2025; 13:e011483. [PMID: 40132907 PMCID: PMC11938238 DOI: 10.1136/jitc-2025-011483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Neoadjuvant immunotherapy has become the new standard of care for stage III melanoma. This study sought to describe the metabolic changes seen with fludeoxyglucose-18-positron emission tomography (FDG-PET) following neoadjuvant immunotherapy in patients with melanoma and explore associations with pathological response and recurrence-free survival (RFS). METHODS Data from patients with macroscopic stage III nodal melanoma treated with neoadjuvant checkpoint inhibitor therapy were pooled from five melanoma centers. All patients underwent baseline and preoperative FDG-PET and CT assessments, and all had surgery. Pathological response was determined using the International Neoadjuvant Melanoma Consortium criteria, radiological response using Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and FDG-PET response using European Organization for Research and Treatment of Cancer (EORTC) criteria. The primary endpoint was to explore the associations of metabolic and radiological responses with pathological response; secondary endpoints were RFS outcomes stratified by each response category. RESULTS 115 patients were included, 69% male, median age 59 years (27-92), 43% BRAF mutant, and median follow-up was 22.2 months (95% CI 13.7 to 26.4). 40 patients received anti-PD-1 monotherapy, 20 patients received pembrolizumab combined with lenvatinib, and 55 patients received ipilimumab and nivolumab. The major pathological response (MPR) rate was 62%, and the pathological complete response rate was 51%. RECIST response underestimated pathological response; patients achieving RECIST stable disease (38%) had a 50% chance of achieving MPR. The FDG-PET metabolic response rate was 73%, with most achieving an MPR (80%), especially in patients with a complete metabolic response (CMR, 96% MPR). A small proportion of patients (10%) had stable metabolic disease on FDG-PET, and all these patients were non-MPR. Patients with progressive metabolic disease were also in the majority non-MPR (79%). Patients with MPR, complete response/partial response on CT, and CMR/partial metabolic response on FDG-PET had a favorable 24-month RFS (95.6%, 97.3%, and 93.7%, respectively), with FDG-PET able to identify a greater proportion of patients with favorable progression-free survival (PFS) than pathology or CT (73%, 62%, and 43%, respectively). CONCLUSION Neoadjuvant immunotherapy has high FDG-PET response rates in melanoma. FDG-PET response associates with pathological response and confers impressive RFS, suggesting this could be an important clinical tool.
Collapse
Affiliation(s)
- Li Zhou
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing, China
| | | | - Edward Hsiao
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Zeynep Eroglu
- H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Sir Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Igor Samoylenko
- N N Blokhin Russian Cancer Research Center, Moscow, Russian Federation
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Westmead and Blacktown Hospitals, Sydney, New South Wales, Australia
| | - George Au-Yeung
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Sir Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| | - Thomas E Pennington
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Rony Kapoor
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | | | - Hussein A Tawbi
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rodabe N Amaria
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - David E Gyorki
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Sir Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Sydney Ch'Ng
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Robert V Rawson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Groheux D, Vaz SC, de Geus-Oei LF, Dibble EH, Ulaner GA, Cook GJR, Hindié E, Poortmans P, Mann RM, Jacene H, Pilkington Woll JP, Rubio IT, Vrancken Peeters MJ, Graff SL, Cardoso F. 18F-Labeled Fluorodeoxyglucose-Positron Emission Tomography/Computed Tomography in Staging and Restaging Patients With Breast Cancer. J Clin Oncol 2025:JCO2401945. [PMID: 40132148 DOI: 10.1200/jco-24-01945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 03/27/2025] Open
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France
- University Paris-Diderot, INSERM U976, Paris, France
- Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France
| | - Sofia C Vaz
- Department of Nuclear Medicine and Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
- Department of Radiation Science & Technology, Delft University of Technology, the Netherlands
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI
| | - Gary A Ulaner
- Department of Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA
- Departments of Radiology and Translational Genomics, University of Southern Caliifornia, Los Angeles, CA
| | - Gary J R Cook
- Department of Cancer Imaging, King's College London, London, United Kingdom
- King's College London and Guy's & St Thomas' PET Centre, London, United Kingdom
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Elif Hindié
- Department of Nuclear Medicine, Bordeaux University Hospital, University of Bordeaux, Bordeaux, France
- Institut Universitaire de France (IUF), Paris, France
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, Belgium
- Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk-Antwerp, Belgium
| | - Ritse M Mann
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Radiology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Heather Jacene
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | | | - Isabel T Rubio
- Department of Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Spain
- Cancer Center Clinica Universidad de Navarra, Spain
| | - Marie-Jeanne Vrancken Peeters
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Stephanie L Graff
- Brown University Health Cancer Institute, Providence, RI
- Legorreta Cancer Center at Brown University, Providence, RI
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
- Advanced Breast Cancer Global Alliance, Lisbon, Portugal
| |
Collapse
|
8
|
Smeenk MM, van Diessen JN, Boellaard TN, Hartemink KJ, de Vries JF, van der Noort V, Badrising SK, Owers EC, Monkhorst K, van den Heuvel MM, Theelen WS. Tremelimumab plus Durvalumab prior to Chemoradiotherapy in Unresectable, Locally Advanced Non-Small Cell Lung Cancer: The Induction Trial. Clin Cancer Res 2025; 31:1037-1046. [PMID: 39821070 PMCID: PMC11911803 DOI: 10.1158/1078-0432.ccr-24-3476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/05/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE The phase I induction trial (NCT04287894) assessed the feasibility and safety of induction immunotherapy (IIT) prior to concurrent chemoradiotherapy (cCRT) in patients with locally advanced non-small cell lung cancer (NSCLC). PATIENTS AND METHODS Patients with unresectable stage II/III NSCLC were eligible for inclusion. Patients received either one cycle of tremelimumab (75 mg) with two cycles of durvalumab (1,500 mg) in cohort I, one cycle of tremelimumab (300 mg) with two cycles of durvalumab in cohort II, or one cycle of tremelimumab (300 mg) with one cycle of durvalumab in cohort III. After IIT, a comprehensive radiological and pathological restaging was performed followed by cCRT. The combined primary endpoint was the feasibility and safety of IIT-cCRT. RESULTS Fifteen of 17 included patients were treated per protocol. IIT-cCRT was completed in 13 of the 15 patients within the predefined feasibility criteria. Grade ≥3 immune-related adverse events occurred in seven of the 15 patients, of which six were treated in the high-dose tremelimumab cohorts, thereby violating the safety criteria in cohorts II and III. The low-dose tremelimumab cohort (I) complied with safety criteria. Eleven patients had multilevel N2 or N3 disease at baseline; eight of these patients were downstaged to either N0/N1 or single-level N2 after IIT. Multiparametric MRI accurately identified nodal downstaging in all seven patients. CONCLUSIONS Induction with high-dose tremelimumab plus durvalumab prior to cCRT in unresectable locally advanced NSCLC was associated with unacceptable toxicity, although IIT resulted in clinically relevant nodal downstaging in eight of the 11 patients with baseline multilevel N2 or N3 disease. Multiparametric MRI showed potential for evaluating treatment response.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Female
- Male
- Middle Aged
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Aged
- Chemoradiotherapy/methods
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Adult
- Neoplasm Staging
- Treatment Outcome
Collapse
Affiliation(s)
- Michiel M. Smeenk
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Judi N.A. van Diessen
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Thierry N. Boellaard
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Koen J. Hartemink
- Department of Surgery, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Thoracic Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeltje F. de Vries
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Sushil K. Badrising
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Emilia C. Owers
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kim Monkhorst
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | |
Collapse
|
9
|
Badarna M, Keidar Z, Arnon-Sheleg E. Current and Future Perspective of PET/CT in Response Assessment of Malignant Pleural Mesothelioma. Semin Nucl Med 2025; 55:252-263. [PMID: 40021361 DOI: 10.1053/j.semnuclmed.2025.02.001] [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: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
Malignant pleural mesothelioma (MPM) is a rare but aggressive cancer characterized by its unique growth patterns, presenting substantial diagnostic challenges. With the shift toward immunotherapy for MPM treatment, assessing therapeutic responses has become increasingly complex. Recent studies indicate that FDG PET/CT may provide more effective response criteria compared to traditional CT-based methods. This review emphasizes the important role of PET/CT in offering deep insights into the disease state and monitoring treatment responses. It also addresses the challenges associated with current imaging criteria, particularly the nonspecificity of FDG uptake that may represent inflammatory responses following treatments or procedures rather than tumor activity. Furthermore, the review discusses the potential of emerging radiopharmaceuticals and advanced volumetric assessments, discussing their implications for improving diagnostic accuracy and treatment evaluation in MPM.
Collapse
Affiliation(s)
- Manar Badarna
- Department of Nuclear Medicine, Rambam Healthcare Campus, Haifa, Israel
| | - Zohar Keidar
- Department of Nuclear Medicine, Rambam Healthcare Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion - The Israeli Institute of Technology, Haifa, Israel.
| | - Elite Arnon-Sheleg
- Departments of Nuclear Medicine and Diagnostic Radiology, Galilee Medical Center, Nahariya, and the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
10
|
Sarikaya I. Radionuclide treatments of cancer: molecular mechanisms, biological responses, histopathological changes, and role of PET imaging. Nucl Med Commun 2025; 46:193-203. [PMID: 39654504 DOI: 10.1097/mnm.0000000000001941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Radiation treatments [radiotherapy and radionuclide treatments (RNTs)] are one of the main and effective treatment modalities of cancer. Globally, the number of cancer patients treated with radionuclides are much less as compared to number of radiotherapy cases but with the development of new radiotracers, most notably 177 Lu and 225 Ac-labeled prostate-specific membrane antigen ligands, and 223 Ra-dichloride for prostate cancer and 177 Lu-somatostatin analogs for neuroendocrine tumors, there is a significant rise in RNTs in the last decade. As therapeutic applications of nuclear medicine is on the rise, the aim of this review is to summarize biological responses to radiation treatments and molecular mechanisms of radiation-induced cell death (e.g. ionization, DNA damages such as double-strand breaks, DNA repair mechanisms, types of cell deaths such as apoptosis, necrosis, and immunogenic cell death), histopathological changes with radiation treatments, and role of PET imaging in RNTs as part of radionuclide theranostics for selecting and planning patients for RNTs, dosimetry, predicting and assessing response to RNTs, predicting toxicities, and other possible PET findings which may be seen after RNTs such as activation of immune system.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kirklareli University, Kirklareli, Turkey
| |
Collapse
|
11
|
Hribernik N, Strasek K, Studen A, Zevnik K, Skalic K, Jeraj R, Rebersek M. Early-time-point 18F-FDG-PET/CT and other prognostic biomarkers of survival in metastatic melanoma patients receiving immunotherapy. Radiol Oncol 2025; 59:43-53. [PMID: 40014787 PMCID: PMC11867565 DOI: 10.2478/raon-2025-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 01/04/2024] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND A considerable proportion of metastatic melanoma (mM) patients do not respond to immune checkpoint inhibitors (ICIs). There is a great need to develop noninvasive biomarkers to detect patients, who do not respond to ICIs early during the course of treatment. The aim of this study was to evaluate the role of early [18F]2fluoro-2-deoxy-D-glucose PET/CT (18F-FDG PET/CT) at week four (W4) and other possible prognostic biomarkers of survival in mM patients receiving ICIs. PATIENTS AND METHODS . In this prospective noninterventional clinical study, mM patients receiving ICIs regularly underwent 18F-FDG PET/CT: at baseline, at W4 after ICI initiation, at week sixteen and every 16 weeks thereafter. The tumor response to ICIs at W4 was assessed via modified European Organisation for Research and Treatment of Cancer (EORTC) criteria. Patients with progressive metabolic disease (PMD) were classified into the no clinical benefit group (no-CB), and those with other response types were classified into the clinical benefit group (CB). The primary end point was survival analysis on the basis of the W4 18F-FDG PET/CT response. The secondary endpoints were survival analysis on the basis of LDH, the number of metastatic localizations, and immune-related adverse events (irAEs). Kaplan-Meier analysis and univariate Cox regression analysis were used to assess the impact on survival. RESULTS Overall, 71 patients were included. The median follow-up was 37.1 months (952% CI = 30.1-38.0). Three (4%) patients had only baseline scans due to rapid disease progression and death prior to W4 18F-FDG-PET/CT. Fifty-one (72%) patients were classified into the CB group, and 17 (24%) were classified into the no-CB group. There was a statistically significant difference in median overall survival (OS) between the CB group (median OS not reached [NR]; 95% CI = 17.8 months - NR) and the no-CB group (median OS 6.2 months; 95% CI = 4.6 months - NR; p = 0.003). Univariate Cox analysis showed HR of 0.4 (95% CI = 0.18 - 0.72; p = 0.004). median OS was also significantly longer in the group with normal serum LDH levels and the group with irAEs and cutaneous irAEs. CONCLUSIONS Evaluation of mM patients with early 18F-FDG-PET/CT at W4, who were treated with ICIs, could serve as prognostic imaging biomarkers. Other recognized prognostic biomarkers were the serum LDH level and occurrence of cutaneous irAEs.
Collapse
Affiliation(s)
- Nezka Hribernik
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Strasek
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Studen
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
- Experimental Particle Physics Department, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Katarina Zevnik
- Department of Nuclear Medicine, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Katja Skalic
- Department of Nuclear Medicine, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Robert Jeraj
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
- University of Wisconsin Carbone Cancer Centre, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
- Experimental Particle Physics Department, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Martina Rebersek
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
12
|
Yang R, Tang H, Xie Y, Cai D, He Y, Zheng Z, Lin Y, Gao H, Tang W, Yan Y, Tan L, Shi H. [ 18F]FDG PET/CT for predicting neoadjuvant PD-L1 blockade monotherapy treatment response in patients with locally advanced esophageal squamous cell carcinoma: a preliminary study. Eur J Nucl Med Mol Imaging 2025; 52:1422-1435. [PMID: 39743618 DOI: 10.1007/s00259-024-07051-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE To investigate the predictive value of 2-[18F]-fluoro-2-deoxy-D-glucose ([18F]FDG) PET/CT for evaluating primary tumor (PT) and lymph node (LN) responses after neoadjuvant programmed death-ligand 1 (PD-L1) blockade monotherapy in patients with locally advanced esophageal squamous cell carcinoma (LA-ESCC). METHODS In the single-arm phase 1b NATION-1907 trial (NCT04215471), 23 patients with LA-ESCC received two cycles of neoadjuvant PD-L1 blockade Adebrelimab followed by surgery. Among these, 18 patients underwent [18F]FDG PET/CT scans both before immunotherapy and prior to surgery. Standardized uptake value corrected for lean body mass (SUL)-derived parameters, including SULmax and SULpeak, were documented for PTs and LNs. Lesions > 1cm3 were segmented using thresholds of 41% and 50% of SULmax, respectively, following European Association of Nuclear Medicine (EANM) guidelines, with metabolic tumor volume (MTV) and total lesion glycolysis (TLG) calculated. Percentage changes of all metabolic parameters were also recorded. Residual viable tumor ≤ 33% were classified as well-responders, whereas residual viable tumor > 33% were classified as poor-responders based on histological evaluation. RESULTS In the PT analysis, 10 patients were classified as PT well-responders and 8 as PT poor-responders. All post-treatment metabolic parameters, except MTV, were significantly lower in well-responders compared to poor-responders. The %ΔMTV, %ΔTLG were significantly higher in the poor-responder group (all P < 0.05). ROC curves indicated %ΔMTV41 exhibited optimum performance in predicting well-responders, with an AUC of 0.875 (cut-off: -31.01). Furthermore, %ΔMTV41 significantly predicted patients' recurrence-free survival (RFS) (P < 0.1). In the LN analysis, 7 LNs were classified as well-responders and 10 as poor-responders. Pre-treatment SULmax, SULpeak were significantly lower in poor-responders compared to well-responders. Post-treatment MTV50 and all percentage changes in parameters were significantly higher in the poor-responder group (all P < 0.05). Receiver operating characteristic curve (ROC) analysis indicated %ΔTLG50 had excellent predictive performance for well-responders, with an AUC of 1.000 (cut-off: -7.5). However, there was no significant correlation between the metabolic response evaluations for PTs and LNs. CONCLUSION The metabolic parameters of [18F]FDG PET/CT, particularly %ΔMTV and %ΔTLG, could effectively predict well-responders among both PTs and LNs to neoadjuvant PD-L1 blockade monotherapy in LA-ESCC, which may facilitate personalized immunotherapy and serve as a stratification tool in future larger-scale studies.
Collapse
Affiliation(s)
- Runjun Yang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Han Tang
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
| | - Yunze Xie
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Danjie Cai
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yibo He
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhe Zheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu Lin
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huaping Gao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wenxin Tang
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yihan Yan
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lijie Tan
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China.
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, No. 180 in Fenglin Road, Shanghai, 200032, P.R. China.
- Shanghai Institute of Medical Imaging, Shanghai, 200032, China.
- Nuclear Medicine Institute of Fudan University, Shanghai, 200032, China.
- Cancer Prevention and Treatment Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Lopci E. Current Status of Staging and Restaging Malignant Pleural Mesothelioma. Semin Nucl Med 2025; 55:240-251. [PMID: 39934006 DOI: 10.1053/j.semnuclmed.2025.01.003] [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: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/13/2025]
Abstract
Malignant pleural mesothelioma (MPM) is the most frequent aggressive tumor affecting the pleura, accounting for over 38,000 deaths worldwide. It originates from the mesothelial cells and is mostly associated to asbestos exposure. Depending on the extent of the disease, the management of MPM varies from surgical intervention to a combination of systemic chemotherapy, immunotherapy, and radiation therapy. Major International scientific societies provide continuous updates on proper management of the disease, including recommendations on the optimal imaging algorithms, which are crucial for determining effective treatment options and optimizing clinical outcomes. However, despite the continuous efforts to improve patients' prognosis, median overall survival remains poor, ranging from 8 to 14 months. And even in case of initial response to treatment, local or distant recurrences represent almost a certainty, requiring appropriate imaging for the assessment of tumor sites. The aim of the present article is to illustrate the current status of imaging for staging and restaging of MPM, not forgetting most recent novelties in the diagnostic work-up of the disease.
Collapse
Affiliation(s)
- Egesta Lopci
- Nuclear Medicine Unit, IRCCS - Humanitas Research Hospital, Rozzano Milano, Italy.
| |
Collapse
|
14
|
Torkaman M, Jemaa S, Fredrickson J, Fernandez Coimbra A, De Crespigny A, Carano RAD. Comparative analysis of intestinal tumor segmentation in PET CT scans using organ based and whole body deep learning. BMC Med Imaging 2025; 25:52. [PMID: 39962481 PMCID: PMC11834234 DOI: 10.1186/s12880-025-01587-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND 18-Fluoro-deoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) is a valuable imaging tool widely used in the management of cancer patients. Deep learning models excel at segmenting highly metabolic tumors but face challenges in regions with complex anatomy and normal cell uptake, such as the gastro-intestinal tract. Despite these challenges, it remains important to achieve accurate segmentation of gastro-intestinal tumors. METHODS Here, we present an international multicenter comparative study between a novel organ-focused approach and a whole-body training method to evaluate the effectiveness of training data homogeneity in accurately identifying gastro-intestinal tumors. In the organ-focused method, the training data is limited to cases with intestinal tumors which makes the network trained with more homogeneous data and with stronger presence of intestinal tumor signals. The whole body approach extracts the intestinal tumors from the results of a model trained on the whole-body scans. Both approaches were trained using diffuse large B cell (DLBCL) patients from a large multi-center clinical trial (NCT01287741). RESULTS We report an improved mean(±std) Dice score of 0.78(±0.21) for the organ-based approach on the hold-out set, compared to 0.63(±0.30) for the whole-body approach, with the p-value of less than 0.0001. At the lesion level, the proposed organ-based approach also shows increased precision, recall, and F1-score. An independent trial was used to evaluate the generalizability of the proposed method to non-Hodgkin's lymphoma (NHL) patients with follicular lymphoma (FL). CONCLUSION Given the variability in structure and metabolism across tissues in the body, our quantitative findings suggest organ-focused training enhances intestinal tumor segmentation by leveraging tissue homogeneity in the training data, contrasting with the whole-body training approach, which, by its very nature, is a more heterogeneous data set.
Collapse
Affiliation(s)
| | | | | | | | | | - Richard A D Carano
- Genentech, Inc, South San Francisco, CA, USA
- F. Hoffman-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
15
|
Zwarthoed C, Jaraudias C, Evesque L, Baron D, François E, Chardin D, Marie L, Mitrea D, Château Y, Gal J, Bailleux C. Prognostic Values of Pre- and Post-Therapeutic FDG-PET in Anal Canal Cancer: Analysis of a Prospective Study. Clin Colorectal Cancer 2025:S1533-0028(25)00017-9. [PMID: 39988512 DOI: 10.1016/j.clcc.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The aim of this post hoc study was to assess the prognostic value of 18F-FDG PET/CT quantitative parameters recorded before and after treatment for anal canal neoplasm for the disease free survival. MATERIALS AND METHODS Consecutive, previously untreated patients with histologically proved anal cancer, with 18F-FDG PET/CT pre- and 2 months post treatment were included. The following criteria were analyzed: baseline primary tumor lesion glycolysis (TLG), metabolic tumor volume (MTV), standardized tumor volume (SUV) max and mean, SUV normalized by lean body mass (SUL) max, mean and peak, variations between pre- and post-treatment examinations for SUVmax (Delta SUVmax), TLG (Delta TLG), MTV (Delta MTV), as well as post-treatment SULpeak and SUVmax for the primary tumor, and baseline sum of lesions TLG and MTV. RESULTS About 78 consecutive patients were included in this study. Median follow-up was 49 months. Baseline TLG, SUVmax, SULpeak, SULmax, sum of lesions for TLG and MTV, Delta SUVmax, and post-therapeutic SULpeak and SUVmax for the primary tumor, were statistically significant for disease free survival. CONCLUSION Pretherapeutic 18F-FDG PET/CT has a statistically significant prognostic value. The wide variability of results published in literature compels us to specifically explore the interest of uptake variations between pre- and post-treatment examinations.
Collapse
Affiliation(s)
- C Zwarthoed
- Department of Nuclear Medicine, Centre Antoine Lacassagne, Nice, France
| | - C Jaraudias
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France.
| | - L Evesque
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - D Baron
- Department of Radiation Oncology, Centre Antoine Lacassagne, Nice, France
| | - E François
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - D Chardin
- Department of Nuclear Medicine, Centre Antoine Lacassagne, Nice, France
| | - L Marie
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - D Mitrea
- Department of Radiation Oncology, Centre Antoine Lacassagne, Nice, France
| | - Y Château
- Department of Medical Statistic, Centre Antoine Lacassagne, Nice, France
| | - J Gal
- Department of Medical Statistic, Centre Antoine Lacassagne, Nice, France
| | - C Bailleux
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
16
|
Van den Bossche V, Vignau J, Vigneron E, Rizzi I, Zaryouh H, Wouters A, Ambroise J, Van Laere S, Beyaert S, Helaers R, van Marcke C, Mignion L, Lepicard EY, Jordan BF, Guilbaud C, Lowyck O, Dahou H, Mendola A, Desgres M, Aubert L, Gerin I, Bommer GT, Boidot R, Vermonden P, Warnant A, Larondelle Y, Machiels JP, Feron O, Schmitz S, Corbet C. PPARα-mediated lipid metabolism reprogramming supports anti-EGFR therapy resistance in head and neck squamous cell carcinoma. Nat Commun 2025; 16:1237. [PMID: 39890801 PMCID: PMC11785796 DOI: 10.1038/s41467-025-56675-3] [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: 07/14/2023] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
Anti-epidermal growth factor receptor (EGFR) therapy (cetuximab) shows a limited clinical benefit for patients with locally advanced or recurrent/metastatic head and neck squamous cell carcinoma (HNSCC), due to the frequent occurrence of secondary resistance mechanisms. Here we report that cetuximab-resistant HNSCC cells display a peroxisome proliferator-activated receptor alpha (PPARα)-mediated lipid metabolism reprogramming, with increased fatty acid uptake and oxidation capacities, while glycolysis is not modified. This metabolic shift makes cetuximab-resistant HNSCC cells particularly sensitive to a pharmacological inhibition of either carnitine palmitoyltransferase 1A (CPT1A) or PPARα in 3D spheroids and tumor xenografts in mice. Importantly, the PPARα-related gene signature, in human clinical datasets, correlates with lower response to anti-EGFR therapy and poor survival in HNSCC patients, thereby validating its clinical relevance. This study points out lipid metabolism rewiring as a non-genetic resistance-causing mechanism in HNSCC that may be therapeutically targeted to overcome acquired resistance to anti-EGFR therapy.
Collapse
Affiliation(s)
- Valentin Van den Bossche
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
| | - Julie Vignau
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Engy Vigneron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Isabella Rizzi
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Hannah Zaryouh
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Jérôme Ambroise
- Centre des Technologies Moléculaires Appliquées (CTMA), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 54, B-1200, Brussels, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit (TCRU), GZA Ziekenhuizen, Antwerp, Belgium
| | - Simon Beyaert
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
- Department of Head and Neck Surgery, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
| | - Raphaël Helaers
- Laboratory of Human Molecular Genetics, de Duve Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Cédric van Marcke
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
| | - Lionel Mignion
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Elise Y Lepicard
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Céline Guilbaud
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Olivier Lowyck
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
| | - Hajar Dahou
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
| | - Antonella Mendola
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
| | - Manon Desgres
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Léo Aubert
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
| | - Isabelle Gerin
- Metabolic Research Group, de Duve Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Guido T Bommer
- Metabolic Research Group, de Duve Institute, UCLouvain, B-1200, Brussels, Belgium
| | - Romain Boidot
- Unit of Molecular Biology, Department of Biology and Pathology of Tumors, Georges‑François Leclerc Cancer Center‑UNICANCER, 21079, Dijon, France
- ICMUB UMR CNRS 6302, 21079, Dijon, France
| | - Perrine Vermonden
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Croix du Sud 4-5/L7.07.03, B-1348, Louvain-la-Neuve, Belgium
| | - Aurélien Warnant
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Croix du Sud 4-5/L7.07.03, B-1348, Louvain-la-Neuve, Belgium
| | - Yvan Larondelle
- Louvain Institute of Biomolecular Science and Technology (LIBST), UCLouvain, Croix du Sud 4-5/L7.07.03, B-1348, Louvain-la-Neuve, Belgium
| | - Jean-Pascal Machiels
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
- Department of Head and Neck Surgery, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium
- WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium
| | - Sandra Schmitz
- King Albert II Cancer Institute, Department of Medical Oncology, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
- Pole of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B-1200, Brussels, Belgium
- Department of Head and Neck Surgery, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10, B-1200, Brussels, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Avenue Hippocrate 57, B1.57.04, B-1200, Brussels, Belgium.
- WEL Research Institute, Avenue Pasteur 6, B-1300, Wavre, Belgium.
| |
Collapse
|
17
|
Lee G, Moon SH, Kim JH, Jeong DY, Choi J, Choi JY, Lee HY. Multimodal Imaging Approach for Tumor Treatment Response Evaluation in the Era of Immunotherapy. Invest Radiol 2025; 60:11-26. [PMID: 39018248 DOI: 10.1097/rli.0000000000001096] [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: 07/19/2024]
Abstract
ABSTRACT Immunotherapy is likely the most remarkable advancement in lung cancer treatment during the past decade. Although immunotherapy provides substantial benefits, their therapeutic responses differ from those of conventional chemotherapy and targeted therapy, and some patients present unique immunotherapy response patterns that cannot be judged under the current measurement standards. Therefore, the response monitoring of immunotherapy can be challenging, such as the differentiation between real response and pseudo-response. This review outlines the various tumor response patterns to immunotherapy and discusses methods for quantifying computed tomography (CT) and 18 F-fluorodeoxyglucose positron emission tomography (PET) in the field of lung cancer. Emerging technologies in magnetic resonance imaging (MRI) and non-FDG PET tracers are also explored. With immunotherapy responses, the role for imaging is essential in both anatomical radiological responses (CT/MRI) and molecular changes (PET imaging). Multiple aspects must be considered when assessing treatment responses using CT and PET. Finally, we introduce multimodal approaches that integrate imaging and nonimaging data, and we discuss future directions for the assessment and prediction of lung cancer responses to immunotherapy.
Collapse
Affiliation(s)
- Geewon Lee
- From the Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (G.L., D.Y.J., J.C., H.Y.L.); Department of Radiology and Medical Research Institute, Pusan National University Hospital, Pusan National University School of Medicine, Busan, South Korea (G.L.); Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (S.H.M., J.Y.C.); Industrial Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea (J.H.K.); Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, South Korea (J.C.); and Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea (H.Y.L.)
| | | | | | | | | | | | | |
Collapse
|
18
|
Giunta EF, Caroli P, Scarpi E, Altavilla A, Rossetti V, Marini I, Celli M, Casadei C, Lolli C, Schepisi G, Bleve S, Brighi N, Cursano MC, Paganelli G, Matteucci F, De Giorgi U. Correlation of [ 68Ga]Ga-PSMA PET/CT response and PSA decline in first-line enzalutamide for metastatic castration-resistant prostate cancer patients. Eur J Nucl Med Mol Imaging 2024; 52:326-334. [PMID: 39207484 PMCID: PMC11599341 DOI: 10.1007/s00259-024-06887-4] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE to assess the utility of response monitoring to enzalutamide by using [68Ga]Ga-PSMA PET in mCRPC patients treated with enzalutamide as first-line therapy. METHODS patients underwent [68Ga]Ga-PSMA PET less than 8 weeks before and 3 months after starting enzalutamide. On the basis of EAU/EANM criteria, patients were categorized as PSMA responders (PET-R) or PSMA non-responders (PET-NR), whilst, based on PSA, they were classified as biochemical responders (PSA-R) or non-responders (PSA-NR). Survival analysis was performed using the Cox regression hazard model and the Kaplan-Meier method. RESULTS 69 patients were considered fully evaluable. We observed 47.8% of concordance between [68Ga]Ga-PSMA PET and PSA monitoring at 3 months after starting enzalutamide. For discordant cases, the PSA reduction has a weak impact on PFS and a significant impact on OS in PET-NR patients, whilst this change has no impact either for PFS and OS in PET-R ones. CONCLUSIONS [68Ga]Ga-PSMA PET could be a useful imaging tool for monitoring response to enzalutamide in mCRPC patients, being more informative than PSA in this setting, and possibly better guiding clinicians in therapeutic decisions.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Paola Caroli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Amelia Altavilla
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Virginia Rossetti
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Marini
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Monica Celli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicole Brighi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Concetta Cursano
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Paganelli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Matteucci
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
19
|
Vos JL, Traets JJ, Qiao X, Seignette IM, Peters D, Wouters MW, Hooijberg E, Broeks A, van der Wal JE, Karakullukcu MB, Klop WMC, Navran A, van Beurden M, Brouwer OR, Morris LG, van Poelgeest MI, Kapiteijn E, Haanen JB, Blank CU, Zuur CL. Diversity of the immune microenvironment and response to checkpoint inhibitor immunotherapy in mucosal melanoma. JCI Insight 2024; 9:e179982. [PMID: 39513365 PMCID: PMC11601749 DOI: 10.1172/jci.insight.179982] [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: 02/02/2024] [Accepted: 08/21/2024] [Indexed: 11/15/2024] Open
Abstract
Mucosal melanoma (MucM) is a rare cancer with a poor prognosis and low response rate to immune checkpoint inhibition (ICI) compared with cutaneous melanoma (CM). To explore the immune microenvironment and potential drivers of MucM's relative resistance to ICI drugs, we characterized 101 MucM tumors (43 head and neck [H&N], 31 female urogenital, 13 male urogenital, 11 anorectal, and 3 other gastrointestinal) using bulk RNA-Seq and immunofluorescence. RNA-Seq data show that MucM has a significantly lower IFN-γ signature levels than CM. MucM tumors of the H&N region show a significantly greater abundance of CD8+ T cells, cytotoxic cells, and higher IFN-γ signature levels than MucM from lower body sites. In the subcohort of 35 patients with MucM treated with ICI, hierarchical clustering reveals clusters with a high and low degree of immune infiltration, with a differential ICI response rate. Immune-associated gene sets were enriched in responders. Signatures associated with cancer-associated fibroblasts, macrophages, and TGF-β signaling may be higher in immune-infiltrated, but ICI-unresponsive tumors, suggesting a role for these resistance mechanisms in MucM. Our data show organ region-specific differences in immune infiltration and IFN-γ signature levels in MucM, with H&N MucM displaying the most favorable immune profile. Our study might offer a starting point for developing more personalized treatment strategies for this disease.
Collapse
Affiliation(s)
- Joris L. Vos
- Department of Head and Neck Surgery and Oncology and
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joleen J.H. Traets
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Oncology and Immunology
| | - Xiaohang Qiao
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, and
| | | | | | | | | | - M. Baris Karakullukcu
- Department of Head and Neck Surgery and Oncology and
- Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Center – Location Amsterdam Medical Center, Amsterdam, Netherlands
| | - W. Martin C. Klop
- Department of Head and Neck Surgery and Oncology and
- Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Center – Location Amsterdam Medical Center, Amsterdam, Netherlands
| | | | | | - Oscar R. Brouwer
- Department of Urology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Luc G.T. Morris
- Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - John B.A.G. Haanen
- Division of Molecular Oncology and Immunology
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Christian U. Blank
- Division of Molecular Oncology and Immunology
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Charlotte L. Zuur
- Department of Head and Neck Surgery and Oncology and
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
- Department of Oral and Maxillofacial Surgery, Amsterdam University Medical Center – Location Amsterdam Medical Center, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
20
|
Topkan E, Kucuk A, Ozturk D, Ozkan EE, Kılıç Durankuş N, Şenyürek Ş, Selek U, Pehlivan B. High Systemic Immune-Inflammation Index Values Before Treatment Predict Poor Pancreatic Cancer Outcomes After Definitive Chemoradiotherapy. Clin Med Insights Oncol 2024; 18:11795549241298552. [PMID: 39525980 PMCID: PMC11544671 DOI: 10.1177/11795549241298552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Background The systemic immune-inflammation index (SII) is an effective tool for predicting the prognosis of patients with cancer. However, its value in patients with locally advanced pancreatic ductal adenocarcinoma (LA-PDAC) undergoing definitive chemoradiotherapy has yet to be addressed. Therefore, we aimed to retrospectively investigate the prognostic significance of the pretreatment SII on the survival outcomes of patients with unresectable LA-PDAC treated with concurrent chemoradiotherapy (C-CRT). Methods The study included 163 patients with LA-PDAC who had received C-CRT. Using receiver operating characteristic (ROC) curve analysis, the utility of a pre-C-CRT cutoff that could stratify survival results was investigated. The primary and secondary endpoints were the correlations between SII levels and overall survival (OS) and progression-free survival (PFS). Results At a median follow-up period of 15 months (range: 3.2-94.5), the median OS and PFS rates for the entire group were 15.7 months (95% confidence interval [CI]: 13.4-17.9), and 7.8 months (95% CI: 6.1-9.4), respectively. We divided the patients into 2 SII cohorts based on the ROC curve analysis (area under the curve [AUC]: 71.9%; sensitivity: 68.9%; specificity: 66.7%): SII < 538 (N = 70) and SII ⩾ 538 (N = 93). Comparative survival analysis showed significantly inferior median OS (13.0 vs 25.4 months; P < .001) and PFS (7.0 vs 15.2 months; P = .003) in patients with SII ⩾ 538 compared with those with SII < 538 before treatment. In multivariate analyses, the Eastern Cooperative Oncology Group (ECOG) performance of 2, N1-2 lymph node, CA 19-9 > 90 U/mL, and SII ⩾ 538 status emerged as independent prognosticators of inferior OS and PFS. Conclusions Present results indicate that patients with unresectable LA-PDAC who underwent C-CRT and had a pretreatment SII ⩾ 538 had significantly worse OS and PFS outcomes compared with those with lower SII values.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana, Turkey
| | - Ahmet Kucuk
- Clinic of Radiation Oncology, Mersin Education and Research Hospital, Mersin, Turkey
| | - Duriye Ozturk
- Department of Radiation Oncology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Emine Elif Ozkan
- Department of Radiation Oncology, Suleyman Demirel University, Isparta, Turkey
| | | | - Şükran Şenyürek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Ugur Selek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Berrin Pehlivan
- Department of Radiation Oncology, Bahcesehir University, Istanbul, Turkey
| |
Collapse
|
21
|
Xu H, Zhou Y, Liang L, Shen J, Yan W, Wang J, Li J, Zhang X, Huang G, Bi W, Guo Z, Xiao Y, Lin J, Yao W, Tong Z, Zhou W, Zhang G, Ye Z, Wang D, Yang J, Fan Z, Liu C, Qu G, Zhang Q, Wei F, Liu W, Tu C, Li H, Yuan J, Niu X. Efficacy and safety of JMT103 in patients with unresectable or surgically-challenging giant cell tumor of bone: a multicenter, phase Ib/II study. Nat Commun 2024; 15:9541. [PMID: 39500883 PMCID: PMC11538294 DOI: 10.1038/s41467-024-53686-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
This was a multicenter, single-arm, open-label, phase Ib/II study (NCT04255576), aimed to evaluate the efficacy and safety of JMT103 in patients with unresectable or surgically-challenging giant cell tumor of bone (GCTB). JMT103 (2 mg/kg) was administered subcutaneously every four weeks, with loading doses on days 8 and 15. The primary endpoint was the objective tumor response rate (OTR) based on best response, defined as the proportion of patients who achieved elimination of at least 90% of the giant cells or radiologic complete or partial response per the modified Inverse Choi density/size (mICDS) or modified European Organization for Research and Treatment of Cancer (mEORTC) within 12 weeks. Secondary endpoints included objective response rate (ORR) per mICDS and mEORTC, and safety. A total of 139 patients were enrolled, and 135 were analyzed for efficacy. OTR, determined by the independent review committee (IRC) was 93.3% (95% CI 87.7-96.9). Treatment-related adverse events occurred in 90 (64.7%) patients, with hypophosphatemia and hypocalcemia being the most common. No serious treatment-related adverse events were observed. Thus, JMT103 demonstrates potential as a therapeutic option for GCTB.
Collapse
Affiliation(s)
- Hairong Xu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yong Zhou
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Li Liang
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jin Wang
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Jianmin Li
- Department of Orthopedic Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaojing Zhang
- Department of Bone and Soft Tissue Surgery, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Gang Huang
- Department of Bone and Soft Tissue Surgery, Hunan Cancer Hospital, Changsha, China
| | - Wenzhi Bi
- Senior Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zheng Guo
- Department of Orthopedics, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yanbin Xiao
- The Second Department of Orthopedic Surgery, Yunnan Cancer Hospital, Kunming, China
| | - Jianhua Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Weitao Yao
- Department of Bone and Soft Tissue, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Zhichao Tong
- Department of Bone Tumor, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Wenxian Zhou
- Department of Breast, Bone and Soft Tissue Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guochuan Zhang
- Department of Bone and Soft Tissue Tumor, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhaoming Ye
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dong Wang
- Department of Orthopedics, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Zhengfu Fan
- Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital, Beijing, China
| | - Caigang Liu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guofan Qu
- Department of Orthopedic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qing Zhang
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Feng Wei
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, China
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Weifeng Liu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Chongqi Tu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- CSPC Pharmaceutical Group Limited, Shijiazhuang, China
| | - Jing Yuan
- CSPC Pharmaceutical Group Limited, Shijiazhuang, China
| | - Xiaohui Niu
- Department of Orthopedic Oncology Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Vöö S, Baldeweg SE, Kumar R, Singh H, Mittal BR, Bomanji J. Perennial inconsistencies and lack of evidence-based recommendations in current guidelines addressing blood glucose level regulations for optimal 18F-fluorodeoxyglucose PET imaging: 25-year 'silver jubilee' of an ongoing unsolved problem in nuclear medicine. Nucl Med Commun 2024; 45:897-900. [PMID: 39466710 DOI: 10.1097/mnm.0000000000001894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Affiliation(s)
- Stefan Vöö
- Institute of Nuclear Medicine, University College London Hospital, University College London Hospitals NHS Foundation Trust (UCLH),
| | - Stephanie E Baldeweg
- Department of Endocrinology, UCLH,
- Division of Medicine, University College London, London, UK and
| | - Rajender Kumar
- Department of Nuclear Medicine and PET, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harmandeep Singh
- Department of Nuclear Medicine and PET, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant R Mittal
- Department of Nuclear Medicine and PET, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, University College London Hospital, University College London Hospitals NHS Foundation Trust (UCLH),
| |
Collapse
|
23
|
Di Franco M, Mei R, Garcia C, Fanti S. Treatment response assessment in mCRPC: is PSMA-PET/CT going to take the lead? Ther Adv Med Oncol 2024; 16:17588359241258367. [PMID: 39386313 PMCID: PMC11462558 DOI: 10.1177/17588359241258367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/14/2024] [Indexed: 10/12/2024] Open
Abstract
The assessment of response to therapy in prostate cancer (PCa) patients is an ongoing, open issue. Prostate-specific antigen has limitations, especially in advanced metastatic PCa, which often displays intratumor variability in terms of response to therapy. Conventional imaging (i.e. computerized tomography and bone scan) is of limited use for its low sensitivity and specificity. Positron-emission tomography (PET) with prostate-specific membrane antigen (PSMA) demonstrated higher sensitivity and specificity, and novel PSMA-based criteria have been recently proposed for treatment response, with promising results in different scenarios, from chemotherapy to radioligand therapy. PSMA-based criteria have been found to outperform the current RECIST 1.1 and Prostate Cancer Working Group 3 frameworks in describing the behavior of PCa, precisely assessing tumor phenotypes through molecular-imaging-derived parameters. This review critically explores the current evidence about the role of PSMA PET/computed tomography in the assessment of treatment response.
Collapse
Affiliation(s)
- Martina Di Franco
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Via Massarenti 9, Bologna 40138, Italy
| | - Riccardo Mei
- Nuclear Medicine Unit, University Hospital of Modena, Modena, Italy
| | - Camilo Garcia
- Department of Nuclear Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Stefano Fanti
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
24
|
Brezun J, Aide N, Peroux E, Lamboley JL, Gutman F, Lussato D, Helissey C. [18F]FDG PET/CT Integration in Evaluating Immunotherapy for Lung Cancer: A Clinician's Practical Approach. Diagnostics (Basel) 2024; 14:2104. [PMID: 39335783 PMCID: PMC11431382 DOI: 10.3390/diagnostics14182104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized the treatment paradigm of lung cancer, resulting in notable enhancements in patient survival. Nevertheless, evaluating treatment response in patients undergoing immunotherapy poses distinct challenges due to unconventional response patterns like pseudoprogressive disease (PPD), dissociated response (DR), and hyperprogressive disease (HPD). Conventional response criteria such as the RECIST 1.1 may not adequately address these complexities. To tackle this issue, novel response criteria such as the iRECIST and imRECIST have been proposed, enabling a more comprehensive assessment of treatment response by incorporating additional scans and considering the best overall response even after radiologic progressive disease evaluation. Additionally, [18F]FDG PET/CT imaging has emerged as a valuable modality for evaluating treatment response, with various metabolic response criteria such as the PERCIMT, imPERCIST, and iPERCIST developed to overcome the limitations of traditional criteria, particularly in detecting pseudoprogression. A multidisciplinary approach involving oncologists, radiologists, and nuclear medicine specialists is crucial for effectively navigating these complexities and enhancing patient outcomes in the era of immunotherapy for lung cancer. In this review, we delineate the key components of these guidelines, summarizing essential aspects for radiologists and nuclear medicine physicians. Furthermore, we provide insights into how imaging can guide the management of individual lung cancer patients in real-world multidisciplinary settings.
Collapse
Affiliation(s)
- Juliette Brezun
- Department of Medical Oncology and Clinical Research Unit, Military Hospital Bégin, 94160 Saint-Mandé, France
| | - Nicolas Aide
- INSERM ANTICIPE U1086, Caen University, 14000 Caen, France;
| | - Evelyne Peroux
- Department of Radiology, Military Hospital Laveran, 13013 Marseille, France;
| | | | - Fabrice Gutman
- Department of Nuclear Medicine, Paul d’Egine Hospital, 94500 Champigny-sur-Marne, France;
| | - David Lussato
- Department of Nuclear Medicine, Centre Cardiologique du Nord, 93200 Saint-Denis, France;
| | - Carole Helissey
- Department of Medical Oncology and Clinical Research Unit, Military Hospital Bégin, 94160 Saint-Mandé, France
| |
Collapse
|
25
|
Groheux D, Vaz SC, Poortmans P, Mann RM, Ulaner GA, Cook GJR, Hindié E, Pilkington Woll JP, Jacene H, Rubio IT, Vrancken Peeters MJ, Dibble EH, de Geus-Oei LF, Graff SL, Cardoso F. Role of [ 18F]FDG PET/CT in patients with invasive breast carcinoma of no special type: Literature review and comparison between guidelines. Breast 2024; 78:103806. [PMID: 39303572 PMCID: PMC11440802 DOI: 10.1016/j.breast.2024.103806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/29/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE The recently released EANM/SNMMI guideline, endorsed by several important clinical and imaging societies in the field of breast cancer (BC) care (ACR, ESSO, ESTRO, EUSOBI/ESR, EUSOMA), emphasized the role of [18F]FDG PET/CT in management of patients with no special type (NST) BC. This review identifies and summarizes similarities, discrepancies and novelties of the EANM/SNMMI guideline compared to NCCN, ESMO and ABC recommendations. METHODS The EANM/SNMMI guideline was based on a systematic literature search and the AGREE tool. The level of evidence was determined according to NICE criteria, and 85 % agreement or higher was reached regarding each statement. Comparisons with NCCN, ESMO and ABC guidelines were examined for specific clinical scenarios in patients with early stage through advanced and metastatic BC. RESULTS Regarding initial staging of patients with NST BC, [18F]FDG PET/CT is the preferred modality in the EANM-SNMMI guideline, showing superiority as a single modality to a combination of contrast-enhanced CT of thorax-abdomen-pelvis plus bone scan in head-to-head comparisons and a randomized study. Its use is recommended in patients with clinical stage IIB or higher and may be useful in certain stage IIA cases of NST BC. In NCCN, ESMO, and ABC guidelines, [18F]FDG PET/CT is instead recommended as complementary to conventional imaging to solve inconclusive findings, although ESMO and ABC also suggest [18F]FDG PET/CT can replace conventional imaging for staging patients with high-risk and metastatic NST BC. During follow up, NCCN and ESMO only recommend diagnostic imaging if there is suspicion of recurrence. Similarly, EANM-SNMMI states that [18F]FDG PET/CT is useful to detect the site and extent of recurrence only when there is clinical or laboratory suspicion of recurrence, or when conventional imaging methods are equivocal. The EANM-SNMMI guideline is the first to emphasize a role of [18F]FDG PET/CT for assessing early metabolic response to primary systemic therapy, particularly for HER2+ BC and TNBC. In the metastatic setting, EANM-SNMMI state that [18F]FDG PET/CT may help evaluate bone metastases and determine early response to treatment, in agreement with guidelines from ESMO. CONCLUSIONS The recently released EANM/SNMMI guideline reinforces the role of [18F]FDG PET/CT in the management of patients with NST BC supported by extensive evidence of its utility in several clinical scenarios.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, Paris, France; University Paris-Diderot, INSERM, U976, Paris, France; Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France.
| | - Sofia C Vaz
- Department of Nuclear Medicine and Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal; Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Belgium; Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk-Antwerp, Belgium
| | - Ritse M Mann
- Department of Radiology, Radboud umc, Nijmegen, the Netherlands
| | - Gary A Ulaner
- Department of Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, United States; Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, CA, United States
| | - Gary J R Cook
- Department of Cancer Imaging, King's College London, London, UK; King's College London and Guy's & St Thomas' PET Centre, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Elif Hindié
- Department of Nuclear Medicine, Bordeaux University Hospital, Bordeaux, France
| | | | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, and Harvard Medical School, United States
| | - Isabel T Rubio
- Department of Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Cancer Center Clinica Universidad de Navarra, Spain
| | - Marie-Jeanne Vrancken Peeters
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Surgery, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft, the Netherlands
| | - Stephanie L Graff
- Lifespan Cancer Institute, Providence, RI, United States; Legorreta Cancer Center at Brown University, Providence, RI, United States
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
26
|
Pu Y, Penney BC, Zhang J, Little K, Simon CA, Feinberg N, Zhang MH, Hwang G, Appelbaum DE. Comparison of Measurement and Prognostic Power of SUV Between High-Definition and Standard PET Imaging in Non-Small Cell Lung Cancer Patients. J Nucl Med Technol 2024; 52:229-233. [PMID: 39019575 DOI: 10.2967/jnmt.124.267684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/29/2024] [Indexed: 07/19/2024] Open
Abstract
This study aimed to evaluate the measurement and prognostic ability of the SUVmax of whole-body tumors (SUVmaxwb) in non-small cell lung cancer (NSCLC) patients, comparing high-definition (HD) PET imaging with standard-definition (SD) PET imaging. Methods: The study included 242 consecutive NSCLC patients who underwent baseline 18F-FDG PET/CT from April 2018 to January 2021. Two imaging techniques were used: HD PET (using ordered-subsets expectation maximization with point-spread function modeling and time-of-flight techniques and smaller voxels) and SD PET (with ordered-subsets expectation maximization and time-of-flight techniques). SUVmaxwb was determined by measuring all the tumor lesions in the whole body, and tumor-to-background ratio (TBR) was calculated using the background SUVmean of various body parts. Results: The patient cohort had an average age of 68.3 y, with 59.1% being female. During a median follow-up of 29.6 mo, 83 deaths occurred. SUVmaxwb was significantly higher in HD PET than SD PET, with respective medians of 17.4 and 11.8. The TBR of 1,125 tumoral lesions was also higher in HD PET. Univariate Cox regression analysis showed that SUVmaxwb from both HD and SD PET were significantly associated with overall survival. However, after adjusting for TNM (tumor, node, metastasis) stage, only SUVmaxwb from SD PET remained significantly associated with survival. Conclusion: HD PET imaging in NSCLC patients yields higher SUVmaxwb and TBR, enhancing tumor visibility. Despite this, its prognostic value is less significant than SD PET after adjusting clinical TNM stage. Thus, consideration should be given to using HD PET reconstruction to increase tumor visibility, and SD PET is recommended for NSCLC patient prognostication and therapeutic evaluation, as well as for the classification of lung nodules.
Collapse
Affiliation(s)
- Yonglin Pu
- Department of Radiology, University of Chicago, Chicago, Illinois;
| | - Bill C Penney
- Department of Radiology, University of Chicago, Chicago, Illinois
| | - Jingmian Zhang
- Fourth Hospital of Hebei Medical University, Shijiazhuang, China; and
| | - Kevin Little
- Department of Radiology, University of Chicago, Chicago, Illinois
| | - Cassie A Simon
- Cancer Registry, University of Chicago, Chicago, Illinois
| | | | | | - Gloria Hwang
- Department of Radiology, University of Chicago, Chicago, Illinois
| | | |
Collapse
|
27
|
Zhang J, Xiong J, Wang M, Wu B, Zhang C. Comparison of the diagnostic value of 68Ga-FAPI and 18F-FDG PET/CT in breast cancer: a systematic review. Clin Transl Imaging 2024; 12:787-798. [DOI: 10.1007/s40336-024-00656-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/14/2024] [Indexed: 01/03/2025]
|
28
|
Jemaa S, Ounadjela S, Wang X, El-Galaly TC, Kostakoglu L, Knapp A, Ku G, Musick L, Sahin D, Wei MC, Yin S, Bengtsson T, De Crespigny A, Carano RA. Automated Lugano Metabolic Response Assessment in 18F-Fluorodeoxyglucose-Avid Non-Hodgkin Lymphoma With Deep Learning on 18F-Fluorodeoxyglucose-Positron Emission Tomography. J Clin Oncol 2024; 42:2966-2977. [PMID: 38843483 PMCID: PMC11361360 DOI: 10.1200/jco.23.01978] [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: 09/14/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 08/30/2024] Open
Abstract
PURPOSE Artificial intelligence can reduce the time used by physicians on radiological assessments. For 18F-fluorodeoxyglucose-avid lymphomas, obtaining complete metabolic response (CMR) by end of treatment is prognostic. METHODS Here, we present a deep learning-based algorithm for fully automated treatment response assessments according to the Lugano 2014 classification. The proposed four-stage method, trained on a multicountry clinical trial (ClinicalTrials.gov identifier: NCT01287741) and tested in three independent multicenter and multicountry test sets on different non-Hodgkin lymphoma subtypes and different lines of treatment (ClinicalTrials.gov identifiers NCT02257567, NCT02500407; 20% holdout in ClinicalTrials.gov identifier NCT01287741), outputs the detected lesions at baseline and follow-up to enable focused radiologist review. RESULTS The method's response assessment achieved high agreement with the adjudicated radiologic responses (eg, agreement for overall response assessment of 93%, 87%, and 85% in ClinicalTrials.gov identifiers NCT01287741, NCT02500407, and NCT02257567, respectively) similar to inter-radiologist agreement and was strongly prognostic of outcomes with a trend toward higher accuracy for death risk than adjudicated radiologic responses (hazard ratio for end of treatment by-model CMR of 0.123, 0.054, and 0.205 in ClinicalTrials.gov identifiers NCT01287741, NCT02500407, and NCT02257567, compared with, respectively, 0.226, 0.292, and 0.272 for CMR by the adjudicated responses). Furthermore, a radiologist review of the algorithm's assessments was conducted. The radiologist median review time was 1.38 minutes/assessment, and no statistically significant differences were observed in the level of agreement of the radiologist with the model's response compared with the level of agreement of the radiologist with the adjudicated responses. CONCLUSION These results suggest that the proposed method can be incorporated into radiologic response assessment workflows in cancer imaging for significant time savings and with performance similar to trained medical experts.
Collapse
Affiliation(s)
| | | | | | - Tarec C. El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
- Hematology Research Unit, Department of Hematology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Medicine Solna, Clinical Epidemiology Division, Karolinska Institutet, Stockholm, Sweden
| | - Lale Kostakoglu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA
| | | | - Grace Ku
- Genentech, Inc, South San Francisco, CA
| | | | | | | | - Shen Yin
- Genentech, Inc, South San Francisco, CA
| | - Thomas Bengtsson
- Department of Statistics, University of California, Berkeley, CA
| | | | | |
Collapse
|
29
|
Long GV, Carlino MS, Au-Yeung G, Spillane AJ, Shannon KF, Gyorki DE, Hsiao E, Kapoor R, Thompson JR, Batula I, Howle J, Ch'ng S, Gonzalez M, Saw RPM, Pennington TE, Lo SN, Scolyer RA, Menzies AM. Neoadjuvant pembrolizumab, dabrafenib and trametinib in BRAF V600-mutant resectable melanoma: the randomized phase 2 NeoTrio trial. Nat Med 2024; 30:2540-2548. [PMID: 38907159 PMCID: PMC11405264 DOI: 10.1038/s41591-024-03077-5] [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: 03/04/2024] [Accepted: 05/17/2024] [Indexed: 06/23/2024]
Abstract
Immune checkpoint inhibitors and BRAF-targeted therapy each improve survival in melanoma. Immune changes early during targeted therapy suggest the mechanisms of each drug class could work synergistically. In the non-comparative, randomized, phase 2 NeoTrio trial, we investigated whether targeted therapy could boost the proportion of patients achieving long-term recurrence-free survival with neoadjuvant immunotherapy in resectable stage III BRAFV600-mutant melanoma. Sixty patients (42% females) were randomized to pembrolizumab alone (n = 20), sequential therapy (dabrafenib plus trametinib followed by pembrolizumab; n = 20) or concurrent (triple) therapy (n = 20), followed by surgery and adjuvant therapy. The primary outcome was pathological response; secondary outcomes included radiographic response, recurrence-free survival, overall survival, surgical outcomes, peripheral blood and tumor analyses and safety. The pathological response rate was 55% (11/20; including six pathological complete responses (pCRs)) with pembrolizumab, 50% (10/20; three pCRs) with sequential therapy and 80% (16/20; ten pCRs) with concurrent therapy, which met the primary outcome in each arm. Treatment-related adverse events affected 75-100% of patients during neoadjuvant treatment, with seven early discontinuations (all in the concurrent arm). At 2 years, event-free survival was 60% with pembrolizumab, 80% with sequential therapy and 71% with concurrent therapy. Recurrences after major pathological response were more common in the targeted therapy arms, suggesting a reduction in response 'quality' when targeted therapy is added to neoadjuvant immunotherapy. Risking the curative potential of immunotherapy in melanoma cannot be justified. Pending longer follow-up, we suggest that immunotherapy and targeted therapy should not be combined in the neoadjuvant setting for melanoma. ClinicalTrials.gov registration: NCT02858921 .
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Royal North Shore Hospital, Sydney, New South Wales, Australia.
- Mater Hospital, Sydney, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Westmead Hospital, Westmead, New South Wales, Australia
- Blacktown Hospital, Blacktown, New South Wales, Australia
| | - George Au-Yeung
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- Concord Repatriation Hospital, Concord, New South Wales, Australia
| | - David E Gyorki
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Edward Hsiao
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Rony Kapoor
- Mater Hospital, Sydney, New South Wales, Australia
- I-MED Radiology Network, Mater Hospital, Sydney, New South Wales, Australia
| | - Jake R Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Iris Batula
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Julie Howle
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Maria Gonzalez
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Thomas E Pennington
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- NSW Health Pathology, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
30
|
Weyts K, Lequesne J, Johnson A, Curcio H, Parzy A, Coquan E, Lasnon C. The impact of introducing deep learning based [ 18F]FDG PET denoising on EORTC and PERCIST therapeutic response assessments in digital PET/CT. EJNMMI Res 2024; 14:72. [PMID: 39126532 DOI: 10.1186/s13550-024-01128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/06/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND [18F]FDG PET denoising by SubtlePET™ using deep learning artificial intelligence (AI) was previously found to induce slight modifications in lesion and reference organs' quantification and in lesion detection. As a next step, we aimed to evaluate its clinical impact on [18F]FDG PET solid tumour treatment response assessments, while comparing "standard PET" to "AI denoised half-duration PET" ("AI PET") during follow-up. RESULTS 110 patients referred for baseline and follow-up standard digital [18F]FDG PET/CT were prospectively included. "Standard" EORTC and, if applicable, PERCIST response classifications by 2 readers between baseline standard PET1 and follow-up standard PET2 as a "gold standard" were compared to "mixed" classifications between standard PET1 and AI PET2 (group 1; n = 64), or between AI PET1 and standard PET2 (group 2; n = 46). Separate classifications were established using either standardized uptake values from ultra-high definition PET with or without AI denoising (simplified to "UHD") or EANM research limited v2 (EARL2)-compliant values (by Gaussian filtering in standard PET and using the same filter in AI PET). Overall, pooling both study groups, in 11/110 (10%) patients at least one EORTCUHD or EARL2 or PERCISTUHD or EARL2 mixed vs. standard classification was discordant, with 369/397 (93%) concordant classifications, unweighted Cohen's kappa = 0.86 (95% CI: 0.78-0.94). These modified mixed vs. standard classifications could have impacted management in 2% of patients. CONCLUSIONS Although comparing similar PET images is preferable for therapy response assessment, the comparison between a standard [18F]FDG PET and an AI denoised half-duration PET is feasible and seems clinically satisfactory.
Collapse
Affiliation(s)
- Kathleen Weyts
- Nuclear Medicine Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, 3 Avenue du General Harris, BP 45026, Caen Cedex 5, 14076, France.
| | - Justine Lequesne
- Biostatistics Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Alison Johnson
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Hubert Curcio
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Aurélie Parzy
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Elodie Coquan
- Medical Oncology Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, France
| | - Charline Lasnon
- Nuclear Medicine Department, François Baclesse Comprehensive Cancer Centre, UNICANCER, Caen, 3 Avenue du General Harris, BP 45026, Caen Cedex 5, 14076, France
- UNICAEN, INSERM 1086 ANTICIPE, Normandy University, Caen, France
| |
Collapse
|
31
|
Cruz-Montijano M, Amo-Salas M, Cassinello-Espinosa J, García-Carbonero I, Villa-Guzman JC, Garcia-Vicente AM. Predictive and Prognostic 18F-Fluorocholine PET/CT Radiomics Nomogram in Patients with Castration-Resistant Prostate Cancer with Bone Metastases Treated with 223Ra. Cancers (Basel) 2024; 16:2695. [PMID: 39123422 PMCID: PMC11312125 DOI: 10.3390/cancers16152695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
PURPOSE We aimed to develop a nomogram able to predict treatment failure, skeletal events, and overall survival (OS) in patients with castration-resistant prostate cancer with bone metastases (CRPC-BM) treated with Radium-223 dichloride (223Ra). PATIENTS AND METHODS Patients from the Castilla-La Mancha Spanish region were prospectively included in the ChoPET-Rad multicenter study from January 2015 to December 2022. Patients underwent baseline, interim, and end-of-treatment bone scintigraphy (BS) and 18F-Fluorocholine PET/CT (FCH PET/CT) scans, obtaining multiple imaging radiomics as well as clinical and biochemical variables during follow-up and studying their association with the previously defined end-points. Survival analysis was performed using the Kaplan-Meier method and Cox regression. Multivariate logistic and Cox regression models were calculated, and these models were depicted by means of nomograms. RESULTS Median progression-free survival (PFS) and OS were 4 and 14 months (mo), respectively. The variables that showed independent and significant association with therapeutic failure were baseline alkaline phosphatase (AP) levels (p = 0.022) and the characteristics of BM on the CT portion of PET/CT (p = 0.017). In the case of OS, the significant variables were therapeutic failure (p = 0.038), the number of lines received after 223Ra (p < 0.001), average SUVmax (p = 0.002), bone marrow infiltration in FCH PET/CT (p = 0.006), and interim FCH PET/CT response (p = 0.048). Final nomograms included these variables, showing good discrimination among the 100 patients included in our study. In the study of skeletal events, only OS showed a significant association in the multivariate analysis, resulting in an inconsistent nomogram design. CONCLUSIONS FCH PET/CT appears to be a good tool for evaluating patients eligible for treatment with 223Ra, as well as for their follow-up. Thus, findings derived from it, such as the morphological characteristics of BM in the CT, bone marrow infiltration, or the response to 223Ra in the interim study, have proven to be solid and useful variables in the creation of nomograms for predicting therapeutic failure and OS.
Collapse
Affiliation(s)
| | - Mariano Amo-Salas
- Mathematics Department, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain;
| | | | | | | | | |
Collapse
|
32
|
Vaz SC, Woll JPP, Cardoso F, Groheux D, Cook GJR, Ulaner GA, Jacene H, Rubio IT, Schoones JW, Peeters MJV, Poortmans P, Mann RM, Graff SL, Dibble EH, de Geus-Oei LF. Joint EANM-SNMMI guideline on the role of 2-[ 18F]FDG PET/CT in no special type breast cancer : (endorsed by the ACR, ESSO, ESTRO, EUSOBI/ESR, and EUSOMA). Eur J Nucl Med Mol Imaging 2024; 51:2706-2732. [PMID: 38740576 PMCID: PMC11224102 DOI: 10.1007/s00259-024-06696-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/20/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION There is much literature about the role of 2-[18F]FDG PET/CT in patients with breast cancer (BC). However, there exists no international guideline with involvement of the nuclear medicine societies about this subject. PURPOSE To provide an organized, international, state-of-the-art, and multidisciplinary guideline, led by experts of two nuclear medicine societies (EANM and SNMMI) and representation of important societies in the field of BC (ACR, ESSO, ESTRO, EUSOBI/ESR, and EUSOMA). METHODS Literature review and expert discussion were performed with the aim of collecting updated information regarding the role of 2-[18F]FDG PET/CT in patients with no special type (NST) BC and summarizing its indications according to scientific evidence. Recommendations were scored according to the National Institute for Health and Care Excellence (NICE) criteria. RESULTS Quantitative PET features (SUV, MTV, TLG) are valuable prognostic parameters. In baseline staging, 2-[18F]FDG PET/CT plays a role from stage IIB through stage IV. When assessing response to therapy, 2-[18F]FDG PET/CT should be performed on certified scanners, and reported either according to PERCIST, EORTC PET, or EANM immunotherapy response criteria, as appropriate. 2-[18F]FDG PET/CT may be useful to assess early metabolic response, particularly in non-metastatic triple-negative and HER2+ tumours. 2-[18F]FDG PET/CT is useful to detect the site and extent of recurrence when conventional imaging methods are equivocal and when there is clinical and/or laboratorial suspicion of relapse. Recent developments are promising. CONCLUSION 2-[18F]FDG PET/CT is extremely useful in BC management, as supported by extensive evidence of its utility compared to other imaging modalities in several clinical scenarios.
Collapse
Affiliation(s)
- Sofia C Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal.
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - David Groheux
- Nuclear Medicine Department, Saint-Louis Hospital, Paris, France
- University Paris-Diderot, INSERM U976, Paris, France
- Centre d'Imagerie Radio-Isotopique (CIRI), La Rochelle, France
| | - Gary J R Cook
- Department of Cancer Imaging, King's College London, London, UK
- King's College London and Guy's & St Thomas' PET Centre, London, UK
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Newport Beach, CA, USA
- University of Southern California, Los Angeles, CA, USA
| | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Isabel T Rubio
- Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, Cancer Center Clinica Universidad de Navarra, Navarra, Spain
| | - Jan W Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-Jeanne Vrancken Peeters
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Philip Poortmans
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, Belgium
- University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Ritse M Mann
- Radiology Department, RadboudUMC, Nijmegen, The Netherlands
| | - Stephanie L Graff
- Lifespan Cancer Institute, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Elizabeth H Dibble
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands.
- Department of Radiation Science & Technology, Technical University of Delft, Delft, The Netherlands.
| |
Collapse
|
33
|
Swiha M, Gafita A, Nguyen A, Emmett L. Treatment Response Imaging in Prostate Cancer. PET Clin 2024; 19:417-430. [PMID: 38670877 DOI: 10.1016/j.cpet.2024.03.009] [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: 04/28/2024]
Abstract
Objective criteria for measuring treatment response in prostate cancer are critical to clinical research and practice. The Prostate Cancer Working Group 3 criteria are widely accepted relying only on conventional imaging for radiographic treatment response. Prostate-specific membrane antigen PET/computed tomography was proven to be superior to conventional imaging in initial diagnosis and biochemical recurrence of prostate cancer. Moreover, there is growing evidence of its role in treatment response assessment in prostate cancer. This study will review the different criteria for imaging treatment response on conventional and advanced molecular imaging for different therapies, and the future perspective in posttherapy imaging.
Collapse
Affiliation(s)
- Mina Swiha
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; Nuclear Medicine Division, Department of Medical Imaging, University of Western Ontario, London, Canada.
| | - Andrei Gafita
- Nuclear Medicine and Molecular Imaging Division, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medicine, Baltimore, USA
| | - Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia; Garvan Institute of Medical Research, Sydney, Australia
| |
Collapse
|
34
|
Hirota S, Tateishi U, Nakamoto Y, Yamamoto H, Sakurai S, Kikuchi H, Kanda T, Kurokawa Y, Cho H, Nishida T, Sawaki A, Ozaka M, Komatsu Y, Naito Y, Honma Y, Takahashi F, Hashimoto H, Udo M, Araki M, Nishidate S. English version of Japanese Clinical Practice Guidelines 2022 for gastrointestinal stromal tumor (GIST) issued by the Japan Society of Clinical Oncology. Int J Clin Oncol 2024; 29:647-680. [PMID: 38609732 PMCID: PMC11130037 DOI: 10.1007/s10147-024-02488-1] [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: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 04/14/2024]
Abstract
The Japan Society of Clinical Oncology Clinical Practice Guidelines 2022 for gastrointestinal stromal tumor (GIST) have been published in accordance with the Minds Manual for Guideline Development 2014 and 2017. A specialized team independent of the working group for the revision performed a systematic review. Since GIST is a rare type of tumor, clinical evidence is not sufficient to answer several clinical and background questions. Thus, in these guidelines, we considered that consensus among the experts who manage GIST, the balance between benefits and harms, patients' wishes, medical economic perspective, etc. are important considerations in addition to the evidence. Although guidelines for the treatment of GIST have also been published by the National Comprehensive Cancer Network (NCCN) and the European Society for Medical Oncology (ESMO), there are some differences between the treatments proposed in those guidelines and the treatments in the present guidelines because of the differences in health insurance systems among countries.
Collapse
Affiliation(s)
- Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University School of Medicine, Nishinomiya, Japan.
| | - Ukihide Tateishi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetaka Yamamoto
- Department of Pathology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shinji Sakurai
- Department of Diagnostic Pathology, Japan Community Healthcare Organization Gunma Central Hospital, Maebashi, Japan
| | - Hirotoshi Kikuchi
- Department of Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tatsuo Kanda
- Department of Gastroenterology, Southern TOHOKU General Hospital, Koriyama, Japan
| | - Yukinori Kurokawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Haruhiko Cho
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Toshirou Nishida
- Department of Surgery, Japan Community Healthcare Organization Osaka Hospital, Osaka, Japan
| | - Akira Sawaki
- Department of Medical Oncology, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Fumiaki Takahashi
- Department of Information Science, Iwate Medical University, Morioka, Japan
| | | | - Midori Udo
- Nursing Department, Osaka Police Hospital, Osaka, Japan
| | - Minako Araki
- Association of Chubu GIST Patients and Their Families, Nagoya, Japan
| | | |
Collapse
|
35
|
Timmers HJLM, Taïeb D, Pacak K, Lenders JWM. Imaging of Pheochromocytomas and Paragangliomas. Endocr Rev 2024; 45:414-434. [PMID: 38206185 PMCID: PMC11074798 DOI: 10.1210/endrev/bnae001] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Pheochromocytomas/paragangliomas are unique in their highly variable molecular landscape driven by genetic alterations, either germline or somatic. These mutations translate into different clusters with distinct tumor locations, biochemical/metabolomic features, tumor cell characteristics (eg, receptors, transporters), and disease course. Such tumor heterogeneity calls for different imaging strategies in order to provide proper diagnosis and follow-up. This also warrants selection of the most appropriate and locally available imaging modalities tailored to an individual patient based on consideration of many relevant factors including age, (anticipated) tumor location(s), size, and multifocality, underlying genotype, biochemical phenotype, chance of metastases, as well as the patient's personal preference and treatment goals. Anatomical imaging using computed tomography and magnetic resonance imaging and functional imaging using positron emission tomography and single photon emission computed tomography are currently a cornerstone in the evaluation of patients with pheochromocytomas/paragangliomas. In modern nuclear medicine practice, a multitude of radionuclides with relevance to diagnostic work-up and treatment planning (theranostics) is available, including radiolabeled metaiodobenzylguanidine, fluorodeoxyglucose, fluorodihydroxyphenylalanine, and somatostatin analogues. This review amalgamates up-to-date imaging guidelines, expert opinions, and recent discoveries. Based on the rich toolbox for anatomical and functional imaging that is currently available, we aim to define a customized approach in patients with (suspected) pheochromocytomas/paragangliomas from a practical clinical perspective. We provide imaging algorithms for different starting points for initial diagnostic work-up and course of the disease, including adrenal incidentaloma, established biochemical diagnosis, postsurgical follow-up, tumor screening in pathogenic variant carriers, staging and restaging of metastatic disease, theranostics, and response monitoring.
Collapse
Affiliation(s)
- Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France and European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1583, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
36
|
Castagnoli F, Mencel J, Ap Dafydd D, Gough J, Drake B, Mcaddy NC, Withey SJ, Riddell AM, Koh DM, Shur JD. Response Evaluation Criteria in Gastrointestinal and Abdominal Cancers: Which to Use and How to Measure. Radiographics 2024; 44:e230047. [PMID: 38662587 DOI: 10.1148/rg.230047] [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: 05/29/2024]
Abstract
As the management of gastrointestinal malignancy has evolved, tumor response assessment has expanded from size-based assessments to those that include tumor enhancement, in addition to functional data such as those derived from PET and diffusion-weighted imaging. Accurate interpretation of tumor response therefore requires knowledge of imaging modalities used in gastrointestinal malignancy, anticancer therapies, and tumor biology. Targeted therapies such as immunotherapy pose additional considerations due to unique imaging response patterns and drug toxicity; as a consequence, immunotherapy response criteria have been developed. Some gastrointestinal malignancies require assessment with tumor-specific criteria when assessing response, often to guide clinical management (such as watchful waiting in rectal cancer or suitability for surgery in pancreatic cancer). Moreover, anatomic measurements can underestimate therapeutic response when applied to molecular-targeted therapies or locoregional therapies in hypervascular malignancies such as hepatocellular carcinoma. In these cases, responding tumors may exhibit morphologic changes including cystic degeneration, necrosis, and hemorrhage, often without significant reduction in size. Awareness of pitfalls when interpreting gastrointestinal tumor response is required to correctly interpret response assessment imaging and guide appropriate oncologic management. Data-driven image analyses such as radiomics have been investigated in a variety of gastrointestinal tumors, such as identifying those more likely to respond to therapy or recur, with the aim of delivering precision medicine. Multimedia-enhanced radiology reports can facilitate communication of gastrointestinal tumor response by automatically embedding response categories, key data, and representative images. ©RSNA, 2024 Test Your Knowledge questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Francesca Castagnoli
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Justin Mencel
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Derfel Ap Dafydd
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Jessica Gough
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Brent Drake
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Naami Charlotte Mcaddy
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Samuel Joseph Withey
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Angela Mary Riddell
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Dow-Mu Koh
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| | - Joshua David Shur
- From the Departments of Radiology (F.C., D.a.D., N.C.M., S.J.W., A.M.R., D.M.K., J.D.S.), Oncology (J.M.), Radiotherapy (J.G.), and Nuclear Medicine (B.D.), Royal Marsden Hospital, Downs Road, Sutton SM2 5PT, UK; and Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK (F.C., D.M.K.)
| |
Collapse
|
37
|
Sun D, Zhang X, Sun Y, Mu Z, Wang H, Zhang Y, Liang J, Lin Y. Early Structural, Biochemical, and Metabolic Responses to Anlotinib in Patients With Progressive Radioactive Iodine Refractory Differentiated Thyroid Cancer. Endocr Pract 2024; 30:456-464. [PMID: 38447630 DOI: 10.1016/j.eprac.2024.02.005] [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: 10/15/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE We aimed to assess the early efficacy of anlotinib in patients with progressive radioactive iodine refractory differentiated thyroid cancer at the structural, biochemical, and metabolic levels. METHODS Ten eligible patients were prospectively enrolled to receive anlotinib. Their responses were assessed at 6 weeks. Apart from the structural response according to Response Evaluation Criteria in Solid Tumors version 1.1, the biochemical response was assessed by serum thyroglobulin (Tg), and the metabolic response was assessed by 2-deoxy-2-[18F] fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) according to the European Organization for Research and Treatment of Cancer criteria. A safety profile was recorded. RESULTS Structurally controlled disease (20% partial response + 80% stable disease) was observed in all patients. The median longest diameter of target lesions shrank from 20.8 mm (IQR, 14.9-27.5) to 17.0 mm (IQR, 14.1-23.7) (P < .001), and the average shrinkage rate was -15.1 ± 14.1%. Sharp serum Tg reduction by 72.8 ± 16.4% was observed in 8 measurable patients. The 18F-FDG PET/CT-mapped glucose metabolic response was not quite comparable to the structural response, with 90% of the patients having controlled disease (30% partial metabolic response + 60% stable metabolic disease), whereas 10% presented progressive metabolic disease. The most common treatment-emergent adverse events (AEs) were hypertension (100%) and proteinuria (70%). Most AEs were grade 1 or 2, whereas grade 3 AEs occurred only in hypertension. CONCLUSION Anlotinib is generally well tolerated and can bring early disease control within the initial 6 weeks of treatment. The sharp biochemical response suggests Tg to be an early sensitive biomarker to anlotinib, whereas the heterogeneous metabolic response might play a complementary role.
Collapse
Affiliation(s)
- Di Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Yuqing Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Zhuanzhuan Mu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Hao Wang
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao 266012, China
| | - Yingqiang Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Peking University, Beijing 102206, China; Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China.
| | - Yansong Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China; Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China.
| |
Collapse
|
38
|
Chlorogiannis DD, Moussa AM, Zhao K, Alexander ES, Sofocleous CT, Sotirchos VS. Imaging Considerations before and after Liver-Directed Locoregional Treatments for Metastatic Colorectal Cancer. Diagnostics (Basel) 2024; 14:772. [PMID: 38611685 PMCID: PMC11011364 DOI: 10.3390/diagnostics14070772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer is a leading cause of cancer-related death. Liver metastases will develop in over one-third of patients with colorectal cancer and are a major cause of morbidity and mortality. Even though surgical resection has been considered the mainstay of treatment, only approximately 20% of the patients are surgical candidates. Liver-directed locoregional therapies such as thermal ablation, Yttrium-90 transarterial radioembolization, and stereotactic body radiation therapy are pivotal in managing colorectal liver metastatic disease. Comprehensive pre- and post-intervention imaging, encompassing both anatomic and metabolic assessments, is invaluable for precise treatment planning, staging, treatment response assessment, and the prompt identification of local or distant tumor progression. This review outlines the value of imaging for colorectal liver metastatic disease and offers insights into imaging follow-up after locoregional liver-directed therapy.
Collapse
Affiliation(s)
| | - Amgad M. Moussa
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ken Zhao
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Erica S. Alexander
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Vlasios S. Sotirchos
- Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
39
|
Ghodsi A, Hicks RJ, Iravani A. PET/Computed Tomography Transformation of Oncology: Immunotherapy Assessment. PET Clin 2024; 19:291-306. [PMID: 38199917 DOI: 10.1016/j.cpet.2023.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Immunotherapy approaches have changed the treatment landscape in a variety of malignancies with a high anti-tumor response. Immunotherapy may be associated with novel response and progression patterns that pose a substantial challenge to the conventional criteria for assessing treatment response, including response evaluation criteria in solid tumors (RECIST) 1.1. In addition to the morphologic details provided by computed tomography (CT) and MRI, hybrid molecular imaging emerges as a comprehensive imaging modality with the capacity to interrogate pathophysiological mechanisms like glucose metabolism. This review highlights the current status of 2-deoxy-2-[18F]fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) in prognostication, response monitoring, and identifying immune-related adverse events. Furthermore, it investigates the potential role of novel immuno-PET tracers that could complement the utilization of 18F-FDG PET/CT by imaging the specific pathways involved in immunotherapeutic strategies.
Collapse
Affiliation(s)
- Alireza Ghodsi
- Department of Radiology, University of Washington, 1144 Eastlake Avenue East, Seattle, WA 98109, USA
| | - Rodney J Hicks
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Australia; Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, Australia; The Melbourne Theranostic Innovation Centre, North Melbourne, Australia
| | - Amir Iravani
- Department of Radiology, University of Washington, 1144 Eastlake Avenue East, Seattle, WA 98109, USA.
| |
Collapse
|
40
|
Muzi M, Peterson LM, Specht JM, Hippe DS, Novakova-Jiresova A, Lee JH, Kurland BF, Mankoff DA, Obuchowski N, Linden HM, Kinahan PE. Repeatability of 18F-FDG uptake in metastatic bone lesions of breast cancer patients and implications for accrual to clinical trials. EJNMMI Res 2024; 14:32. [PMID: 38536511 PMCID: PMC10973316 DOI: 10.1186/s13550-024-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/06/2024] [Indexed: 04/01/2024] Open
Abstract
BACKGROUND Standard measures of response such as Response Evaluation Criteria in Solid Tumors are ineffective for bone lesions, often making breast cancer patients that have bone-dominant metastases ineligible for clinical trials with potentially helpful therapies. In this study we prospectively evaluated the test-retest uptake variability of 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) in a cohort of breast cancer patients with bone-dominant metastases to determine response criteria. The thresholds for 95% specificity of change versus no-change were then applied to a second cohort of breast cancer patients with bone-dominant metastases. METHODS For this study, nine patients with 38 bone lesions were imaged with 18F-FDG in the same calibrated scanner twice within 14 days. Tumor uptake was quantified by the most commonly used PET parameter, the maximum tumor voxel normalized by dose and body weight (SUVmax) and also by the mean of a 1-cc maximal uptake volume normalized by dose and lean-body-mass (SULpeak). The asymmetric repeatability coefficients with confidence intervals for SUVmax and SULpeak were used to determine the limits of 18F-FDG uptake variability. A second cohort of 28 breast cancer patients with bone-dominant metastases that had 146 metastatic bone lesions was imaged with 18F-FDG before and after standard-of-care therapy for response assessment. RESULTS The mean relative difference of SUVmax and SULpeak in 38 bone tumors of the first cohort were 4.3% and 6.7%. The upper and lower asymmetric limits of the repeatability coefficient were 19.4% and - 16.3% for SUVmax, and 21.2% and - 17.5% for SULpeak. 18F-FDG repeatability coefficient confidence intervals resulted in the following patient stratification using SULpeak for the second patient cohort: 11-progressive disease, 5-stable disease, 7-partial response, and 1-complete response with three inevaluable patients. The asymmetric repeatability coefficients response criteria for SULpeak changed the status of 3 patients compared to the standard Positron Emission Tomography Response Criteria in Solid Tumors of ± 30% SULpeak. CONCLUSION In evaluating bone tumor response for breast cancer patients with bone-dominant metastases using 18F-FDG SUVmax, the repeatability coefficients from test-retest studies show that reductions of more than 17% and increases of more than 20% are unlikely to be due to measurement variability. Serial 18F-FDG imaging in clinical trials investigating bone lesions in these patients, such as the ECOG-ACRIN EA1183 trial, benefit from confidence limits that allow interpretation of response.
Collapse
Affiliation(s)
- Mark Muzi
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA.
| | - Lanell M Peterson
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Jennifer M Specht
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Daniel S Hippe
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | | | - Jean H Lee
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Brenda F Kurland
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | | | | | - Hannah M Linden
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| | - Paul E Kinahan
- Department of Radiology, University of Washington Medical Center, 1959 NE Pacific Street, UW Box 356465, Seattle, Washington, 98195, USA
| |
Collapse
|
41
|
Rischin D, Hughes BGM, Basset-Séguin N, Schadendorf D, Bowyer S, Trabelsi Messai S, Meier F, Eigentler TK, Casado Echarren V, Stein B, Beylot-Barry M, Dalac S, Dréno B, Migden MR, Hauschild A, Schmults CD, Lim AM, Yoo SY, Paccaly AJ, Papachristos A, Nguyen JH, Okoye E, Seebach F, Booth J, Lowy I, Fury MG, Guminski A. High response rate with extended dosing of cemiplimab in advanced cutaneous squamous cell carcinoma. J Immunother Cancer 2024; 12:e008325. [PMID: 38471711 PMCID: PMC10936461 DOI: 10.1136/jitc-2023-008325] [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] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Cemiplimab (Libtayo®), a human monoclonal immunoglobulin G4 antibody to the programmed cell death-1 receptor, is approved for the treatment of patients with advanced cutaneous squamous cell carcinoma (CSCC), who are not candidates for curative surgery or curative radiation, using an every-3-weeks (Q3W) dosing interval. Pharmacokinetic modeling indicated that Ctrough of extended intravenous dosing of 600 mg every 4 weeks (Q4W) would be comparable to the approved intravenous dosage of 350 mg Q3W. We examined the efficacy, pharmacokinetics, and safety of cemiplimab dosed Q4W. METHODS In this open-label, phase II trial (ClinicalTrials.gov identifier NCT02760498), the cohort of patients ≥18 years old with advanced CSCC received cemiplimab 600 mg intravenously Q4W for up to 48 weeks. Tumor measurements were recorded every 8 weeks. The primary endpoint was objective response rate by independent central review. RESULTS Sixty-three patients with advanced CSCC were treated with cemiplimab. The median duration of follow-up was 22.4 months (range: 1.0-39.8). An objective response was observed in 39 patients (62%; 95% CI: 48.8% to 73.9%), with 22% of patients (n=14) achieving complete response and 40% (n=25) achieving partial response. The most common treatment-emergent adverse events were diarrhea, pruritus, and fatigue. CONCLUSIONS Extended dosing of cemiplimab 600 mg intravenously Q4W exhibited substantial antitumor activity, rapid and durable responses, and an acceptable safety profile in patients with advanced CSCC. These results confirm that cemiplimab is a highly active therapy for advanced CSCC. Additional data would help ascertain the benefit-risk profile for the 600 mg intravenous dosing regimen compared with the approved regimen.
Collapse
Affiliation(s)
- Danny Rischin
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital and University of Queensland, Brisbane, Queensland, Australia
| | | | - Dirk Schadendorf
- University Hospital Essen and Essen and German Cancer Consortium, Essen, Germany
| | - Samantha Bowyer
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | | | - Friedegund Meier
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases Dresden, Dresden, Germany
- Department of Dermatology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Thomas K Eigentler
- Department of Dermatology, Venereology and Allergology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Victoria Casado Echarren
- Department of Oncology, Oncohealth Institute, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Brian Stein
- ICON Cancer Centre, Adelaide, South Australia, Australia
| | - Marie Beylot-Barry
- Department of Dermatology, University Hospital of Bordeaux, Bordeaux, France
| | - Sophie Dalac
- Hospital Center University Dijon Bourgogne, Dijon, France
| | | | - Michael R Migden
- Departments of Dermatology and Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Chrysalyne D Schmults
- Dermatologic Surgery, Mohs Micrographic Surgery Center, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Annette M Lim
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Suk-Young Yoo
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Anne J Paccaly
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | | | | | - Emmanuel Okoye
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Frank Seebach
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Jocelyn Booth
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Israel Lowy
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Matthew G Fury
- Regeneron Pharmaceuticals, Inc, Tarrytown, New York, USA
| | - Alexander Guminski
- Department of Medical Oncology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
42
|
Ozturk H. Role of 18F-fluorodeoxyglucose positron-emission tomography/computed tomography in restaging of adrenocortical carcinoma. Rep Pract Oncol Radiother 2024; 28:728-736. [PMID: 38515818 PMCID: PMC10954270 DOI: 10.5603/rpor.98727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/04/2023] [Indexed: 03/23/2024] Open
Abstract
Background The objective was to retrospectively evaluate the contribution of fluorodeoxyglucose [18F] positron emission tomography/computed tomography (18FDG-PET/CT) to the re-staging of adrenocortical carcinoma (ACC). Materials and methods A total of 16 patients (10 males and 6 females), who underwent adrenalectomy due to adrenocortical carcinoma and 18FDG-PET/CT scan to re-stage the tumor between July 2007 and April 2013, were included in the present study. The mean age was 53.37 ± 13.91 years (min: 30, max: 74) The patients were required to fast for six hours prior to scanning, and whole-body PET scanning from the skull base to the upper thighs was performed approximately 1 h after the intravenous injection of 555 MBq of F-18 FDG. Whole body CT scanning was performed in the cranio-caudal direction. FDG-PET images were reconstructed using CT data for attenuation correction. Suspicious recurrent or metastatic lesions were confirmed by histopathology or clinical follow-up. Results Sensitivity, specificity, positive predictive value, negative predictive value and accuracy of 18FDG-PET/CT were 100%, 83.3%, 90.9%, 83.3%, and 93.7%, respectively. Conclusion 18FDG-PET/CT detects local recurrence and/or distant metastases with high accuracy in the re-staging of operated adrenocortical carcinoma. It is considered that the procedure could play an important role in treatment decision after the operation and post-operative follow-up and could influence the entire decision-making process.
Collapse
Affiliation(s)
- Hakan Ozturk
- Department of Urology, Izmir University of Economics, Izmir, Türkiye
| |
Collapse
|
43
|
van der Hiel B, Aalbersberg EA, van den Eertwegh AJM, de Wit-van der Veen LJ, Stokkel MPM, Lopez-Yurda M, Boellaard R, Kapiteijn EW, Hospers GAP, Aarts MJB, de Vos FYFL, Boers-Sonderen MJ, van der Veldt AAM, de Groot JWB, Haanen JBAG. The Predictive Value of FDG PET/CT for Determining Progression-Free Survival in Advanced Stage III-IV BRAF -Mutated Melanoma Patients Treated With Targeted Therapy-What Can Be Learned From Progression? Clin Nucl Med 2024; 49:138-145. [PMID: 38113329 DOI: 10.1097/rlu.0000000000004988] [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: 12/21/2023]
Abstract
PURPOSE The aims of this study were to investigate whether (early) PERCIST response monitoring with 18 F-FDG PET/CT is predictive for progression-free survival (PFS) in unresectable stage III or IV melanoma patients treated with BRAF/MEK inhibitor (MEKi) and to define dissemination patterns at progression with a lesion-based evaluation in direct comparison to baseline to improve our understanding of 18 F-FDG PET/CT during BRAF/MEKi. PATIENTS AND METHODS This prospective multicenter single-arm study included 70 patients with unresectable stage III/IV BRAF -mutated melanoma who underwent contrast-enhanced CT and 18 F-FDG PET/CT at baseline and 2 and 7 weeks during treatment with vemurafenib plus cobimetinib and at progression if possible. Tumor response assessment was done with RECIST1.1 and PERCIST. Follow-up PET/CT scans were visually compared with baseline to assess dissemination patterns. RESULTS Using RECIST1.1, PFS was not significantly different between the response groups ( P = 0.26). At 2 weeks, PERCIST median PFS was 15.7 months for patients with complete metabolic response (CMR) versus 8.3 months for non-CMR ( P = 0.035). The hazards ratio (HR) for progression/death in non-CMR versus CMR was 1.99 (95% confidence interval [CI], 1.03-3.84; P = 0.040) and 1.77 (95% CI, 0.91-3.43; P = 0.0935) when adjusting for lactate dehydrogenase (LDH). At 7 weeks, median PFS for PERCIST CMR was 16.7 months versus 8.5 months for non-CMR ( P = 0.0003). The HR for progression/death in the non-CMR group was significantly increased (HR, 2.94; 95% CI, 1.60-5.40; P = 0.0005), even when adjusting for LDH (HR, 2.65; 95% CI, 1.43-4.91; P = 0.0020). At week 7, 18 F-FDG PET/CT was false-positive in all 4 (6%) patients with new FDG-avid lesions but CMR of known metastases. When 18 F-FDG PET/CT was performed at progressive disease, 18/22 (82%) patients had progression of known metastases with or without new 18 F-FDG-avid lesions. CONCLUSIONS This study shows that PERCIST response assessment at week 7 is predictive for PFS, regardless of LDH. At 2 weeks, patients with CMR have longer PFS than patients with non-CMR, but different PET parameters should be investigated to further evaluate the added value of early 18 F-FDG PET/CT. Disease progression on PET/CT is predominated by progression of known metastases, and new 18 F-FDG-avid lesions during BRAF/MEKi are not automatically a sign of recurrent disease.
Collapse
Affiliation(s)
- Bernies van der Hiel
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Else A Aalbersberg
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | | | | | - Marcel P M Stokkel
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Ronald Boellaard
- Department of Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, Groningen
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht
| | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht
| | | | | | | | | |
Collapse
|
44
|
Al Mansour L, De Mestier L, Haissaguerre M, Afchain P, Hadoux J, Lecomte T, Morland D, Cottereau AS, De Rycke O, Tlili G, Tordo J, Janier M, Deville A, Walter T. Outcome on Mesenteric Mass Response of Small-Intestinal Neuroendocrine Tumors Treated by 177Lu-DOTATATE Peptide Receptor Radionuclide Therapy: The MesenLuth Study, a National Study from the French Group of Endocrine Tumors and Endocan-RENATEN Network. J Nucl Med 2024; 65:258-263. [PMID: 38212066 DOI: 10.2967/jnumed.123.266063] [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: 05/25/2023] [Revised: 10/25/2023] [Indexed: 01/13/2024] Open
Abstract
A mesenteric mass (MM), characterized by fibrotic reaction, is present in most small-intestinal neuroendocrine tumors (SI-NETs). 177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) has shown its efficacy in patients with progressive SI-NETs. However, because of specific tissue characteristics of desmoplastic MMs, we hypothesize that these lesions may be refractory to 177Lu-DOTATATE PRRT. Methods: From the national French Groupe d'étude des Tumeurs Endocrines database, we identified patients with an advanced SI-NET and a MM (≥2 cm with a retractile aspect) of a SI-NET treated by at least 1 course of 177Lu-DOTATATE PRRT. The primary endpoint was a MM objective response rate (ORR) of less than 5%. Secondary endpoints were metabolic response, MM-related safety, and clinical response, as well as MM progression-free survival (PFS) and non-MM PFS. Results: In total, 52 patients were included. The MM ORR was 4% (n = 2), and the non-MM ORR was 8% (n = 4). No patient had a MM metabolic response, and the non-MM metabolic response rate was 12% (n = 6). Among the 26 patients with baseline MM-related symptoms, 46% had a clinical response. Four patients presented with gastrointestinal complications during PRRT. The median MM-related PFS was not reached, and the non-MM PFS was 50.3 mo (95% CI, 38.2-61.7 mo). Conclusion: This study confirms that 177Lu-DOTATATE PRRT does not lead to morphologic response on MMs (ORR < 5%). However, it allows MM stability, with few MM-related side effects, and has a relevant impact on MM-related symptoms.
Collapse
Affiliation(s)
- Laure Al Mansour
- Service de Médecine Nucléaire, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
| | - Louis De Mestier
- Service de Pancréatologie et Oncologie Digestive, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Université Paris-Cité, Clichy, France
| | - Magalie Haissaguerre
- Service d'Endocrinologie, Hôpital Haut-Lévêque, Université de Bordeaux, Pessac, France
| | - Pauline Afchain
- Service d'Oncologie, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Julien Hadoux
- Service d'Oncologie, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry Lecomte
- Service de d'Hépato-gastro-entérologie et de Cancérologie Digestive, CHRU Hôpitaux de Tours, Tours, France
| | - David Morland
- Service de Médecine Nucléaire et Centre de Recherche en STIC EA 3804, Institut Godinot, Université de Reims Champagne-Ardenne, Reims, France
| | - Anne Segolene Cottereau
- Service de Médecine Nucléaire, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université René Descartes, Paris, France
| | - Ophelie De Rycke
- Service de Pancréatologie et Oncologie Digestive, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Université Paris-Cité, Clichy, France
| | - Ghoufrane Tlili
- Service de Médecine Nucléaire, CHU de Bordeaux, Bordeaux, France
| | - Jérémie Tordo
- Service de Médecine Nucléaire, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Marc Janier
- Service de Médecine Nucléaire, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
| | - Agathe Deville
- Service de Médecine Nucléaire, Hôpital Louis Pradel, Hospices Civils de Lyon, Bron, France
| | - Thomas Walter
- Service d'Oncologie Digestive, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France; and
- Gastro-entérologie et Technologies pour la Santé, Unité de Recherche, Centre de Recherche en Cancérologie de Lyon, INSERM, Lyon, France
| |
Collapse
|
45
|
Muzi M, Peterson LM, Specht JM, Hippe DS, Novakova-Jiresova A, Lee JH, Kurland BF, Mankoff DA, Obuchowski N, Linden HM, Kinahan PE. Repeatability of 18F-FDG uptake in metastatic bone lesions of breast cancer patients and implications for accrual to clinical trials. RESEARCH SQUARE 2024:rs.3.rs-3818932. [PMID: 38313279 PMCID: PMC10836099 DOI: 10.21203/rs.3.rs-3818932/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
BACKGROUND Standard measures of response such as Response Evaluation Criteria in Solid Tumors are ineffective for bone lesions, often making breast cancer patients with bone-dominant metastases ineligible for clinical trials with potentially helpful therapies. In this study we prospectively evaluated the test-retest uptake variability of 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) in a cohort of breast cancer patients with bone-dominant metastases to determine response criteria. The thresholds for 95% specificity of change versus no-change were then applied to a second cohort of breast cancer patients with bone-dominant metastases.In this study, nine patients with 38 bone lesions were imaged with 18F-FDG in the same calibrated scanner twice within 14 days. Tumor uptake was quantified as the maximum tumor voxel normalized by dose and body weight (SUVmax) and the mean of a 1-cc maximal uptake volume normalized by dose and lean-body-mass (SULpeak). The asymmetric repeatability coefficients with confidence intervals of SUVmax and SULpeak were used to determine limits of 18F-FDG uptake variability. A second cohort of 28 breast cancer patients with bone-dominant metastases that had 146 metastatic bone lesions was imaged with 18F-FDG before and after standard-of-care therapy for response assessment. RESULTS The mean relative difference of SUVmax in 38 bone tumors of the first cohort was 4.3%. The upper and lower asymmetric limits of the repeatability coefficient were 19.4% and -16.3%, respectively. The 18F-FDG repeatability coefficient confidence intervals resulted in the following patient stratification for the second patient cohort: 11-progressive disease, 5-stable disease, 7-partial response, and 1-complete response with three inevaluable patients. The asymmetric repeatability coefficients response criteria changed the status of 3 patients compared to standard the standard Positron Emission Tomography Response Criteria in Solid Tumors of ±30% SULpeak. CONCLUSIONS In evaluating bone tumor response for breast cancer patients with bone-dominant metastases using 18F-FDG uptake, the repeatability coefficients from test-retest studies show that reductions of more than 17% and increases of more than 20% are unlikely to be due to measurement variability. Serial 18F-FDG imaging in clinical trials investigating bone lesions from these patients, such as the ECOG-ACRIN EA1183 trial, benefit from confidence limits that allow interpretation of response.
Collapse
Affiliation(s)
- Mark Muzi
- University of Washington School of Medicine
| | | | | | | | | | - Jean H Lee
- University of Washington Department of Radiology
| | | | | | | | | | | |
Collapse
|
46
|
Mileva M, de Vries EGE, Guiot T, Wimana Z, Deleu AL, Schröder CP, Lefebvre Y, Paesmans M, Stroobants S, Huizing M, Aftimos P, Tol J, Van der Graaf WTA, Oyen WJG, Vugts DJ, Menke-van der Houven van Oordt CW, Brouwers AH, Piccart-Gebhart M, Flamen P, Gebhart G. Molecular imaging predicts lack of T-DM1 response in advanced HER2-positive breast cancer (final results of ZEPHIR trial). NPJ Breast Cancer 2024; 10:4. [PMID: 38184611 PMCID: PMC10771456 DOI: 10.1038/s41523-023-00610-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/04/2023] [Indexed: 01/08/2024] Open
Abstract
Efficacy of the human epidermal growth factor receptor (HER)2-targeting trastuzumab emtansine (T-DM1) in breast cancer (BC) relies on HER2 status determined by immunohistochemistry or fluorescence in-situ hybridization. Heterogeneity in HER2 expression, however, generates interest in "whole-body" assessment of HER2 status using molecular imaging. We evaluated the role of HER2-targeted molecular imaging in detecting HER2-positive BC lesions and patients unlikely to respond to T-DM1. Patients underwent zirconium-89 (89Zr) trastuzumab (HER2) PET/CT and [18F]-2-fluoro-2-deoxy-D-glucose (FDG) PET/CT before T-DM1 initiation. Based on 89Zr-trastuzumab uptake, lesions were visually classified as HER2-positive (visible/high uptake) or HER2-negative (background/close to background activity). According to proportion of FDG-avid tumor load showing 89Zr-trastuzumab uptake (entire/dominant part or minor/no part), patients were classified as HER2-positive and HER2-negative, respectively. Out of 265 measurable lesions, 93 (35%) were HER2-negative, distributed among 42 of the 90 included patients. Of these, 18 (19%) lesions belonging to 11 patients responded anatomically (>30% decrease in axial diameter from baseline) after three T-DM1 cycles, resulting in an 81% negative predictive value (NPV) of the HER2 PET/CT. In combination with early metabolic response assessment on FDG PET/CT performed before the second T-DM1 cycle, NPVs of 91% and 100% were reached in predicting lesion-based and patient-based (RECIST1.1) response, respectively. Therefore, HER2 PET/CT, alone or in combination with early FDG PET/CT, can successfully identify BC lesions and patients with a low probability of clinical benefit from T-DM1.
Collapse
Affiliation(s)
- Magdalena Mileva
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Thomas Guiot
- Department of Medical Physics, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Zéna Wimana
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Radiopharmacy, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Anne-Leen Deleu
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Carolien P Schröder
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yolene Lefebvre
- Department of Radiology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marianne Paesmans
- Data Center, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Sigrid Stroobants
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Manon Huizing
- Department of Medical Oncology, Antwerp University Hospital, Edegem, Belgium
| | - Philippe Aftimos
- Department of Medical Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jolien Tol
- Department of Internal Medicine, Jeroen Bosch Ziekenhuis, Den Bosch, The Netherlands
| | - Winette T A Van der Graaf
- Department of Medical Oncology, Antoni van Leeuwenhoek-Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Wim J G Oyen
- Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
- Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, The Netherlands
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Danielle J Vugts
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Adrienne H Brouwers
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martine Piccart-Gebhart
- Department of Medical Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Géraldine Gebhart
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
47
|
Martella S, Aiello MM, Bertaglia V, Cau R, Denaro N, Cadoni A, Novello S, Scartozzi M, Novello G, Soto Parra HJ, Saba L, Solinas C, Porcu M. Malignant Pleural Mesothelioma: Staging and Radiological Response Criteria in Patients Treated with Immune Checkpoint Inhibitors. Target Oncol 2024; 19:13-28. [PMID: 38063957 DOI: 10.1007/s11523-023-01017-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 02/01/2024]
Abstract
Malignant pleural mesothelioma (MPM) is a rare and challenging cancer associated with asbestos fiber exposure, which offers limited treatment options. Historically, platinum-based chemotherapy has been the primary approach, but recent developments have introduced immunotherapy as a promising alternative for the treatment of this disease. Nevertheless, the unique growth patterns and occasionally ambiguous progressive characteristics of MPM make the interpretation of radiological assessments complex. Immunotherapy further complicates matters by introducing unconventional treatment response patterns such as hyperprogression and pseudoprogression. Consequently, there is a growing imperative to integrate the standard RECIST criteria with the mesothelioma-specific mRECIST criteria (version 1.1), as outlined in iRECIST. This comprehensive review is driven by the intent to provide a valuable resource for radiologists and clinicians engaged in the diagnosis, treatment, and monitoring of MPM in the era of immunotherapy. Specifically, the current imaging methods employed for staging and follow-up will be exposed and discussed, with a focus on the technical specificities and the mRECIST 1.1 methodology. Furthermore, we will provide a discussion about major clinical trials related to the use of immunotherapy in MPM patients. Finally, the latest advancements in radiomics, the applications of artificial intelligence in MPM, and their potential impact on clinical practice for prognosis and therapy, are discussed.
Collapse
Affiliation(s)
- Serafina Martella
- Department of Medical Oncology, University Hospital Policlinico San Marco, Catania, Italy
| | - Marco Maria Aiello
- Department of Medical Oncology, University Hospital Policlinico San Marco, Catania, Italy
| | - Valentina Bertaglia
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Turin, Italy
| | - Riccardo Cau
- Department of Radiology, AOU Cagliari, S.S: 554, km 4,500, CAP: 09042, Monserrato (CA), Italy
| | - Nerina Denaro
- Department of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Andrea Cadoni
- Department of Medical Oncology, AOU Cagliari, Monserrato (CA), Italy
| | - Silvia Novello
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Turin, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, AOU Cagliari, Monserrato (CA), Italy
| | - Giuseppe Novello
- Department of Medical Oncology, University Hospital Policlinico San Marco, Catania, Italy
| | - Hector Josè Soto Parra
- Department of Medical Oncology, University Hospital Policlinico San Marco, Catania, Italy
| | - Luca Saba
- Department of Radiology, AOU Cagliari, S.S: 554, km 4,500, CAP: 09042, Monserrato (CA), Italy
| | - Cinzia Solinas
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Turin, Italy
| | - Michele Porcu
- Department of Radiology, AOU Cagliari, S.S: 554, km 4,500, CAP: 09042, Monserrato (CA), Italy.
| |
Collapse
|
48
|
Swiha M, Ayati N, Oprea-Lager DE, Ceci F, Emmett L. How to Report PSMA PET. Semin Nucl Med 2024; 54:14-29. [PMID: 37558507 DOI: 10.1053/j.semnuclmed.2023.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
Prostate cancer (PCa) is the most common cancer diagnosed in men in most developed countries and a leading cause of cancer-related morbidity and mortality. Prostate-specific membrane antigen positron emission tomography (PSMA-PET) has become a valuable tool in the staging and assessment of disease recurrence in PCa, and more recently for assessment for treatment eligibility to PSMA radioligand therapy (RLT). Harmonization of PSMA-PET interpretation and synoptic reports are needed to communicate concisely and reproducibly PSMA-PET/CT to referring physicians and to support clinician therapeutic management decisions in various stages of the disease. Uniform image interpretation is also important to provide comparable data between clinical trials and to translate such data from research to daily practice. This review provides an overview of the value of PSMA-PET across the different clinical stages of PCa, discusses published reporting criteria for PSMA-PET, identifies pitfalls in reporting PSMA, and provides recommendations for synoptic reports.
Collapse
Affiliation(s)
- Mina Swiha
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; Nuclear Medicine Division, Department of Medical Imaging, University of Western Ontario, London, Canada
| | - Narjess Ayati
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia; Garvan Institute of Medical Research, Sydney, Australia
| | - Daniela E Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University. Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia; Garvan Institute of Medical Research, Sydney, Australia.
| |
Collapse
|
49
|
Wang N. Editorial: Case reports in breast cancer : 2022. Front Oncol 2023; 13:1330225. [PMID: 38162508 PMCID: PMC10755864 DOI: 10.3389/fonc.2023.1330225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Affiliation(s)
- Nan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
50
|
Pavoine M, Thuillier P, Karakatsanis N, Legoupil D, Amrane K, Floch R, Le Pennec R, Salaün PY, Abgral R, Bourhis D. Clinical application of a population-based input function (PBIF) for a shortened dynamic whole-body FDG-PET/CT protocol in patients with metastatic melanoma treated by immunotherapy. EJNMMI Phys 2023; 10:79. [PMID: 38062278 PMCID: PMC10703763 DOI: 10.1186/s40658-023-00601-3] [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: 08/30/2023] [Accepted: 11/28/2023] [Indexed: 10/16/2024] Open
Abstract
BACKGROUND The aim was to investigate the feasibility of a shortened dynamic whole-body (dWB) FDG-PET/CT protocol and Patlak imaging using a population-based input function (PBIF), instead of an image-derived input function (IDIF) across the 60-min post-injection period, and study its effect on the FDG influx rate (Ki) quantification in patients with metastatic melanoma (MM) undergoing immunotherapy. METHODS Thirty-seven patients were enrolled, including a PBIF modeling group (n = 17) and an independent validation cohort (n = 20) of MM from the ongoing prospective IMMUNOPET2 trial. All dWB-PET data were acquired on Vision 600 PET/CT systems. The PBIF was fitted using a Feng's 4-compartments model and scaled to the individual IDIF tail's section within the shortened acquisition time. The area under the curve (AUC) of PBIFs was compared to respective IDIFs AUC within 9 shortened time windows (TW) in terms of linear correlation (R2) and Bland-Altman tests. Ki metrics calculated with PBIF vs IDIF on 8 organs with physiological tracer uptake, 44 tumoral lesions of MM and 11 immune-induced inflammatory sites of pseudo-progression disease were also compared (Mann-Whitney test). RESULTS The mean ± SD relative AUC bias was calculated at 0.5 ± 3.8% (R2 = 0.961, AUCPBIF = 1.007 × AUCIDIF). In terms of optimal use in routine practice and statistical results, the 5th-7th pass (R2 = 0.999 for both Ki mean and Ki max) and 5th-8th pass (mean ± SD bias = - 4.9 ± 6.5% for Ki mean and - 4.8% ± 5.6% for Ki max) windows were selected. There was no significant difference in Ki values from PBIF5_7 vs IDIF5_7 for physiological uptakes (p > 0.05) as well as for tumor lesions (mean ± SD Ki IDIF5_7 3.07 ± 3.27 vs Ki PBIF5_7 2.86 ± 2.96 100ml/ml/min, p = 0.586) and for inflammatory sites (mean ± SD Ki IDIF5_7 1.13 ± 0.59 vs Ki PBIF5_7 1.13 ± 0.55 100ml/ml/min, p = 0.98). CONCLUSION Our study showed the feasibility of a shortened dWB-PET imaging protocol with a PBIF approach, allowing to reduce acquisition duration from 70 to 20 min with reasonable bias. These findings open perspectives for its clinical use in routine practice such as treatment response assessment in oncology.
Collapse
Affiliation(s)
- Mathieu Pavoine
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Philippe Thuillier
- UMR INSERM 1304 GETBO, Brest, France
- Department of Endocrinology, University Hospital, Brest, France
| | - Nicolas Karakatsanis
- Department of Radiology, Weil Cornell Medical College of Cornell University, New York, NY, USA
| | | | - Karim Amrane
- Department of Oncology, Regional Hospital, Morlaix, France
| | - Romain Floch
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France
| | - Romain Le Pennec
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France
- UMR INSERM 1304 GETBO, Brest, France
| | - Pierre-Yves Salaün
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France
- UMR INSERM 1304 GETBO, Brest, France
| | - Ronan Abgral
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France
- UMR INSERM 1304 GETBO, Brest, France
| | - David Bourhis
- Department of Nuclear Medicine, University Hospital, 2 Avenue Foch, 29200, Brest, France.
- UMR INSERM 1304 GETBO, Brest, France.
| |
Collapse
|