1
|
Li H, Zhang H, Feng Z, Zhang X, Bi G, Du L, Zhao Y. A high biocompatible near-infrared fluorescent probe for tracking cysteine in multi-biosystem and its application in cervical cancer imaging. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 326:125185. [PMID: 39332175 DOI: 10.1016/j.saa.2024.125185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/27/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
Cysteine (Cys) plays a crucial role in the biological system and many related diseases. However, the detection of Cys in living organisms are still hindered by shortage of small molecule fluorophores that exhibit excitation and emission in the near-infrared region. Herein, we designed and synthesized a high water-soluble Cys probe (Cy7-SS) based on heptamethine cyanine scaffold. The prepared Cy7-SS displayed an enhanced emission at near-infrared region (NIR) after the recognition of Cys. Moreover, Cy7-SS not only exhibited high selectivity and sensitivity on the detection of in vitro Cys, but also could be used for endogenous Cys in living cells and C.elegans. The prepared strategy for the design of fluorophores expands the in vivo sensing toolkit for the precise analysis in clinical diagnosis.
Collapse
Affiliation(s)
- Hualong Li
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo, Shandong Province 255049, PR China; State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Huiling Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Zhixuan Feng
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China; School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Xiaojie Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Gehua Bi
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo, Shandong Province 255049, PR China.
| | - Libo Du
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| | - Yi Zhao
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo, Shandong Province 255049, PR China.
| |
Collapse
|
2
|
Cepero A, Yang Y, Young L, Huang J, Ji X, Yang F. Longitudinal FDG-PET Radiomics for Early Prediction of Treatment Response to Chemoradiation in Locally Advanced Cervical Cancer: A Pilot Study. Cancers (Basel) 2024; 16:3813. [PMID: 39594768 PMCID: PMC11592998 DOI: 10.3390/cancers16223813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Objectives: This study aimed to assess the capacity of longitudinal FDG-PET radiomics for early distinguishing between locally advanced cervical cancer (LACC) patients who responded to treatment and those who did not. Methods: FDG-PET scans were obtained before and midway through concurrent chemoradiation for a study cohort of patients with LACC. Radiomics features related to image textures were extracted from the primary tumor volumes and stratified for relevance to treatment response status with the aid of random forest recursive feature elimination. Predictive models based on the k-nearest neighbors time series classifier were developed using the top-selected features to differentiate between responders and non-responders. The performance of the developed models was evaluated using receiver operating characteristic (ROC) curve analysis and n-fold cross-validation. Results: The top radiomics features extracted from scans taken midway through treatment showed significant differences between the two responder groups (p-values < 0.0005). In contrast, those from pretreatment scans did not exhibit significant differences. The AUC of the mean ROC curve for the predictive model based on the top features from pretreatment scans was 0.8529, while it reached 0.9420 for those derived midway through treatment scans. Conclusions: The study highlights the potential of longitudinal FDG-PET radiomics extracted midway through treatment for predicting response to chemoradiation in LACC patients and emphasizes that interim PET scans could be crucial in personalized medicine, ultimately enhancing therapeutic outcomes for LACC.
Collapse
Affiliation(s)
- Alejandro Cepero
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| | - Yidong Yang
- The First Affiliated Hospital of the University of Science and Technology of China, Hefei 230001, China
| | - Lori Young
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jianfeng Huang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Xuemei Ji
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Fei Yang
- Department of Radiation Oncology, University of Miami School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
3
|
Smith CLC, Zwezerijnen GJC, Wiegers SE, Jauw YWS, Lugtenburg PJ, Zijlstra JM, Yaqub M, Boellaard R. Feasibility of Using 18F-FDG PET/CT Radiomics and Machine Learning to Detect Drug-Induced Interstitial Lung Disease. Diagnostics (Basel) 2024; 14:2531. [PMID: 39594197 PMCID: PMC11592839 DOI: 10.3390/diagnostics14222531] [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: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Bleomycin is an oncolytic and antibiotic agent used to treat various human cancers because of its antitumor activity. Unfortunately, up to 46% of the patients treated with bleomycin develop drug-induced interstitial lung disease (DIILD) and potentially life-threatening interstitial pulmonary fibrosis. Tools and biomarkers for predicting and detecting DIILD are limited. Therefore, we aimed to evaluate the feasibility of 18F-FDG PET/CT, PET radiomics, and machine learning in distinguishing DIILD in an explorative pilot study. METHODS Eighteen Hodgkin's lymphoma (HL) patients, of whom 10 developed DIILD after treatment with bleomycin, were retrospectively included. Five diffuse large B-cell lymphoma (DLBCL) patients were included as a control group since they were not treated with bleomycin. All patients underwent 18F-FDG PET/CT scans before (baseline) and during treatment (interim). Structural changes were assessed by changes in Hounsfield Units (HUs). The 18F-FDG PET scans were used to assess metabolic changes by examining the feasibility of 504 radiomics features, including the mean activity of the lungs (SUVmean). A Random Forest (RF) classifier evaluated the identification and prediction of DIILD based on PET radiomics features. RESULTS HL patients who developed DIILD showed a significant increase in standard SUV metrics (SUVmean; p = 0.012, median increase 37.4%), and in some regional PET radiomics features (texture strength; p = 0.009, median increase 101.6% and zone distance entropy; p = 0.019, median increase 18.5%), while this was not found in HL patients who did not develop DIILD and DLBCL patients. The RF classifier correctly identified DIILD in 72.2% of the patients and predicted the development of DIILD correctly in 50% of the patients. There were no significant differences in HUs over time within all three patient groups. CONCLUSIONS Our explorative longitudinal pilot study suggests that certain regional 18F-FDG PET radiomics features can effectively identify DIILD in HL patients treated with bleomycin, as significant longitudinal increases were observed in SUVmean, texture strength, and zone distance entropy after the development of DIILD. The metabolic activity of these features did not significantly increase over time in DLBCL patients and HL patients who did not develop DIILD. This indicates that 18F-FDG PET radiomics, with and without machine learning, might serve as potential biomarkers for detecting DIILD.
Collapse
Affiliation(s)
- Charlotte L. C. Smith
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Gerben J. C. Zwezerijnen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sanne E. Wiegers
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Yvonne W. S. Jauw
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Pieternella J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands
| | - Josée M. Zijlstra
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
4
|
Kim SY, Chung HW, So Y, Lee MH, Lee EJ. Recent Updates of PET in Lymphoma: FDG and Beyond. Biomedicines 2024; 12:2485. [PMID: 39595051 PMCID: PMC11592097 DOI: 10.3390/biomedicines12112485] [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: 09/07/2024] [Revised: 10/12/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Lymphoma is one of the most common cancers worldwide, categorized into Hodgkin lymphoma and non-Hodgkin lymphoma. 18F-fluorodeoxyglucose positron emission tomography (FDG PET) has become an essential imaging tool for evaluating patients with lymphoma in terms of initial diagnosis, staging, prognosis, and treatment response assessment. Recent advancements in imaging technology and methodologies, along with the development of artificial intelligence, have revolutionized the evaluation of complex imaging data, enhancing the diagnostic and predictive power of PET in lymphoma. However, FDG is not cancer-specific, but it primarily reflects glucose metabolism, which has prompted the investigation of alternative PET tracers to address this limitation. Novel PET radiotracers, such as fibroblast activation protein inhibitors targeting the tumor microenvironment, have recently shown promising results in evaluating various malignancies compared to FDG PET. Furthermore, with the rapid advancements in immunotherapy and the favorable imaging properties of 89Zr, immunoPET has emerged as a promising modality, offering insights into the functional and molecular status of the immune system. ImmunoPET can also facilitate the development of new antibody therapeutics and radioimmunotherapy by providing pharmacokinetic and pharmacodynamic data. This review provides comprehensive insights into the current clinical applications of FDG PET in lymphoma, while also exploring novel PET imaging radiotracers beyond FDG, discussing their mechanisms of action and potential impact on patient management.
Collapse
Affiliation(s)
- Sung-Yong Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Hyun Woo Chung
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
- Research Institute of Medical Science, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Young So
- Department of Nuclear Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea;
| | - Mark Hong Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea; (S.-Y.K.); (M.H.L.)
| | - Eun Jeong Lee
- Department of Nuclear Medicine, Seoul Medical Center, 156 Sinnae-ro, Jungnang-gu, Seoul 02053, Republic of Korea;
| |
Collapse
|
5
|
Chauvie S, Castellino A, Bergesio F, De Maggi A, Durmo R. Lymphoma: The Added Value of Radiomics, Volumes and Global Disease Assessment. PET Clin 2024; 19:561-568. [PMID: 38910057 DOI: 10.1016/j.cpet.2024.05.009] [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: 06/25/2024]
Abstract
Lymphoma represents a condition that holds promise for cure with existing treatment modalities; nonetheless, the primary clinical obstacle lies in advancing therapeutic outcomes by pinpointing high-risk individuals who are unlikely to respond favorably to standard therapy. In this article, the authors will delineate the significant strides achieved in the lymphoma field, with a particular emphasis on the 3 prevalent subtypes: Hodgkin lymphoma, diffuse large B-cell lymphomas, and follicular lymphoma.
Collapse
Affiliation(s)
- Stéphane Chauvie
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy.
| | | | - Fabrizio Bergesio
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy
| | - Adriano De Maggi
- Department of Medical Physics, 'Santa Croce e Carle Hospital, Cuneo, Italy
| | - Rexhep Durmo
- Nuclear Medicine Division, Department of Radiology, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
6
|
Kishida M, Fujisawa M, Steidl C. Molecular biomarkers in classic Hodgkin lymphoma. Semin Hematol 2024:S0037-1963(24)00069-6. [PMID: 38969539 DOI: 10.1053/j.seminhematol.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/27/2024] [Indexed: 07/07/2024]
Abstract
Classic Hodgkin lymphoma is a unique B-cell derived malignancy featuring rare malignant Hodgkin and Reed Sternberg (HRS) cells that are embedded in a quantitively dominant tumor microenvironment (TME). Treatment of classic Hodgkin lymphoma has significantly evolved in the past decade with improving treatment outcomes for newly diagnosed patients and the minority of patients suffering from disease progression. However, the burden of toxicity and treatment-related long-term sequelae remains high in a typically young patient population. This highlights the need for better molecular biomarkers aiding in risk-adapted treatment strategies and predicting response to an increasing number of available treatments that now prominently involve multiple immunotherapy options. Here, we review modern molecular biomarker approaches that reflect both the biology of the malignant HRS cells and cellular components in the TME, while holding the promise to improve diagnostic frameworks for clinical decision-making and be feasible in clinical trials and routine practice. In particular, technical advances in sequencing and analytic pipelines using liquid biopsies, as well as deep phenotypic characterization of tissue architecture at single-cell resolution, have emerged as the new frontier of biomarker development awaiting further validation and implementation in routine diagnostic procedures.
Collapse
Affiliation(s)
- Makoto Kishida
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada
| | - Manabu Fujisawa
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada; Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Christian Steidl
- Centre for Lymphoid Cancer department, BC Cancer, Vancouver, British Columbia, Canada; Institute of Medicine, University of Tsukuba, Ibaraki, Japan; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
7
|
Chen H, Fang Y, Gu J, Sun P, Yang L, Pan F, Wu H, Ye T. Dual-Layer Spectral Detector Computed Tomography Quantitative Parameters: A Potential Tool for Lymph Node Activity Determination in Lymphoma Patients. Diagnostics (Basel) 2024; 14:149. [PMID: 38248026 PMCID: PMC10814325 DOI: 10.3390/diagnostics14020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Dual-energy CT has shown promising results in determining tumor characteristics and treatment effectiveness through spectral data by assessing normalized iodine concentration (nIC), normalized effective atomic number (nZeff), normalized electron density (nED), and extracellular volume (ECV). This study explores the value of quantitative parameters in contrast-enhanced dual-layer spectral detector CT (SDCT) as a potential tool for detecting lymph node activity in lymphoma patients. A retrospective analysis of 55 lymphoma patients with 289 lymph nodes, assessed through 18FDG-PET/CT and the Deauville five-point scale, revealed significantly higher values of nIC, nZeff, nED, and ECV in active lymph nodes compared to inactive ones (p < 0.001). Generalized linear mixed models showed statistically significant fixed-effect parameters for nIC, nZeff, and ECV (p < 0.05). The area under the receiver operating characteristic curve (AUROC) values of nIC, nZeff, and ECV reached 0.822, 0.845, and 0.811 for diagnosing lymph node activity. In conclusion, the use of g nIC, nZeff, and ECV as alternative imaging biomarkers to PET/CT for identifying lymph node activity in lymphoma holds potential as a reliable diagnostic tool that can guide treatment decisions.
Collapse
Affiliation(s)
- Hebing Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Yuxiang Fang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Jin Gu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Peng Sun
- Clinical & Technical Support, Philips Healthcare, Floor 7, Building 2, World Profit Center, Beijing 100000, China;
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Hongying Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| | - Tianhe Ye
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue #1277, Wuhan 430022, China; (H.C.); (Y.F.); (J.G.); (L.Y.); (F.P.)
- Hubei Province Key Laboratory of Molecular Imaging, Jiefang Avenue #1277, Wuhan 430022, China
| |
Collapse
|
8
|
Alderuccio JP, Reis IM, Hamadani M, Nachiappan M, Leslom S, Kahl BS, Ai WZ, Radford J, Solh M, Ardeshna KM, Hess BT, Lunning MA, Zinzani PL, Stathis A, Carlo-Stella C, Lossos IS, Caimi PF, Han S, Yang F, Kuker RA, Moskowitz CH. PET/CT Biomarkers Enable Risk Stratification of Patients with Relapsed/Refractory Diffuse Large B-cell Lymphoma Enrolled in the LOTIS-2 Clinical Trial. Clin Cancer Res 2024; 30:139-149. [PMID: 37855688 PMCID: PMC10872617 DOI: 10.1158/1078-0432.ccr-23-1561] [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: 07/07/2023] [Revised: 09/11/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE Significant progress has occurred in developing quantitative PET/CT biomarkers in diffuse large B-cell lymphoma (DLBCL). Total metabolic tumor volume (MTV) is the most extensively studied, enabling assessment of FDG-avid tumor burden associated with outcomes. However, prior studies evaluated the outcome of cytotoxic chemotherapy or chimeric antigen receptor T-cell therapy without data on recently approved FDA agents. Therefore, we aimed to assess the prognosis of PET/CT biomarkers in patients treated with loncastuximab tesirine. EXPERIMENTAL DESIGN We centrally reviewed screening PET/CT scans of patients with relapsed/refractory DLBCL enrolled in the LOTIS-2 (NCT03589469) study. MTV was obtained by computing individual volumes using the SUV ≥4.0 threshold. Other PET/CT metrics, clinical factors, and the International Metabolic Prognostic Index (IMPI) were evaluated. Logistic regression was used to assess the association between biomarkers and treatment response. Cox regression was used to determine the effect of biomarkers on time-to-event outcomes. We estimated biomarker prediction as continuous and binary variables defined by cutoff points. RESULTS Across 138 patients included in this study, MTV with a cutoff point of 96 mL was the biomarker associated with the highest predictive performance in univariable and multivariable models to predict failure to achieve complete metabolic response (OR, 5.42; P = 0.002), progression-free survival (HR, 2.68; P = 0.002), and overall survival (HR, 3.09; P < 0.0001). IMPI demonstrated an appropriate performance, however, not better than MTV alone. CONCLUSIONS Pretreatment MTV demonstrated robust risk stratification, with those patients demonstrating high MTV achieving lower responses and survival to loncastuximab tesirine in relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Isildinha M. Reis
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mehdi Hamadani
- Medical College of Wisconsin, Milwaukee, WI, United States
| | - Muthiah Nachiappan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Salman Leslom
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Brad S. Kahl
- Washington University, St. Louis, MO, United States
| | - Weiyun Z. Ai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - John Radford
- NIHR Clinical Research Facility, University of Manchester and the Christie NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Melhem Solh
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA, United States
| | - Kirit M. Ardeshna
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Brian T. Hess
- Medical University of South Carolina, Charleston, SC, United States
| | - Matthew A. Lunning
- University of Nebraska Medical Center- Fred and Pamela Buffett Cancer Center, Omaha, NE, United States
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and Department of Oncology and Hematology, Humanitas Research Hospital–IRCCS, Milano, Italy
| | - Izidore S. Lossos
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Paolo F. Caimi
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH, United States
| | - Sunwoo Han
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Fei Yang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Russ A. Kuker
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Craig H. Moskowitz
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|