1
|
Ferrández MC, Golla SSV, Eertink JJ, Wiegers SE, Zwezerijnen GJC, Heymans MW, Lugtenburg PJ, Kurch L, Hüttmann A, Hanoun C, Dührsen U, Barrington SF, Mikhaeel NG, Ceriani L, Zucca E, Czibor S, Györke T, Chamuleau MED, Zijlstra JM, Boellaard R. Validation of an Artificial Intelligence-Based Prediction Model Using 5 External PET/CT Datasets of Diffuse Large B-Cell Lymphoma. J Nucl Med 2024; 65:1802-1807. [PMID: 39362767 DOI: 10.2967/jnumed.124.268191] [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: 06/06/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
The aim of this study was to validate a previously developed deep learning model in 5 independent clinical trials. The predictive performance of this model was compared with the international prognostic index (IPI) and 2 models incorporating radiomic PET/CT features (clinical PET and PET models). Methods: In total, 1,132 diffuse large B-cell lymphoma patients were included: 296 for training and 836 for external validation. The primary outcome was 2-y time to progression. The deep learning model was trained on maximum-intensity projections from PET/CT scans. The clinical PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, SUVpeak, age, and performance status. The PET model included metabolic tumor volume, maximum distance from the bulkiest lesion to another lesion, and SUVpeak Model performance was assessed using the area under the curve (AUC) and Kaplan-Meier curves. Results: The IPI yielded an AUC of 0.60 on all external data. The deep learning model yielded a significantly higher AUC of 0.66 (P < 0.01). For each individual clinical trial, the model was consistently better than IPI. Radiomic model AUCs remained higher for all clinical trials. The deep learning and clinical PET models showed equivalent performance (AUC, 0.69; P > 0.05). The PET model yielded the highest AUC of all models (AUC, 0.71; P < 0.05). Conclusion: The deep learning model predicted outcome in all trials with a higher performance than IPI and better survival curve separation. This model can predict treatment outcome in diffuse large B-cell lymphoma without tumor delineation but at the cost of a lower prognostic performance than with radiomics.
Collapse
Affiliation(s)
- Maria C Ferrández
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sandeep S V Golla
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sanne E Wiegers
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn W Heymans
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lars Kurch
- Clinic and Polyclinic for Nuclear Medicine, Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sally F Barrington
- School of Biomedical Engineering and Imaging Sciences, King's College London and Guy's and St Thomas' PET Centre, King's Health Partners, King's College London, London, United Kingdom
| | - N George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Luca Ceriani
- Department of Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Emanuele Zucca
- SAKK Swiss Group for Clinical Cancer Research, Bern, Switzerland
- Department of Oncology, Oncology Institute of Southern Switzerland-EOC, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; and
| | - Sándor Czibor
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Tamás Györke
- Department of Nuclear Medicine, Medical Imaging Centre, Semmelweis University, Budapest, Hungary
| | - Martine E D Chamuleau
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
2
|
Frontzek F, Renaud L, Dührsen U, Poeschel V, Bernard S, Chartier L, Ketterer N, Récher C, Fitoussi O, Held G, Casasnovas O, Haioun C, Mounier N, Tilly H, Morschhauser F, Le Gouill S, Karsten IE, Duns G, Steidl C, Scott DW, Klapper W, Rosenwald A, Ott G, Molina T, Lenz G, Ziepert M, Altmann B, Thieblemont C, Schmitz N. Identification, risk factors, and clinical course of CNS relapse in DLBCL patients across 19 prospective phase 2 and 3 trials-a LYSA and GLA/ DSHNHL collaboration. Leukemia 2024; 38:2225-2234. [PMID: 39152324 DOI: 10.1038/s41375-024-02371-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
Progression or relapse in the central nervous system (CNS) remains a rare but mostly fatal event for patients with diffuse large B-cell lymphoma (DLBCL). In a retrospective analysis of 5189 patients treated within 19 prospective German and French phase 2/3 trials, we identified 159 patients experiencing a CNS event (relapse: 62%, progression: 38%). Intracerebral, meningeal, intraspinal, or combined involvement was reported in 44%, 31%, 3%, and 22% of patients, respectively. 62 of 155 evaluable patients (40%) showed concurrent systemic progression/ relapse. 82% of all CNS events occurred within two years after study inclusion or randomization. 87% of patients showed extranodal involvement outside the CNS. Patients generally had poor outcomes with a median overall survival (OS) of 3.4 months (95% CI 2.9-4.2) and a 2-year OS of 15% (10-22%). Outcomes did not differ depending on the site or time point of CNS events. Patients with isolated CNS events demonstrated significantly better OS (p = 0.023). Twenty-five patients were consolidated with autologous or allogeneic stem cell transplantation and achieved a 3-year OS of 36% (20-66%). This large study including more than 5000 DLBCL patients highlights the unmet medical need to improve the outcome of DLBCL patients suffering from CNS relapse.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Central Nervous System Neoplasms/therapy
- Central Nervous System Neoplasms/secondary
- Central Nervous System Neoplasms/mortality
- Central Nervous System Neoplasms/pathology
- Middle Aged
- Female
- Adult
- Aged
- Risk Factors
- Prospective Studies
- Young Adult
- Retrospective Studies
- Neoplasm Recurrence, Local/pathology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Disease Progression
- Aged, 80 and over
- Clinical Trials, Phase III as Topic
- Prognosis
- Recurrence
- Clinical Trials, Phase II as Topic
Collapse
Affiliation(s)
- Fabian Frontzek
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada.
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany.
| | - Loïc Renaud
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Viola Poeschel
- Department of Internal Medicine 1 (Oncology, Hematology, Clinical Immunology and Rheumatology), Saarland University Medical School, Homburg/Saar, Germany
| | - Sophie Bernard
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | | | - Nicolas Ketterer
- Centre d'Oncologie-Hématologie, Clinique Bois-Cerf, Lausanne, Switzerland
| | - Christian Récher
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Olivier Fitoussi
- Oncologie-Hematologie, Polyclinique Bordeaux Nord Aquitaine, Bordeaux, France
| | - Gerhard Held
- Department for Hematology and Oncology, Westpfalz - Klinikum Kaiserslautern, Kaiserslautern, Germany
| | - Olivier Casasnovas
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Corinne Haioun
- Lymphoid malignancies unit-APHP, Hopital Henri Mondor, Creteil, France
| | | | - Hervé Tilly
- INSERM U1245, Centre Henri Becquerel, Rouen, France
| | | | | | - Imke E Karsten
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany
| | - Gerben Duns
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David W Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | | | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart, Germany
| | - Thierry Molina
- Universite de Paris, Assistance Publique-Hôpitaux de Paris, Hopital Necker, Anatomo-pathologie, Paris, France
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany
| | - Marita Ziepert
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Bettina Altmann
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Catherine Thieblemont
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | - Norbert Schmitz
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany.
| |
Collapse
|
3
|
Maas CCHM, van Klaveren D, Durmaz M, Visser O, Issa DE, Posthuma EFM, Zijlstra JM, Chamuleau MED, Lugtenburg PJ, Kersten MJ, Dinmohamed AG. Comparative effectiveness of 6x R-CHOP21 versus 6x R-CHOP21 + 2 R for patients with advanced-stage diffuse large B-cell lymphoma. Blood Cancer J 2024; 14:157. [PMID: 39266543 PMCID: PMC11393348 DOI: 10.1038/s41408-024-01137-0] [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: 06/18/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
First-line treatment for advanced-stage diffuse large B-cell lymphoma (DLBCL) typically involves 6x R-CHOP21 or 6x R-CHOP21 with two additional rituximab administrations (6x R-CHOP21 + 2 R). In contemporary practice, this treatment choice might be guided by interim PET scan results. This nationwide, population-based study investigates the comparative effectiveness of these treatment regimens in an era where interim PET-guided treatment decisions were not standard practice. Utilizing the Netherlands Cancer Registry, we identified 1577 adult patients diagnosed with advanced-stage DLBCL between 2014-2018 who completed either 6x R-CHOP21 (43%) or 6x R-CHOP21 + 2 R (57%). We used propensity scores to assess differences in event-free survival (EFS) and overall survival (OS). At five years, EFS (hazard ratio of 6x R-CHOP21 + 2 R versus 6x R-CHOP21 [HR] = 0.89; 95% confidence interval [CI], 0.72-1.09) and OS (HR = 0.93; 95% CI, 0.73-1.18) were not significantly different between both regimens. In exploratory risk-stratified analysis according to the International Prognostic Index (IPI), high-IPI patients (i.e., scores of 4-5) benefit most from 6x R-CHOP21 + 2 R (5-year absolute risk difference of EFS = 16.8%; 95% CI, -0.4%-34.1% and OS = 12.1%; 95% CI, -5.4-29.6%). Collectively, this analysis reveals no significant differences on average in EFS and OS between the two treatments. However, the potential benefits for high-risk patients treated with 6x R-CHOP21 + 2 R underscore the need for future research.
Collapse
Affiliation(s)
- Carolien C H M Maas
- Department of Public Health, Erasmus University Medical Centre, Rotterdam, The Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands.
| | - David van Klaveren
- Department of Public Health, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Müjde Durmaz
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Otto Visser
- Department of Registration, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Djamila E Issa
- Department of Internal Medicine, Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - Eduardus F M Posthuma
- Department of Internal Medicine, Reinier de Graaf Gasthuis, Delft, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Josée M Zijlstra
- Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martine E D Chamuleau
- Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, The Netherlands
| | - Marie José Kersten
- Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
- LYMMCARE (Lymphoma and Myeloma Center Amsterdam), Amsterdam, The Netherlands
| | - Avinash G Dinmohamed
- Department of Public Health, Erasmus University Medical Centre, Rotterdam, The Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands.
- Amsterdam UMC, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands.
- LYMMCARE (Lymphoma and Myeloma Center Amsterdam), Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Patel K, Ivanov A, Jocelyn T, Hantel A, Garcia JS, Abel GA. Patient-Reported Outcomes in Phase 3 Clinical Trials for Blood Cancers: A Systematic Review. JAMA Netw Open 2024; 7:e2414425. [PMID: 38829615 PMCID: PMC11148691 DOI: 10.1001/jamanetworkopen.2024.14425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 06/05/2024] Open
Abstract
Importance Published research suggests that patient-reported outcomes (PROs) are neither commonly collected nor reported in randomized clinical trials (RCTs) for solid tumors. Little is known about these practices in RCTs for hematological malignant neoplasms. Objective To evaluate the prevalence of PROs as prespecified end points in RCTs of hematological malignant neoplasms, and to assess reporting of PROs in associated trial publications. Evidence Review All issues of 8 journals known for publishing high-impact RCTs (NEJM, Lancet, Lancet Hematology, Lancet Oncology, Journal of Clinical Oncology, Blood, JAMA, and JAMA Oncology) between January 1, 2018, and December 13, 2022, were searched for primary publications of therapeutic phase 3 trials for adults with hematological malignant neoplasms. Studies that evaluated pretransplant conditioning regimens, graft-vs-host disease treatment, or radiotherapy as experimental treatment were excluded. Data regarding trial characteristics and PROs were extracted from manuscripts and trial protocols. Univariable analyses assessed associations between trial characteristics and PRO collection or reporting. Findings Ninety RCTs were eligible for analysis. PROs were an end point in 66 (73%) trials: in 1 trial (1%) as a primary end point, in 50 (56%) as a secondary end point, and in 15 (17%) as an exploratory end point. PRO data were reported in 26 of 66 primary publications (39%): outcomes were unchanged in 18 and improved in 8, with none reporting worse PROs with experimental treatment. Trials sponsored by for-profit entities were more likely to include PROs as an end point (49 of 55 [89%] vs 17 of 35 [49%]; P < .001) but were not significantly more likely to report PRO data (20 of 49 [41%] vs 6 of 17 [35%]; P = .69). Compared with trials involving lymphoma (18 of 29 [62%]) or leukemia or myelodysplastic syndrome (18 of 28 [64%]), those involving plasma cell disorders or multiple myeloma (27 of 30 [90%]) or myeloproliferative neoplasms (3 of 3 [100%]) were more likely to include PROs as an end point (P = .03). Similarly, compared with trials involving lymphoma (3 of 18 [17%]) or leukemia or myelodysplastic syndrome (5 of 18 [28%]), those involving plasma cell disorders or multiple myeloma (16 of 27 [59%]) or myeloproliferative neoplasms (2 of 3 [67%]) were more likely to report PROs in the primary publication (P = .01). Conclusions and Relevance In this systematic review, almost 3 of every 4 therapeutic RCTs for blood cancers collected PRO data; however, only 1 RCT included PROs as a primary end point. Moreover, most did not report resulting PRO data in the primary publication and when reported, PROs were either better or unchanged, raising concern for publication bias. This analysis suggests a critical gap in dissemination of data on the lived experiences of patients enrolled in RCTs for hematological malignant neoplasms.
Collapse
Affiliation(s)
- Kishan Patel
- Department of Internal Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Alexandra Ivanov
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tajmah Jocelyn
- Center for Clinical Investigation, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Andrew Hantel
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacqueline S. Garcia
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gregory A. Abel
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
5
|
Cherng HJJ, Herrera A. Circulating Tumor DNA in Diffuse Large B-Cell Lymphoma: from Bench to Bedside? Curr Treat Options Oncol 2024; 25:659-678. [PMID: 38656685 DOI: 10.1007/s11864-024-01201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
OPINION STATEMENT Diffuse large B-cell lymphoma (DLBCL) is a curable disease with variable outcomes due to underlying heterogeneous clinical and molecular features-features that are insufficiently characterized with our current tools. Due to these limitations, treatment largely remains a "one-size-fits-all" approach. Circulating tumor DNA (ctDNA) is a novel biomarker in cancers that is increasingly utilized for risk stratification and response assessment. ctDNA is readily detectable from the plasma of patients with DLBCL but has not yet been incorporated into clinical care to guide treatment. Here, we describe how ctDNA sequencing represents a promising technology in development to personalize the care of patients with DLBCL. We will review the different types of ctDNA assays being studied and the rapidly growing body of evidence supporting the utility of ctDNA in different treatment settings in DLBCL. Risk stratification by estimation of tumor burden and liquid genotyping, molecular response assessment during treatment, and monitoring for measurable residual disease (MRD) to identify therapy resistance and predict clinical relapse are all potential applications of ctDNA. It is time for clinical trials in DLBCL to utilize ctDNA as an integral biomarker for patient selection, response-adapted designs, and surrogate endpoints. As more ctDNA assays become commercially available for routine use, clinicians should consider liquid biopsy when treatment response is equivocal on imaging. Incorporating MRD may also guide decision-making if patients experience severe treatment toxicities. Though important barriers remain, we believe that ctDNA will soon be ready to transition from bench to bedside to individualize treatment for our patients with DLBCL.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/genetics
- Humans
- Circulating Tumor DNA/blood
- Biomarkers, Tumor/blood
- Liquid Biopsy/methods
- Disease Management
- Translational Research, Biomedical
- Precision Medicine/methods
- Prognosis
- Clinical Decision-Making
- Disease Susceptibility
Collapse
Affiliation(s)
- Hua-Jay J Cherng
- Lymphoma Service, Division of Hematology & Oncology, Columbia University Irving Medical Center, 177 Fort Washington Avenue, 6GN-Rm 435, New York, NY, 10032, USA.
| | - Alex Herrera
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
6
|
Parihar AS, Pant N, Subramaniam RM. Quarter-Century PET/CT Transformation of Oncology: Lymphoma. PET Clin 2024; 19:281-290. [PMID: 38403384 DOI: 10.1016/j.cpet.2023.12.014] [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: 02/27/2024]
Abstract
The clinical landscape of lymphomas has changed dramatically over the last 2 decades, including significant progress made in the understanding and utilization of imaging modalities and the available treatment options for both indolent and aggressive lymphomas. Since the introduction of hybrid PET/CT scanners in 2001, the indications of 18F-fluorodeoxyglucose (FDG) PET/CT in the management of lymphomas have grown rapidly. In today's clinical practice, FDG PET/CT is used in successful management of the vast majority patients with lymphomas.
Collapse
Affiliation(s)
- Ashwin Singh Parihar
- Mallinckrodt Institute of Radiology; Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA.
| | | | - Rathan M Subramaniam
- Faculty of Medicine, Nursing, Midwifery & Health Sciences, The University of Notre Dame Australia, Sydney, Australia; Department of Radiology, Duke University, Durham, NC, USA; Department of Medicine, University of Otago Medical School, Dunedin, New Zealand
| |
Collapse
|
7
|
Fox CP, Chaganti S, McIlroy G, Barrington SF, Burton C, Cwynarski K, Eyre TA, Illidge T, Kalakonda N, Kuhnl A, McKay P, Davies AJ. The management of newly diagnosed large B-cell lymphoma: A British Society for Haematology Guideline. Br J Haematol 2024; 204:1178-1192. [PMID: 38247115 PMCID: PMC7616447 DOI: 10.1111/bjh.19273] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/20/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024]
Affiliation(s)
| | - Sridhar Chaganti
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, UK
| | - Graham McIlroy
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Cathy Burton
- Department of Haematology, The Leeds Teaching Hospitals, Leeds, UK
| | - Kate Cwynarski
- Department of Haematology, University College London Hospitals, London, UK
| | - Toby A Eyre
- Oxford Cancer and Haematology Centre, Oxford University Hospitals, Oxford, UK
| | - Timothy Illidge
- Division of Cancer Sciences, Manchester NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Andrea Kuhnl
- Department of Haematology, King's College Hospital, London, UK
| | - Pam McKay
- Department of Haematology, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Andrew J Davies
- Cancer Sciences Division, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| |
Collapse
|
8
|
Abrisqueta P. New Insights into First-Line Therapy in Diffuse Large B-Cell Lymphoma: Are We Improving Outcomes? J Clin Med 2024; 13:1929. [PMID: 38610693 PMCID: PMC11012802 DOI: 10.3390/jcm13071929] [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: 01/15/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most prevalent subtype of lymphoma, comprising heterogeneous patient subgroups with distinctive biological and clinical characteristics. The R-CHOP combination (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) has been the standard initial treatment, yielding prolonged remissions in over 60% of patients with advanced-stage disease. Several attempts to enhance the outcomes of this regimen over the last two decades have shown limited success. Various novel therapeutic approaches have recently emerged in lymphoma, demonstrating promising results. These include small molecules, novel monoclonal antibodies, antibody-drug conjugates (ADC), bispecific antibodies (BsAbs), and chimeric antigen receptor (CAR) T-cell therapy. This review explores recent advancements in therapeutic strategies for DLBCL and their potential impact on the initial management of DLBCL patients.
Collapse
Affiliation(s)
- Pau Abrisqueta
- Department of Hematology, Vall d’Hebron Hospital Universitari, Experimental Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
9
|
Zhao W, Wu X, Huang S, Wang H, Fu H. Evaluation of therapeutic effect and prognostic value of 18F-FDG PET/CT in different treatment nodes of DLBCL patients. EJNMMI Res 2024; 14:20. [PMID: 38372908 PMCID: PMC10876506 DOI: 10.1186/s13550-024-01074-w] [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: 09/22/2023] [Accepted: 01/28/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND In the present study, we aimed to investigate the role of baseline (B), interim (I) and end-of-treatment (Eot) 18F-FDG PET/CT in assessing the prognosis of diffuse large B cell lymphoma (DLBCL), so as to identify patients who need intensive treatment at an early stage. METHODS A total of 127 DLBCL patients (62 men; 65 women; median age 62 years) were retrospectively analyzed in this study. Baseline (n = 127), interim (n = 127, after 3-4 cycles) and end-of-treatment (n = 53, after 6-8 cycles) PET/CT images were re-evaluated; semi-quantitative parameters such as maximum standardized uptake value of lesion-to-liver ratio (SUVmax(LLR)) and lesion-to-mediastinum ratio (SUVmax(LMR)), total metabolic tumor volume (TMTV) and total metabolic tumor volume (TLG) were recorded. ΔTLG1 was the change of interim relative to baseline TLG (I to B), ΔTLG2 (Eot to B). ΔSUVmax and ΔTMTV were the same algorithm. The visual Deauville 5-point scale (D-5PS) has been adopted as the major criterion for PET evaluation. Visual analysis (VA) and semi-quantitative parameters were assessed for the ability to predict progression-free survival (PFS) and overall survival (OS) by using Kaplan-Meier method, cox regression and logistic regression analysis. When visual and semi-quantitative analysis are combined, the result is only positive if both are positive. RESULTS At a median follow-up of 34 months, the median PFS and OS were 20 and 32 months. The survival curve analysis showed that advanced stage and IPI score with poor prognosis, ΔSUVmax(LLR)1 < 89.2%, ΔTMTV1 < 91.8% and ΔTLG1 < 98.8%, ΔSUVmax(LLR)2 < 86.4% were significantly related to the shortening of PFS in patient (p < 0.05). ΔSUVmax(LLR)1 < 83.2% and ΔTLG1 < 97.6% were significantly correlated with the shortening of OS in patients (p < 0.05). Visual analysis showed that incomplete metabolic remission at I-PET and Eot-PET increased the risk of progress and death. In terms of predicting recurrence by I-PET, the combination of visual and semi-quantitative parameters showed higher positive predictive value (PPV) and specificity than a single index. CONCLUSION Three to four cycles of R-CHOP treatment may be a time point for early prediction of early recurrence/refractory (R/R) patients and active preemptive treatment. Combined visual analysis with semi-quantitative parameters of 18F-FDG PET/CT at interim can improve prognostic accuracy and may allow for more precise screening of patients requiring early intensive therapy.
Collapse
Affiliation(s)
- Wenyu Zhao
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaodong Wu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Department of Radiology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Shuo Huang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hongliang Fu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
10
|
Tabari E, Lovejoy AF, Lin H, Bolen CR, Lor Saelee S, Lefkowitz JP, Kurtz DM, Bottos A, Nielsen TG, Parreira JM, Luong KT. NGS-determined molecular markers and disease burden metrics from ctDNA correlate with PFS in previously untreated DLBCL. Leuk Lymphoma 2024:1-11. [PMID: 38337191 DOI: 10.1080/10428194.2024.2301924] [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: 06/13/2023] [Accepted: 01/02/2024] [Indexed: 02/12/2024]
Abstract
Personalized risk stratification and treatment may help improve outcomes among patients with diffuse large B-cell lymphoma (DLBCL). We developed a next-generation sequencing (NGS)-based method to assess a range of potential prognostic indicators, and evaluated it using pretreatment plasma samples from 310 patients with previously untreated DLBCL from the GOYA trial (NCT01287741). Variant calls and DLBCL subtyping with the plasma-based method were concordant with corresponding tissue-based methods. Patients with a tumor burden greater than the median (p = .003) and non-germinal center B-cell-like (non-GCB) DLBCL (p = .049) had worse progression-free survival than patients with a tumor burden less than the median or GCB DLBCL. Multi-factor assessment combining orthogonal features from a single pretreatment plasma sample has promise as a prognostic indicator in this setting (p = .085). This minimally invasive plasma-based NGS assay could enable comprehensive prognostic assessment of patients in a clinical setting, with greater accessibility than current methods.
Collapse
Affiliation(s)
| | | | - Hai Lin
- Roche Sequencing Solutions, Pleasanton, CA, USA
| | | | | | | | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | | | | | |
Collapse
|
11
|
Zhang W, Liu X, Zhong Q, Wu T, Yang Y, Chen B, Jing H, Tang Y, Jin J, Liu Y, Song Y, Fang H, Lu N, Li N, Zhai Y, Zhang W, Wang S, Chen F, Yin L, Qi S, Li Y. Prediction of 5-year overall survival of diffuse large B-cell lymphoma on the pola-R-CHP regimen based on 2-year event-free survival and progression-free survival. Cancer Med 2024; 13:e6899. [PMID: 38180169 PMCID: PMC10807604 DOI: 10.1002/cam4.6899] [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: 09/25/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
This study aimed to predict the 5-year overall survival (OS) benefit of pola-R-CHP versus R-CHOP in the POLARIX trial based on the 2-year event-free survival (EFS) and progression-free survival (PFS) rates in diffuse large B-cell lymphoma (DLBCL). We identified randomized controlled trials (RCT) published before 31 May 2023. The correlation between the logarithmic (log) hazard ratio (HR) for EFS (HREFS) or PFS (HRPFS) and the HR for OS (HROS) was estimated at the trial-level. Correlation analysis was performed between 2-year PFS or EFS and 5-year OS rates at the treatment arm-level. Linear regression models were used to calculate the 5-year OS of pola-R-CHP and R-CHOP. In the included 20 RCTs, a linear correlation between HREFS (r = 0.765) or HRPFS (r = 0.534) and HROS was observed at the trial- level. Two-year EFS (r = 0.918) or 2-year PFS (r = 0.865) correlated linearly with 5-year OS. Linear regression analysis between 2-year EFS/PFS and 5-year OS gave estimated 5-year OS rates between pola-R-CHP and R-CHOP of 6.4% and 6.3%, respectively. Two-year EFS and PFS are feasible early endpoints in patients with DLBCL treated primarily with immunochemotherapy. The pola-R-CHP regimen is expected to improve 5-year OS.
Collapse
Affiliation(s)
- Wan‐Ru Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Xin Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Qiu‐Zi Zhong
- Beijing Hospital, National Geriatric Medical CenterBeijingChina
| | - Tao Wu
- Affiliated Hospital of Guizhou Medical University, Guizhou Cancer HospitalGuiyangGuizhouChina
| | - Yong Yang
- Fujian Medical University Union HospitalFuzhouFujianChina
| | - Bo Chen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Hao Jing
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Yuan Tang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Jing Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC)ShenzhenChina
| | - Yue‐Ping Liu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Yong‐Wen Song
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Hui Fang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Ning‐Ning Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Ning Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Yi‐Rui Zhai
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Wen‐Wen Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Shu‐Lian Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Fan Chen
- Affiliated Hospital of Qinghai UniversityQinghaiChina
| | - Lin Yin
- Affiliated Hospital of Qinghai UniversityQinghaiChina
| | - Shu‐Nan Qi
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| | - Ye‐Xiong Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC); Collaborative Innovation Center for Cancer MedicineBeijingChina
| |
Collapse
|
12
|
Puranik AD, Choudhury S, Ghosh S, Dev ID, Ramchandani V, Uppal A, Bhosale V, Palsapure A, Rungta R, Pandey R, Khatri S, George G, Satamwar Y, Maske R, Agrawal A, Shah S, Purandare NC, Rangarajan V. Tata Memorial Centre Evidence Based Use of Nuclear medicine diagnostic and treatment modalities in cancer. Indian J Cancer 2024; 61:S1-S28. [PMID: 38424680 DOI: 10.4103/ijc.ijc_52_24] [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: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/02/2024]
Abstract
ABSTRACT PET/CT and radioisotope therapy are diagnostic and therapeutic arms of Nuclear Medicine, respectively. With the emergence of better technology, PET/CT has become an accessible modality. Diagnostic tracers exploring disease-specific targets has led the clinicians to look beyond FDG PET. Moreover, with the emergence of theranostic pairs of radiopharmaceuticals, radioisotope therapy is gradually making it's way into treatment algorithm of common cancers in India. We therefore would like to discuss in detail the updates in PET/CT imaging and radionuclide therapy and generate a consensus-driven evidence based document which would guide the practitioners of Oncology.
Collapse
Affiliation(s)
- Ameya D Puranik
- Department of Nuclear Medicine and Molecular Imaging, Tata Memorial Hospital and Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Nielsen NB, Gerke O, Nielsen AL, Juul-Jensen K, Larsen TS, Møller MB, Hildebrandt MG. A retrospective head-to-head comparison of the Lugano classification and PERCIST for FDG-PET/CT response assessment in diffuse large B-cell lymphoma. Clin Physiol Funct Imaging 2024; 44:70-78. [PMID: 37501554 DOI: 10.1111/cpf.12851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common form of lymphoma. European guidelines recommend FDG-PET/CT for staging and end of treatment (EOT) response assessment, mid-treatment response assessment is optional. We compared the Lugano classification and PET Response Criteria In Solid Tumours (PERCIST) for FDG-PET/CT response assessment in DLBCL head-to-head. METHODS We retrospectively included patients with DLBCL who underwent first-line R-CHOP(-like) therapy (2013-2020). Interim and EOT FDG-PET/CT response were reevaluated using the Lugano classification and PERCIST. Response was dichotomized into complete metabolic response (CMR) versus non-CMR (interim and EOT) and responders versus nonresponders (interim only). The cutoff for nonresponse at interim was a Deauville score of 5 (DS5) with the Lugano classification and a partial metabolic response with ≤66% reduction in SULpeak using PERCIST (PERCIST66). RESULTS In multivariable Cox regression (N = 170), DS5 at interim, PERCIST66 at interim, non-CMR at EOT with the Lugano classification and non-CMR at EOT with PERCIST were predictive of progression-free survival (PFS). The Lugano classification and PERCIST agreed perfectly at interim and EOT and with 98.4% for the identification of nonresponders at interim. The accuracy for predicting events within 2 years of diagnosis was 84.2% for DS-5 at interim, 87.6% for PERCIST66 at interim, 86% for non-CMR with the Lugano classification at EOT and 83.3% for non-CMR with PERCIST at EOT. CONCLUSION The Lugano classification and PERCIST were equally predictive of PFS. Nonresponse at interim and non-CMR at EOT were predictive of poor PFS with comparable accuracy for predicting events within 2 years.
Collapse
Affiliation(s)
- Nicklas B Nielsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Open Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
- Centre for Personalized Response Monitoring in Oncology, Odense University Hospital, Odense, Denmark
| | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anne L Nielsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Karen Juul-Jensen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Thomas S Larsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Michael B Møller
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Malene G Hildebrandt
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Open Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
- Centre for Personalized Response Monitoring in Oncology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
14
|
Johansson P, Alig S, Richter J, Hanoun C, Rekowski J, Dürig J, Ylstra B, de Jong D, Klapper W, Alizadeh AA, Dührsen U, Hüttmann A. Outcome prediction by interim positron emission tomography and IgM monoclonal gammopathy in diffuse large B-cell lymphoma. Ann Hematol 2023; 102:3445-3455. [PMID: 37566280 PMCID: PMC10640472 DOI: 10.1007/s00277-023-05393-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023]
Abstract
In diffuse large B-cell lymphoma (DLBCL), a positive interim positron emission tomography (PET) scan predicts treatment failure, but the proportion of high-risk patients thus identified is small. To improve prediction, we combined the interim PET result with the presence or absence of an associated IgM gammopathy. Of 108 DLBCL patients participating in a prospective trial, nine (8%) were interim PET positive and 19 (18%) had an IgM gammopathy. The monoclonal protein was not associated with distinguishing genetic features, and its light chain restriction was not always concordant with the light chain restriction of the lymphoma. The information provided by interim PET and IgM gammopathy was combined to dichotomize the population into sizeable high-risk (1-2 adverse factors) and low-risk groups (no adverse factor) with widely different outcomes (population size, 25% vs. 75%; 3-year risk of progression, 51% vs. 10%; 3-year overall survival, 64% vs. 95%). Multivariable analyses including established risk factors revealed the interim PET result and the IgM gammopathy status to be the only factors significantly associated with outcome. Information about interim PET response and IgM gammopathy may be useful in studies testing risk-adapted treatment strategies.
Collapse
Affiliation(s)
- Patricia Johansson
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
- Institute of Cell Biology (Cancer Research), Faculty of Medicine, University of Duisburg-Essen, Essen, Germany
| | - Stefan Alig
- Department of Medicine, Divisions of Oncology and Hematology, Stanford University, Stanford, CA, USA
| | - Julia Richter
- Department of Hematopathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Jan Rekowski
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jan Dürig
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Daphne de Jong
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wolfram Klapper
- Department of Hematopathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ash A Alizadeh
- Department of Medicine, Divisions of Oncology and Hematology, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Institute for Stem Cell Biology & Regenerative Medicine, Stanford, CA, USA
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany.
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147, Essen, Germany
| |
Collapse
|
15
|
Puckrin R, Sterrett R, Chua N, Owen C, Duggan P, Shafey M, Stewart D. Consolidative Autotransplantation Achieves High Cure Rates in Adverse-Risk Large B Cell Lymphoma. Transplant Cell Ther 2023; 29:763.e1-763.e5. [PMID: 37703996 DOI: 10.1016/j.jtct.2023.09.005] [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: 08/12/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023]
Abstract
There remains an unmet need to optimize the first-line treatment of patients with high-risk large B cell lymphoma (LBCL), particularly those with a high International Prognostic Index (IPI) score or a positive interim positron emission tomography (PET) scan who experience poor outcomes with R-CHOP. This study was conducted to evaluate the real-world effectiveness of consolidative autologous stem cell transplantation (ASCT) among patients with high-risk LBCL. This retrospective study included consecutive patients with LBCL and IPI score 4 or 5 who underwent consolidative ASCT as part of first-line therapy in Alberta, Canada. Progression-free survival (PFS), overall survival (OS), and disease-specific survival (DSS) were determined using the Kaplan-Meier method. The study cohort comprised 114 patients with median age of 60 years (range, 18 to 73 years), of whom 81 (71%) had an IPI score of 4 and 33 (29%) had an IPI score of 5. With a median follow-up of 5.6 years, the 5-year PFS was 72% (95% confidence interval [CI], 62% to 79%), 5-year OS was 74% (95% CI, 64% to 81%), and 5-year DSS was 80% (95% CI, 71% to 87%). There was no significant difference in PFS among patients with and patients without positive interim PET scans (n = 24), MYC and BCL2 and/or BCL6 rearrangements (n = 26), or central nervous system involvement (n = 15). Consolidative ASCT is associated with high cure rates and favorable survival outcomes in patients with high-risk LBCL and may overcome the adverse prognostic impact of a positive interim PET scan.
Collapse
Affiliation(s)
- Robert Puckrin
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada.
| | - Russell Sterrett
- Cross Cancer Institute and University of Alberta, Edmonton, Canada
| | - Neil Chua
- Cross Cancer Institute and University of Alberta, Edmonton, Canada
| | - Carolyn Owen
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Peter Duggan
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Mona Shafey
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| | - Douglas Stewart
- Tom Baker Cancer Centre and University of Calgary, Calgary, Canada
| |
Collapse
|
16
|
Cuzzo B, Lipsky A, Cherng HJJ. Measurable Residual Disease Monitoring in Lymphoma. Curr Hematol Malig Rep 2023; 18:292-304. [PMID: 37930608 DOI: 10.1007/s11899-023-00715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
PURPOSE OF REVIEW The utility of analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and disease in the bone marrow as an adjunctive tool in caring for hematologic cancer patients is expanding. This holds true for lymphoma where these biomarkers are being explored as a means of genotyping and quantifying disease. Regarding the latter, they can be used to monitor measurable residual disease (MRD) during and after treatment. This holds potential for aiding clinical decisions amidst treatment, detecting earlier relapse, and improving prognostication. Here, we review the evidence to support these applications in a variety of lymphoma subtypes. RECENT FINDINGS Numerous clinical trials across a variety of lymphomas have demonstrated value in MRD monitoring. MRD monitoring is often prognostic for progression free survival (PFS) and even overall survival (OS) at several time points in a disease course, particularly when utilizing serial measurements. With regards to tailoring treatment, there are a growing number of trials examining MRD-adaptive treatment strategies to intensify or de-escalate treatment to individualize care. Lastly, MRD monitoring has been utilized successfully in detecting earlier relapse when compared to more standard methods of clinical surveillance such as radiographic assessment. Although not routinely implemented into clinical practice, MRD monitoring in lymphoma is helping shape the future landscape of this disease by aiding in prognostication, guiding therapy, and detecting earlier relapse. Steps to standardize and further examine this technology prospectively are being taken to bring MRD monitoring to the forefront of the field.
Collapse
Affiliation(s)
- Brian Cuzzo
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Andrew Lipsky
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Hua-Jay J Cherng
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA.
| |
Collapse
|
17
|
Lewis KL, Trotman J. Integration of PET in DLBCL. Semin Hematol 2023; 60:291-304. [PMID: 38326144 DOI: 10.1053/j.seminhematol.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 02/09/2024]
Abstract
F-fluorodeoxyglucose positron emission tomography-computerized tomography (18FDG-PET/CT) is the gold-standard imaging modality for staging and response assessment for most lymphomas. This review focuses on the utility of 18FDG-PET/CT, and its role in staging, prognostication and response assessment in diffuse large B-cell lymphoma (DLBCL), including emerging possibilities for future use.
Collapse
Affiliation(s)
| | - Judith Trotman
- Concord Repatriation General Hospital, Concord, NSW, Australia
| |
Collapse
|
18
|
Itti E, Blanc-Durand P, Berriolo-Riedinger A, Kanoun S, Kraeber-Bodéré F, Meignan M, Gat E, Gouill SL, Casasnovas RO, Bodet-Milin C. Validation of the ΔSUV max for Interim PET Interpretation in Diffuse Large B-Cell Lymphoma on the Basis of the GAINED Clinical Trial. J Nucl Med 2023; 64:1706-1711. [PMID: 37734837 DOI: 10.2967/jnumed.123.265871] [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: 04/14/2023] [Revised: 08/18/2023] [Indexed: 09/23/2023] Open
Abstract
The GAINED phase 3 trial (ClinicalTrials.gov identifier: NCT01659099) evaluated a PET-driven consolidative strategy in patients with diffuse large B-cell lymphoma. In this post hoc analysis, we aimed to compare the prognostic value of the per-protocol PET interpretation criteria (Menton 2011 consensus) with the change in the SUVmax (ΔSUVmax) alone. Methods: Real-time central review of 18F-FDG PET/CT was performed in 581 patients after 2 cycles (PET2) and 4 cycles (PET4) of immunochemotherapy using the Menton 2011 criteria, combining the ΔSUVmax (cutoffs of 66% and 70% at PET2 and PET4, respectively) and the Deauville scale. In "special cases," when the baseline SUVmax was less than 10.0 or the interim residual tumor SUVmax was greater than 5.0, the Menton 2011 experts' consensus agreed that the ΔSUVmax may not be reliable and that the Deauville score is preferable. Prognostic values of Menton 2011 and ΔSUVmax were evaluated by Kaplan-Meier analyses in terms of progression-free survival (PFS). Results: Seventeen percent of patients at PET2 (100/581) and 8% at PET4 (49/581) had PET-negative results by ΔSUVmax but were considered to have PET-positive results according to Menton 2011 with residual SUVmax of greater than 5.0. For the population with PET2-positive results, 2-y PFS was 70% (range, 58%-80%) with ΔSUVmax alone, whereas the outcome tended to be better for those who were considered to have PET-positive results by Menton 2011, 81% (range, 72%-87%). Conversely, all 10 patients with baseline SUVmax of less than 10.0 had PET2-positive results by ΔSUVmax but were considered to have PET2-negative results by Menton 2011. These patients had the same 2-y PFS as patients with PET2-negative/PET4-negative results, indicating that the ΔSUVmax yielded false-positive results in this situation. Conclusion: We recommend the use of the ΔSUVmax alone rather than the Menton 2011 criteria for assessing the interim metabolic response in patients with diffuse large B-cell lymphoma, except when the baseline SUVmax is less than 10.0.
Collapse
Affiliation(s)
- Emmanuel Itti
- Nuclear Medicine, CHU Henri Mondor, Paris-Est University, Créteil, France;
| | - Paul Blanc-Durand
- Nuclear Medicine, CHU Henri Mondor, Paris-Est University, Créteil, France
| | | | - Salim Kanoun
- Nuclear Medicine, Georges-François Leclerc Center, Dijon, France
| | | | - Michel Meignan
- Nuclear Medicine, CHU Henri Mondor, Paris-Est University, Créteil, France
| | - Elodie Gat
- Lymphoma Study Association Recherche Clinique (LYSARC), Pierre-Bénite, France
| | | | | | - Caroline Bodet-Milin
- Nantes University, Angers University, CHU Nantes, INSERM, CNRS, CRCI2NA, Nantes, France
| |
Collapse
|
19
|
Russler-Germain DA, Calhoun BR, Wu N, Watkins MP, Siegel BA, Bartlett NL, Mhlanga JC. FDG-PET/CT response assessment with qualitative Lugano criteria outperforms change in SUV max as a predictive biomarker in frontline treatment of mantle cell lymphoma. Leuk Lymphoma 2023; 64:1870-1874. [PMID: 37489928 DOI: 10.1080/10428194.2023.2239404] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023]
Affiliation(s)
- David A Russler-Germain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Brendan R Calhoun
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ningying Wu
- Public Health Sciences Division, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Marcus P Watkins
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Barry A Siegel
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Nancy L Bartlett
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Joyce C Mhlanga
- Division of Nuclear Medicine, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
20
|
Decruyenaere P, Giuili E, Verniers K, Anckaert J, De Grove K, Van der Linden M, Deeren D, Van Dorpe J, Offner F, Vandesompele J. Exploring the cell-free total RNA transcriptome in diffuse large B-cell lymphoma and primary mediastinal B-cell lymphoma patients as biomarker source in blood plasma liquid biopsies. Front Oncol 2023; 13:1221471. [PMID: 37954086 PMCID: PMC10634215 DOI: 10.3389/fonc.2023.1221471] [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: 05/12/2023] [Accepted: 09/18/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction Diffuse large B-cell lymphoma (DLBCL) and primary mediastinal B-cell lymphoma (PMBCL) are aggressive histological subtypes of non-Hodgkin's lymphoma. Improved understanding of the underlying molecular pathogenesis has led to new classification and risk stratification tools, including the development of cell-free biomarkers through liquid biopsies. The goal of this study was to investigate cell-free RNA (cfRNA) biomarkers in DLBCL and PMBCL patients. Materials and methods Blood plasma samples (n=168) and matched diagnostic formalin-fixed paraffin-embedded (FFPE) tissue samples (n=69) of DLBCL patients, PMBCL patients and healthy controls were collected between 2016-2021. Plasma samples were collected at diagnosis, at interim evaluation, after treatment, and in case of refractory or relapsed disease. RNA was extracted from 200 µl plasma using the miRNeasy serum/plasma kit and from FFPE tissue using the miRNeasy FFPE kit. RNA was subsequently sequenced on a NovaSeq 6000 instrument using the SMARTer Stranded Total RNA-seq pico v3 library preparation kit. Results Higher cfRNA concentrations were demonstrated in lymphoma patients compared to healthy controls. A large number of differentially abundant genes were identified between the cell-free transcriptomes of DLBCL patients, PMBCL patients, and healthy controls. Overlap analyses with matched FFPE samples showed that blood plasma has a unique transcriptomic profile that significantly differs from that of the tumor tissue. As a good concordance between tissue-derived gene expression and the immunohistochemistry Hans algorithm for cell-of-origin (COO) classification was demonstrated in the FFPE samples, but not in the plasma samples, a 64-gene cfRNA classifier was developed that can accurately determine COO in plasma. High plasma levels of a 9-gene signature (BECN1, PRKCB, COPA, TSC22D3, MAP2K3, UQCRHL, PTMAP4, EHD1, NAP1L1 pseudogene) and a 5-gene signature (FTH1P7, PTMAP4, ATF4, FTH1P8, ARMC7) were significantly associated with inferior progression-free and overall survival in DLBCL patients, respectively, independent of the NCCN-IPI score. Conclusion Total RNA sequencing of blood plasma samples allows the analysis of the cell-free transcriptome in DLBCL and PMBCL patients and demonstrates its unexplored potential in identifying diagnostic, cell-of-origin, and prognostic cfRNA biomarkers.
Collapse
Affiliation(s)
- Philippe Decruyenaere
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Edoardo Giuili
- Interuniversity Institute of Bioinformatics in Brussels (IB), Free University of Brussels, Brussels, Belgium
- Department of Biotechnology and Pharmacy, University of Bologna, Bologna, Italy
| | - Kimberly Verniers
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jasper Anckaert
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Katrien De Grove
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | | | - Dries Deeren
- Department of Hematology, Algemeen Ziekenhuis (AZ) Delta Roeselare-Menen, Roeselare, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Fritz Offner
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Jo Vandesompele
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Cox MC, Esposito F, Postorino M, Venditti A, Di Napoli A. Serum Paraprotein Is Associated with Adverse Prognostic Factors and Outcome, across Different Subtypes of Mature B-Cell Malignancies-A Systematic Review. Cancers (Basel) 2023; 15:4440. [PMID: 37760410 PMCID: PMC10527377 DOI: 10.3390/cancers15184440] [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: 07/20/2023] [Revised: 08/20/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
The presence of a serum paraprotein (PP) is usually associated with plasma-cell dyscrasias, Waldenstrom Macroglobulinemia/lymphoplasmacytic lymphoma, and cryoglobulinemia. However, PP is also often reported in other high- and low-grade B-cell malignancies. As these reports are sparse and heterogeneous, an overall view on this topic is lacking, Therefore, we carried out a complete literature review to detail the characteristics, and highlight differences and similarities among lymphoma entities associated with PP. In these settings, IgM and IgG are the prevalent PP subtypes, and their serum concentration is often low or even undetectable without immunofixation. The relevance of paraproteinemia and its prevalence, as well as the impact of IgG vs. IgM PP, seems to differ within B-NHL subtypes and CLL. Nonetheless, paraproteinemia is almost always associated with advanced disease, as well as with immunophenotypic, genetic, and clinical features, impacting prognosis. In fact, PP is reported as an independent prognostic marker of poor outcome. All the above call for implementing clinical practice, with the assessment of paraproteinemia, in patients' work-up. Indeed, more studies are needed to shed light on the biological mechanism causing more aggressive disease. Furthermore, the significance of paraproteinemia, in the era of targeted therapies, should be assessed in prospective trials.
Collapse
Affiliation(s)
- Maria Christina Cox
- UOC Malattie Linfoproliferative, Fondazione Policlinico Tor Vergata, 00133 Roma, Italy
| | - Fabiana Esposito
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Massimiliano Postorino
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Adriano Venditti
- Ematologia, Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, 00133 Roma, Italy; (F.E.)
| | - Arianna Di Napoli
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University, 00189 Roma, Italy;
| |
Collapse
|
22
|
Gumà J, Palazón-Carrión N, Rueda-Domínguez A, Sequero S, Calvo V, García-Arroyo R, Gómez-Codina J, Llanos M, Martínez-Banaclocha N, Provencio M. SEOM-GOTEL clinical guidelines on diffuse large B cell lymphoma (2022). Clin Transl Oncol 2023; 25:2749-2758. [PMID: 37289353 PMCID: PMC10425474 DOI: 10.1007/s12094-023-03206-5] [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: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 06/09/2023]
Abstract
Diffuse large B-cell lymphoma is the most frequent histological subtype of NHL and the paradigm for the management of aggressive lymphoma. An excisional or incisional lymph node biopsy evaluated by an experienced hemopathologist is recommended to establish the diagnosis. Twenty years following its introduction, R-CHOP remains the standard first-line treatment. No modification of this scheme (increased chemotherapy dose intensity, new monoclonal antibodies, or the addition of immunomodulators or anti-target agents) has significatively improved the clinical outcomes, whereas therapy for recurrence or progression is evolving rapidly. The irruption of CART cells, polatuzumab vedotin, tafasitamab, and CD20/CD3 bispecific antibodies are changing the natural history of relapsed patients and will challenge R-CHOP as the benchmark for newly diagnosed patients.
Collapse
Affiliation(s)
- Josep Gumà
- Medical Oncology Department, Hospital Universitari Sant Joan de Reus, IISPV, URV, Reus, Tarragona, Spain
| | | | - Antonio Rueda-Domínguez
- UGCI Medical Oncology, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Silvia Sequero
- Medical Oncology Department, Hospital Universitario San Cecilio, Granada, Spain
| | - Virginia Calvo
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Ramón García-Arroyo
- Medical Oncology Department, Complejo Hospitalario Universitario, Pontevedra, Spain
| | - José Gómez-Codina
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Marta Llanos
- Medical Oncology Department, Hospital Universitario de Canarias, Tenerife, Spain
| | - Natividad Martínez-Banaclocha
- Oncology Department, Hospital General Universitario Dr. Balmis, Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Mariano Provencio
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| |
Collapse
|
23
|
Zhi Y, Bao S, Mao J, Chai G, Liu C, Zhu J. Development and validation of a survival nomogram in patients with primary testicular diffuse large B-cell lymphoma. J Int Med Res 2023; 51:3000605231197052. [PMID: 37676929 PMCID: PMC10492492 DOI: 10.1177/03000605231197052] [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: 03/28/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
OBJECTIVE We developed and validated a nomogram for overall survival (OS) and cancer-specific survival (CSS) prediction in patients with primary testicular diffuse large B-cell lymphoma (PT-DLBCL). METHODS Patients diagnosed with PT-DLBCL were selected from the Surveillance, Epidemiology, and End Results (SEER) database. Independent prognostic factors were analyzed to establish a nomogram of OS and CSS. Patients were reclassified into high- and low-risk groups; survival was compared using Kaplan-Meier curves and log-rank tests. RESULTS We collected 1099 PT-DLBCL cases (2000-2019) from SEER and randomized into training (n = 771) and validation (n = 328) cohorts. In univariate and multivariate Cox regression analyses, five prognostic indicators (age, treatment modality, diagnosis year, Ann Arbor stage, laterality) were used to establish a nomogram of OS and CSS. The nomogram demonstrated excellent discrimination and calibration, with concordance indices in the training and validation cohorts of 0.702 (95% confidence interval [CI], 0.677-0.727) and 0.705 (95% CI 0.67-0.74) for OS and 0.694 (95% CI 0.663-0.725) and 0.680 (95% CI 0.63-0.72) for CSS. The calibration curve and ROC analysis indicated good predictive capability of the nomogram. CONCLUSIONS The constructed prognostic model showed good predictive value for PT-DLBCL to assist clinicians in developing individualized treatment strategies.
Collapse
Affiliation(s)
- Yongjin Zhi
- Department of Hematology, Taizhou People’s Hospital affiliated to Nanjing Medical University, Taizhou, Jiangsu Province, China
| | - Shuojing Bao
- Department of General Practice, Zhangdian People’s Hospital, Taizhou, Jiangsu Province, China
| | - Jingcheng Mao
- Department of Hematology, Taizhou People’s Hospital affiliated to Nanjing Medical University, Taizhou, Jiangsu Province, China
| | - Gufan Chai
- Department of Hematology, Taizhou People’s Hospital affiliated to Nanjing Medical University, Taizhou, Jiangsu Province, China
| | - Chengjiang Liu
- Department of General Practice, Anhui Medical University, Heifei, Anhui Province, China
| | - Jianfeng Zhu
- Department of Hematology, Taizhou People’s Hospital affiliated to Nanjing Medical University, Taizhou, Jiangsu Province, China
| |
Collapse
|
24
|
Witte HM, Riedl J, Künstner A, Fähnrich A, Ketzer J, Fliedner SMJ, Reimer N, Bernard V, von Bubnoff N, Merz H, Busch H, Feller A, Gebauer N. Molecularly Stratified Treatment Options in Primary Refractory DLBCL/HGBL with MYC and BCL2 or BCL6 Rearrangements (HGBL, NOS with MYC/BCL6). Target Oncol 2023; 18:749-765. [PMID: 37488307 PMCID: PMC10517902 DOI: 10.1007/s11523-023-00983-5] [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] [Accepted: 06/25/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND There is growing evidence supporting multidisciplinary molecular tumor boards (MTB) in solid tumors whereas hematologic malignancies remain underrepresented in this regard. OBJECTIVE The present study aimed to assess the clinical relevance of MTBs in primary refractory diffuse large B-cell lymphomas/high-grade B-cell lymphomas with MYC and BCL2 rearrangements (prDLBCL/HGBL-MYC/BCL2) (n = 13) and HGBL, not otherwise specified (NOS), with MYC and BCL6 rearrangements (prHGBL, NOS-MYC/BCL6) (n = 6) based on our previously published whole-exome sequencing (WES) cohort. PATIENTS AND METHODS For genomic analysis, the institutional MTB WES pipeline (University Cancer Center Schleswig-Holstein: UCCSH), certified for routine clinical diagnostics, was employed and supplemented by a comprehensive immunohistochemical work-up. Consecutive database research and annotation according to established evidence levels for molecularly stratified therapies was performed (NCT-DKTK/ESCAT). RESULTS Molecularly tailored treatment options with NCT-DKTK evidence level of at least m2A were identified in each case. We classified mutations in accordance with biomarker/treatment baskets and detected a heterogeneous spectrum of targetable alterations affecting immune evasion (IE; n = 30), B-cell targets (BCT; n = 26), DNA damage repair (DDR; n = 20), tyrosine kinases (TK; n = 13), cell cycle (CC; n = 7), PI3K-MTOR-AKT pathway (PAM; n = 2), RAF-MEK-ERK cascade (RME; n = 1), and others (OTH; n = 11). CONCLUSION Our virtual MTB approach identified potential molecularly targeted treatment options alongside targetable genomic signatures for both prDLBCL/HGBL-MYC/BCL2 and prHGBL, NOS-MYC/BCL6. These results underline the potential of MTB consultations in difficult-to-treat lymphomas early in the treatment sequence.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany.
- Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Bundeswehrkrankenhaus Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany.
| | - Jörg Riedl
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| | - Axel Künstner
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Anke Fähnrich
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Julius Ketzer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Department of Pediatrics, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Stephanie M J Fliedner
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Niklas Reimer
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Veronica Bernard
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| | - Hartmut Merz
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Hauke Busch
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
- Medical Systems Biology Group, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Alfred Feller
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Maria-Goeppert-Straße 9a, 23562, Lübeck, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Alee 160, 23538, Lübeck, Germany
| |
Collapse
|
25
|
Alderuccio JP, Kuker RA, Yang F, Moskowitz CH. Quantitative PET-based biomarkers in lymphoma: getting ready for primetime. Nat Rev Clin Oncol 2023; 20:640-657. [PMID: 37460635 DOI: 10.1038/s41571-023-00799-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 08/20/2023]
Abstract
The use of functional quantitative biomarkers extracted from routine PET-CT scans to characterize clinical responses in patients with lymphoma is gaining increased attention, and these biomarkers can outperform established clinical risk factors. Total metabolic tumour volume enables individualized estimation of survival outcomes in patients with lymphoma and has shown the potential to predict response to therapy suitable for risk-adapted treatment approaches in clinical trials. The deployment of machine learning tools in molecular imaging research can assist in recognizing complex patterns and, with image classification, in tumour identification and segmentation of data from PET-CT scans. Initial studies using fully automated approaches to calculate metabolic tumour volume and other PET-based biomarkers have demonstrated appropriate correlation with calculations from experts, warranting further testing in large-scale studies. The extraction of computer-based quantitative tumour characterization through radiomics can provide a comprehensive view of phenotypic heterogeneity that better captures the molecular and functional features of the disease. Additionally, radiomics can be integrated with genomic data to provide more accurate prognostic information. Further improvements in PET-based biomarkers are imminent, although their incorporation into clinical decision-making currently has methodological shortcomings that need to be addressed with confirmatory prospective validation in selected patient populations. In this Review, we discuss the current knowledge, challenges and opportunities in the integration of quantitative PET-based biomarkers in clinical trials and the routine management of patients with lymphoma.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Russ A Kuker
- Department of Radiology, Division of Nuclear Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Fei Yang
- Department of Radiation Oncology, Division of Medical Physics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Craig H Moskowitz
- Department of Medicine, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
26
|
Ferrández MC, Golla SSV, Eertink JJ, de Vries BM, Lugtenburg PJ, Wiegers SE, Zwezerijnen GJC, Pieplenbosch S, Kurch L, Hüttmann A, Hanoun C, Dührsen U, de Vet HCW, Zijlstra JM, Boellaard R. An artificial intelligence method using FDG PET to predict treatment outcome in diffuse large B cell lymphoma patients. Sci Rep 2023; 13:13111. [PMID: 37573446 PMCID: PMC10423266 DOI: 10.1038/s41598-023-40218-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
Convolutional neural networks (CNNs) may improve response prediction in diffuse large B-cell lymphoma (DLBCL). The aim of this study was to investigate the feasibility of a CNN using maximum intensity projection (MIP) images from 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) baseline scans to predict the probability of time-to-progression (TTP) within 2 years and compare it with the International Prognostic Index (IPI), i.e. a clinically used score. 296 DLBCL 18F-FDG PET/CT baseline scans collected from a prospective clinical trial (HOVON-84) were analysed. Cross-validation was performed using coronal and sagittal MIPs. An external dataset (340 DLBCL patients) was used to validate the model. Association between the probabilities, metabolic tumour volume and Dmaxbulk was assessed. Probabilities for PET scans with synthetically removed tumors were also assessed. The CNN provided a 2-year TTP prediction with an area under the curve (AUC) of 0.74, outperforming the IPI-based model (AUC = 0.68). Furthermore, high probabilities (> 0.6) of the original MIPs were considerably decreased after removing the tumours (< 0.4, generally). These findings suggest that MIP-based CNNs are able to predict treatment outcome in DLBCL.
Collapse
Affiliation(s)
- Maria C Ferrández
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| | - Sandeep S V Golla
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jakoba J Eertink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bart M de Vries
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Pieternella J Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sanne E Wiegers
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Gerben J C Zwezerijnen
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lars Kurch
- Department of Nuclear Medicine, Clinic and Polyclinic for Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Henrica C W de Vet
- Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Methodology, Amsterdam Public Health Research Institute, Methodology, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Cancer Center Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Shi Q, He Y, Yi H, Mu R, Jiang X, Fu D, Dong L, Qin W, Xu P, Cheng S, Song Q, Chen S, Wang L, Zhao W. Positron emission tomography-adapted therapy in low-risk diffuse large B-cell lymphoma: results of a randomized, phase III, non-inferiority trial. Cancer Commun (Lond) 2023; 43:896-908. [PMID: 37403255 PMCID: PMC10397561 DOI: 10.1002/cac2.12462] [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: 12/11/2022] [Revised: 05/17/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The current standard of care for non-bulky diffuse large B-cell lymphoma (DLBCL) patients with an International Prognostic Index (IPI) of 0 is four cycles of rituximab plus cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) but whether the same efficacy can be achieved with reduced chemotherapy regimen of four cycles for non-bulky DLBCL patients with an IPI of 1 remains unclear. This study compared four cycles versus six cycles of chemotherapy in non-bulky low-risk DLBCL patients with negative interim positron emission tomography with computed tomography (PET-CT, Deauville 1-3), irrespective of age and other IPI risk factors (IPI 0-1). METHODS This was an open-label, randomized, phase III, non-inferiority trial. Patients aged 14-75 years with newly diagnosed low-risk DLBCL, according to IPI, achieving PET-CT confirmed complete response (CR) after four cycles of R-CHOP were randomized (1:1) between four cycles of rituximab (4R-CHOP+4R arm) or two cycles of R-CHOP plus two cycles of rituximab (6R-CHOP+2R arm). The primary endpoint was 2-year progression-free survival (PFS), conducted in the intention-to-treat population. Safety was assessed in patients with at least one cycle of assigned treatment. The non-inferiority margin was -8%. RESULTS A total of 287 patients were included in the intention-to-treat analysis, the median follow-up was 47.3 months, and the 2-year PFS rate was 95% (95% confidence interval [CI], 92% to 99%) and 94% (95% CI, 91% to 98%) for the 4R-CHOP+4R and 6R-CHOP+2R arm. The absolute difference in 2-year PFS between the two arms was 1% (95% CI, -5% to 7%), supporting the non-inferiority of 4R-CHOP+4R. Grade 3-4 neutropenia was lower in the last four cycles of rituximab alone in the 4R-CHOP+4R arm (16.7% versus 76.9%), with decreased risk of febrile neutropenia (0.0% versus 8.4%) and infection (2.1% versus 14.0%). CONCLUSIONS For newly diagnosed low-risk DLBCL patients, interim PET-CT after four cycles of R-CHOP was effective in identifying patients with Deauville 1-3 who would have a good response and Deauville 4-5 patients who might have high-risk biological features or develop resistance. Reducing the standard six cycles to four cycles of chemotherapy had comparable clinical efficacy and fewer adverse events in low-risk, non-bulky DLBCL with interim PET-CT confirmed CR.
Collapse
Affiliation(s)
- Qing Shi
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Yang He
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Hong‐Mei Yi
- Department of PathologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Rong‐Ji Mu
- Clinical Research InstituteShanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Xu‐Feng Jiang
- Department of Nuclear MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Di Fu
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Lei Dong
- Department of PathologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Wei Qin
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Peng‐Peng Xu
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Shu Cheng
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Qi Song
- Department of RadiologyRuijin Hospital affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Sai‐Juan Chen
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
- Laboratory of Molecular PathologyPôle de Recherches Sino‐Français en Science du Vivant et GénomiqueShanghaiChina
| | - Li Wang
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
- Laboratory of Molecular PathologyPôle de Recherches Sino‐Français en Science du Vivant et GénomiqueShanghaiChina
| | - Wei‐Li Zhao
- State Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiShanghai Institute of HematologyRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
- Laboratory of Molecular PathologyPôle de Recherches Sino‐Français en Science du Vivant et GénomiqueShanghaiChina
| |
Collapse
|
28
|
Sworder BJ, Kurtz DM. Cell-free DNA in large B-cell lymphoma: MRD and beyond. Semin Hematol 2023; 60:142-149. [PMID: 37474409 PMCID: PMC10528139 DOI: 10.1053/j.seminhematol.2023.06.004] [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: 03/27/2023] [Revised: 05/31/2023] [Accepted: 06/24/2023] [Indexed: 07/22/2023]
Abstract
Large B-cell lymphomas (LBCLs) are a strikingly diverse set of diseases, including clinical, biological, and molecular heterogeneity. Despite a wealth of information resolving this heterogeneity in the research setting, applying molecular features routinely in the clinic remains challenging. The advent of circulating tumor DNA (ctDNA) liquid biopsies promises to unlock additional molecular information in the clinic, including mutational genotyping, molecular classification, and minimal residual disease detection. Here, we examine the technologies, applications, and studies exploring the utility of ctDNA in LBCLs.
Collapse
Affiliation(s)
- Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA; Stanford Cancer Institute, Stanford University, Stanford, CA.
| |
Collapse
|
29
|
Kaanders JHAM, Bussink J, Aarntzen EHJG, Braam P, Rütten H, van der Maazen RWM, Verheij M, van den Bosch S. [18F]FDG-PET-Based Personalized Radiotherapy Dose Prescription. Semin Radiat Oncol 2023; 33:287-297. [PMID: 37331783 DOI: 10.1016/j.semradonc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
PET imaging with 2'-deoxy-2'-[18F]fluoro-D-glucose ([18F]FDG) has become one of the pillars in the management of malignant diseases. It has proven value in diagnostic workup, treatment policy, follow-up, and as prognosticator for outcome. [18F]FDG is widely available and standards have been developed for PET acquisition protocols and quantitative analyses. More recently, [18F]FDG-PET is also starting to be appreciated as a decision aid for treatment personalization. This review focuses on the potential of [18F]FDG-PET for individualized radiotherapy dose prescription. This includes dose painting, gradient dose prescription, and [18F]FDG-PET guided response-adapted dose prescription. The current status, progress, and future expectations of these developments for various tumor types are discussed.
Collapse
Affiliation(s)
- Johannes H A M Kaanders
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands..
| | - Johan Bussink
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud university medical center, Nijmegen, The Netherlands
| | - Pètra Braam
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Heidi Rütten
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | | | - Marcel Verheij
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| | - Sven van den Bosch
- Department of Radiation Oncology, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Zanoni L, Bezzi D, Nanni C, Paccagnella A, Farina A, Broccoli A, Casadei B, Zinzani PL, Fanti S. PET/CT in Non-Hodgkin Lymphoma: An Update. Semin Nucl Med 2023; 53:320-351. [PMID: 36522191 DOI: 10.1053/j.semnuclmed.2022.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022]
Abstract
Non-Hodgkin lymphomas represents a heterogeneous group of lymphoproliferative disorders characterized by different clinical courses, varying from indolent to highly aggressive. 18F-FDG-PET/CT is the current state-of-the-art diagnostic imaging, for the staging, restaging and evaluation of response to treatment in lymphomas with avidity for 18F-FDG, despite it is not routinely recommended for surveillance. PET-based response criteria (using five-point Deauville Score) are nowadays uniformly applied in FDG-avid lymphomas. In this review, a comprehensive overview of the role of 18F-FDG-PET in Non-Hodgkin lymphomas is provided, at each relevant point of patient management, particularly focusing on recent advances on diffuse large B-cell lymphoma and follicular lymphoma, with brief updates also on other histotypes (such as marginal zone, mantle cell, primary mediastinal- B cell lymphoma and T cell lymphoma). PET-derived semiquantitative factors useful for patient stratification and prognostication and emerging radiomics research are also presented.
Collapse
Affiliation(s)
- Lucia Zanoni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Davide Bezzi
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Paccagnella
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy; Nuclear Medicine Unit, AUSL Romagna, Cesena, Italy
| | - Arianna Farina
- Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Alessandro Broccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Beatrice Casadei
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Nuclear Medicine, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Zeman MN, Akin EA, Merryman RW, Jacene HA. Interim FDG-PET/CT for Response Assessment of Lymphoma. Semin Nucl Med 2023; 53:371-388. [PMID: 36376131 DOI: 10.1053/j.semnuclmed.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022]
Abstract
The clinical use and prognostic value of interim FDG-PET/CT (iPET/CT), which is performed after treatment initiation but prior to its completion, varies by lymphoma subtype. Evidence supporting the prognostic value of iPET/CT is more robust for classical Hodgkin lymphoma (cHL), and in this lymphoma subtype, response-adapted treatment approaches guided by iPET/CT are a widely used standard of care for first-line therapy. The data supporting use of iPET/CT among patients with non-Hodgkin lymphoma (NHL) is less well-established, but failure to achieve complete metabolic response on iPET/CT is generally considered a poor prognostic factor with likely consequences for progression free survival. This review will present the available evidence supporting use of iPET/CT in lymphoma patients, particularly as it relates to prognostication and the ability to inform response-adapted treatment strategies. The latter will be addressed through a discussion on the major iPET-response adapted clinical trials with mention of ongoing trials. Special attention will be given to cHL and a few subtypes of NHL, including diffuse large B cell lymphoma (DLBCL), follicular lymphoma (FL), and peripheral T cell lymphoma (PTCL).
Collapse
Affiliation(s)
- Merissa N Zeman
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - Esma A Akin
- Department of Radiology, Division of Nuclear Medicine, George Washington University, Medical Faculty Associates, Washington, DC
| | - Reid W Merryman
- Harvard Medical School, Boston, MA; Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Heather A Jacene
- Department of Radiology, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA; Department of Imaging, Dana-Farber Cancer Institute, Boston, MA.
| |
Collapse
|
32
|
Michaud L, Bantilan K, Mauguen A, Moskowitz CH, Zelenetz AD, Schöder H. Prognostic Value of 18F-FDG PET/CT in Diffuse Large B-Cell Lymphoma Treated with a Risk-Adapted Immunochemotherapy Regimen. J Nucl Med 2023; 64:536-541. [PMID: 36549918 PMCID: PMC10071786 DOI: 10.2967/jnumed.122.264740] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Early identification of patients with diffuse large B-cell lymphoma (DLBCL) who are likely to experience disease recurrence or refractory disease after rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) would be useful for improving risk-adapted treatment strategies. We aimed to assess the prognostic value of 18F-FDG PET/CT parameters at baseline, interim, and end of treatment (EOT). Methods: We analyzed the prognostic impact of 18F-FDG PET/CT in 166 patients with DLBCL treated with a risk-adapted immunochemotherapy regimen. Scans were obtained at baseline, after 4 cycles of R-CHOP or 3 cycles of RR-CHOP (double dose of R) and 1 cycle of CHOP alone (interim) and 6 wk after completing therapy (EOT). Progression-free survival (PFS) and overall survival (OS) were estimated using Kaplan-Meier and the impact of clinical/PET factors assessed with Cox models. We also assessed the predictive ability of the recently proposed International Metabolic Prognostic Index (IMPI). Results: The median follow-up was 7.9 y. International Prognostic Index (IPI), baseline metabolic tumor volume (MTV), and change in maximum SUV (ΔSUVmax) at interim scans were statistically significant predictors for OS. Baseline MTV, interim ΔSUVmax, and EOT Deauville score were statistically significant predictors of PFS. Combining interim PET parameters demonstrated that patients with Deauville 4-5 and positive ΔSUVmax ≤ 70% at restaging (∼10% of the cohort) had extremely poor prognosis. The IMPI had limited discrimination and slightly overestimated the event rate in our cohort. Conclusion: Baseline MTV and interim ΔSUVmax predicted both PFS and OS with this sequential immunochemotherapy program. Combining interim Deauville score with interim ΔSUVmax may identify an extremely high-risk DLBCL population.
Collapse
Affiliation(s)
- Laure Michaud
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kurt Bantilan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Craig H Moskowitz
- Department of Medicine, University of Miami Health System, Miami, Florida
| | - Andrew D Zelenetz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
| |
Collapse
|
33
|
Torka P, Pederson LD, Knopp MV, Poon D, Zhang J, Kahl BS, Higley HR, Kelloff G, Friedberg JW, Schwartz LH, Wilson WH, Leonard JP, Bartlett NL, Schöder H, Ruppert AS. Is local review of positron emission tomography scans sufficient in diffuse large B-cell lymphoma clinical trials? A CALGB 50303 analysis. Cancer Med 2023; 12:8211-8217. [PMID: 36799072 PMCID: PMC10134372 DOI: 10.1002/cam4.5628] [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: 09/21/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Quantitative methods of Fluorodeoxyglucose Positron Emission Tomography (FDG-PET) interpretation, including the percent change in FDG uptake from baseline (ΔSUV), are under investigation in lymphoma to overcome challenges associated with visual scoring systems (VSS) such as the Deauville 5-point scale (5-PS). METHODS In CALGB 50303, patients with DLBCL received frontline R-CHOP or DA-EPOCH-R, and although there were no significant associations between interim PET responses assessed centrally after cycle 2 (iPET) using 5-PS with progression-free survival (PFS) or overall survival (OS), there were significant associations between central determinations of iPET ∆SUV with PFS/OS. In this patient cohort, we retrospectively compared local vs central iPET readings and evaluated associations between local imaging data and survival outcomes. RESULTS Agreement between local and central review was moderate (kappa = 0.53) for VSS and high (kappa = 0.81) for ∆SUV categories (<66% vs. ≥66%). ∆SUV ≥66% at iPET was significantly associated with PFS (p = 0.03) and OS (p = 0.002), but VSS was not. Associations with PFS/OS when applying local review vs central review were comparable. CONCLUSIONS These data suggest that local PET interpretation for response determination may be acceptable in clinical trials. Our findings also highlight limitations of VSS and call for incorporation of more objective measures of response assessment in clinical trials.
Collapse
Affiliation(s)
- Pallawi Torka
- Department of MedicineRoswell Park Cancer Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Levi D. Pederson
- Alliance Statistics and Data Management CenterMayo ClinicRochesterMinnesotaUSA
| | | | - David Poon
- Department of RadiologyThe Ohio State UniversityColumbusOhioUSA
| | - Jun Zhang
- Department of RadiologyThe Ohio State UniversityColumbusOhioUSA
| | - Brad S. Kahl
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | | | - Gary Kelloff
- Division of Cancer Treatment and DiagnosisNational Cancer Institute, National Institutes of HealthRockvilleMarylandUSA
| | | | | | - Wyndham H. Wilson
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of HealthRockvilleMarylandUSA
| | - John P. Leonard
- Department of MedicineWeill Cornell Medical CollegeNew YorkNew YorkUSA
| | - Nancy L. Bartlett
- Department of MedicineWashington University School of MedicineSt. LouisMissouriUSA
| | - Heiko Schöder
- Department of RadiologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Amy S. Ruppert
- Department of Internal MedicineThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
34
|
Tamaki N, Hirata K, Kotani T, Nakai Y, Matsushima S, Yamada K. Four-dimensional quantitative analysis using FDG-PET in clinical oncology. Jpn J Radiol 2023:10.1007/s11604-023-01411-4. [PMID: 36947283 PMCID: PMC10366296 DOI: 10.1007/s11604-023-01411-4] [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: 01/17/2023] [Accepted: 03/02/2023] [Indexed: 03/23/2023]
Abstract
Positron emission tomography (PET) with F-18 fluorodeoxyglucose (FDG) has been commonly used in many oncological areas. High-resolution PET permits a three-dimensional analysis of FDG distributions on various lesions in vivo, which can be applied for tissue characterization, risk analysis, and treatment monitoring after chemoradiotherapy and immunotherapy. Metabolic changes can be assessed using the tumor absolute FDG uptake as standardized uptake value (SUV) and metabolic tumor volume (MTV). In addition, tumor heterogeneity assessment can potentially estimate tumor aggressiveness and resistance to chemoradiotherapy. Attempts have been made to quantify intratumoral heterogeneity using radiomics. Recent reports have indicated the clinical feasibility of a dynamic FDG PET-computed tomography (CT) in pilot cohort studies of oncological cases. Dynamic imaging permits the assessment of temporal changes in FDG uptake after administration, which is particularly useful for differentiating pathological from physiological uptakes with high diagnostic accuracy. In addition, several new parameters have been introduced for the in vivo quantitative analysis of FDG metabolic processes. Thus, a four-dimensional FDG PET-CT is available for precise tissue characterization of various lesions. This review introduces various new techniques for the quantitative analysis of FDG distribution and glucose metabolism using a four-dimensional FDG analysis with PET-CT. This elegant study reveals the important role of tissue characterization and treatment strategies in oncology.
Collapse
Affiliation(s)
- Nagara Tamaki
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tomoya Kotani
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshitomo Nakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigenori Matsushima
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Yamada
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
35
|
Duarte S, Roque A, Saraiva T, Afonso C, Marques BA, Lima CB, Neves D, Lai AC, Costa G, Cipriano A, Geraldes C, Ruzickova L, Carda JP, Gomes M. Interim FDG 18-PET SUV max Variation Adds Prognostic Value to Deauville 5-Point Scale in the Identification of Patients with Ultra-High-Risk Diffuse Large B Cell Lymphoma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e107-e116. [PMID: 36567213 DOI: 10.1016/j.clml.2022.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/13/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Interim response evaluation by 18F-fluorodeoxyglucose positron emission tomography/computed tomography (iPET) in diffuse large B cell lymphoma (DLBCL) could be important to rule out disease progression and has been suggested to be predictive of survival. However, treatment guidance by iPET is not yet recommended for DLBCL in clinical practice. We aimed to compare the predictive value of iPET when utilizing the visual Deauville 5-point scale (DS) and the semiquantitative variation of maximum standardized uptake value (ΔSUVmax). MATERIALS AND METHODS We included 85 patients diagnosed with DLBCL and uniformly treated with standard protocols. iPET with DS of 1-3 and/or ΔSUVmax ≥66% was defined as negative. Univariable and multivariable Cox regression analyses were performed to determine the independent factors affecting progression free survival (PFS) or overall survival (OS) and to estimate PFS and OS. RESULTS iPET positivity, measured by DS or ΔSUVmax, showed predictive value of disease refractoriness, improved by combining DS and ΔSUVmax. After a median follow-up of 50.1 months, iPET was an independent predictor for both PFS and OS when interpreted by DS, but only for PFS by ΔSUVmax. Combined visual and semiquantitative analysis (D4-5 & ΔSUVmax<66%) was an independent predictor of PFS and OS, and allowed to identify an ultra-high-risk subgroup of patients with very dismal outcome, increasing the discriminating capacity for iPET. CONCLUSION Our study suggests that combined DS and ΔSUVmax in iPET assessment predicts refractory disease and distinguishes ultra-high-risk DLBCL patients with a very dismal prognosis, who may benefit from PET-guided therapy adjustment.
Collapse
Affiliation(s)
- Sara Duarte
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal.
| | - Adriana Roque
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Tiago Saraiva
- Nuclear Medicine Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Carolina Afonso
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Bárbara Almeida Marques
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Carla Barros Lima
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Dulcelena Neves
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Ana Catarina Lai
- Pathology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Gracinda Costa
- Nuclear Medicine Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Augusta Cipriano
- Pathology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - Catarina Geraldes
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lenka Ruzickova
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| | - José Pedro Carda
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Marília Gomes
- Clinical Hematology Department, Hospital and University Centre of Coimbra, Coimbra, Portugal
| |
Collapse
|
36
|
Sworder BJ, Kurtz DM, Alig SK, Frank MJ, Shukla N, Garofalo A, Macaulay CW, Shahrokh Esfahani M, Olsen MN, Hamilton J, Hosoya H, Hamilton M, Spiegel JY, Baird JH, Sugio T, Carleton M, Craig AFM, Younes SF, Sahaf B, Sheybani ND, Schroers-Martin JG, Liu CL, Oak JS, Jin MC, Beygi S, Hüttmann A, Hanoun C, Dührsen U, Westin JR, Khodadoust MS, Natkunam Y, Majzner RG, Mackall CL, Diehn M, Miklos DB, Alizadeh AA. Determinants of resistance to engineered T cell therapies targeting CD19 in large B cell lymphomas. Cancer Cell 2023; 41:210-225.e5. [PMID: 36584673 PMCID: PMC10010070 DOI: 10.1016/j.ccell.2022.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 10/17/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022]
Abstract
Most relapsed/refractory large B cell lymphoma (r/rLBCL) patients receiving anti-CD19 chimeric antigen receptor (CAR19) T cells relapse. To characterize determinants of resistance, we profiled over 700 longitudinal specimens from two independent cohorts (n = 65 and n = 73) of r/rLBCL patients treated with axicabtagene ciloleucel. A method for simultaneous profiling of circulating tumor DNA (ctDNA), cell-free CAR19 (cfCAR19) retroviral fragments, and cell-free T cell receptor rearrangements (cfTCR) enabled integration of tumor and both engineered and non-engineered T cell effector-mediated factors for assessing treatment failure and predicting outcomes. Alterations in multiple classes of genes are associated with resistance, including B cell identity (PAX5 and IRF8), immune checkpoints (CD274), and those affecting the microenvironment (TMEM30A). Somatic tumor alterations affect CAR19 therapy at multiple levels, including CAR19 T cell expansion, persistence, and tumor microenvironment. Further, CAR19 T cells play a reciprocal role in shaping tumor genotype and phenotype. We envision these findings will facilitate improved chimeric antigen receptor (CAR) T cells and personalized therapeutic approaches.
Collapse
Affiliation(s)
- Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Stefan K Alig
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Matthew J Frank
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Navika Shukla
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Andrea Garofalo
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Charles W Macaulay
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mohammad Shahrokh Esfahani
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mari N Olsen
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - James Hamilton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Hitomi Hosoya
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mark Hamilton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jay Y Spiegel
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - John H Baird
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Takeshi Sugio
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Mia Carleton
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Alexander F M Craig
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Sheren F Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bita Sahaf
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Natasha D Sheybani
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Joseph G Schroers-Martin
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chih Long Liu
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Jean S Oak
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael C Jin
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Sara Beygi
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA
| | - Andreas Hüttmann
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Christine Hanoun
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Ulrich Dührsen
- Department of Hematology, University Hospital of Essen, Essen, Germany
| | - Jason R Westin
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S Khodadoust
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robbie G Majzner
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA; Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA; Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Parker Institute for Cancer Immunotherapy, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - David B Miklos
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford, CA 94305, USA
| | - Ash A Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, 265 Campus Drive, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
Witte HM, Fähnrich A, Künstner A, Riedl J, Fliedner SMJ, Reimer N, Hertel N, von Bubnoff N, Bernard V, Merz H, Busch H, Feller A, Gebauer N. Primary refractory plasmablastic lymphoma: A precision oncology approach. Front Oncol 2023; 13:1129405. [PMID: 36923431 PMCID: PMC10008852 DOI: 10.3389/fonc.2023.1129405] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023] Open
Abstract
Introduction Hematologic malignancies are currently underrepresented in multidisciplinary molecular-tumor-boards (MTB). This study assesses the potential of precision-oncology in primary-refractory plasmablastic-lymphoma (prPBL), a highly lethal blood cancer. Methods We evaluated clinicopathological and molecular-genetic data of 14 clinically annotated prPBL-patients from initial diagnosis. For this proof-of-concept study, we employed our certified institutional MTB-pipeline (University-Cancer-Center-Schleswig-Holstein, UCCSH) to annotate a comprehensive dataset within the scope of a virtual MTB-setting, ultimately recommending molecularly stratified therapies. Evidence-levels for MTB-recommendations were defined in accordance with the NCT/DKTK and ESCAT criteria. Results Median age in the cohort was 76.5 years (range 56-91), 78.6% of patients were male, 50% were HIV-positive and clinical outcome was dismal. Comprehensive genomic/transcriptomic analysis revealed potential recommendations of a molecularly stratified treatment option with evidence-levels according to NCT/DKTK of at least m2B/ESCAT of at least IIIA were detected for all 14 prPBL-cases. In addition, immunohistochemical-assessment (CD19/CD30/CD38/CD79B) revealed targeted treatment-recommendations in all 14 cases. Genetic alterations were classified by treatment-baskets proposed by Horak et al. Hereby, we identified tyrosine-kinases (TK; n=4), PI3K-MTOR-AKT-pathway (PAM; n=3), cell-cycle-alterations (CC; n=2), RAF-MEK-ERK-cascade (RME; n=2), immune-evasion (IE; n=2), B-cell-targets (BCT; n=25) and others (OTH; n=4) for targeted treatment-recommendations. The minimum requirement for consideration of a drug within the scope of the study was FDA-fast-track development. Discussion The presented proof-of-concept study demonstrates the clinical potential of precision-oncology, even in prPBL-patients. Due to the aggressive course of the disease, there is an urgent medical-need for personalized treatment approaches, and this population should be considered for MTB inclusion at the earliest time.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany.,Department of Hematology and Oncology, Federal Armed Forces Hospital, Ulm, Germany
| | - Anke Fähnrich
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Jörg Riedl
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany.,Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Stephanie M J Fliedner
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Niklas Reimer
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Nadine Hertel
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Veronica Bernard
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Hartmut Merz
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| | - Alfred Feller
- Hämatopathologie Lübeck, Reference Centre for Lymph Node Pathology and Hematopathology, Lübeck, Germany
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany.,University Cancer Center Schleswig-Holstein, University Hospital of Schleswig- Holstein, Lübeck, Germany
| |
Collapse
|
38
|
Vodicka P, Klener P, Trneny M. Diffuse Large B-Cell Lymphoma (DLBCL): Early Patient Management and Emerging Treatment Options. Onco Targets Ther 2022; 15:1481-1501. [PMID: 36510607 PMCID: PMC9739046 DOI: 10.2147/ott.s326632] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents a curable disease with a 60-70% chance of cure with current R-CHOP chemoimmunotherapy. However, 30-40% of patients are refractory or relapsing. Many attempts failed to improve the outcome of DLBCL patients, including the intensification of R-CHOP regimen, consolidation, or maintenance therapy since the introduction of R-CHOP in 2000. Better understanding of both molecular biology of lymphoma cells and the tumor microenvironment raised the hope for future improvement of DLBCL patients' survival. Novel molecular findings have initiated clinical trials exploring targeted therapy based on driver genetic alterations with an intent to improve survival of high-risk subsets of patients. But the preliminary results remain ambiguous. The approach "agnostic" to specific molecular alterations of lymphoma cell includes antibody-drug conjugates (especially polatuzumab vedotin), immunotherapy comprising different antibodies with immunomodulatory effect (tafasitamab, lenalidomide), and T-cell engaging therapy (bispecific antibodies, early use of CAR T-cell). This approach could increase the cure rates and change the current therapeutic paradigm. However, better prognostic stratification, smarter designs of clinical trials, modification of endpoints including the use of ctDNA are needed. This review covers the complexity of DLBCL management.
Collapse
Affiliation(s)
- Prokop Vodicka
- First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Pavel Klener
- First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Marek Trneny
- First Department of Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
39
|
Seifert R, Kersting D, Rischpler C, Sandach P, Ferdinandus J, Fendler WP, Rahbar K, Weckesser M, Umutlu L, Hanoun C, Hüttmann A, Reinhardt HC, von Tresckow B, Herrmann K, Dührsen U, Schäfers M. Interim FDG-PET analysis to identify patients with aggressive non-Hodgkin lymphoma who benefit from treatment intensification: a post-hoc analysis of the PETAL trial. Leukemia 2022; 36:2845-2852. [PMID: 36241697 PMCID: PMC9712103 DOI: 10.1038/s41375-022-01713-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022]
Abstract
The randomized PETAL trial failed to demonstrate a benefit of interim FDG-PET (iPET)-based treatment intensification over continued standard therapy with CHOP (plus rituximab (R) in CD20-positive lymphomas). We hypothesized that PET analysis of all lymphoma manifestations may identify patients who benefitted from treatment intensification. A previously developed neural network was employed for iPET analysis to identify the highest pathological FDG uptake (max-SUVAI) and the mean FDG uptake of all lymphoma manifestations (mean-SUVAI). High mean-SUVAI uptake was determined separately for iPET-positive and iPET-negative patients. The endpoint was time-to-progression (TTP). There was a significant interaction of additional rituximab and mean-SUVAI in the iPET-negative group (HR = 0.6, p < 0.05). Patients with high mean-SUVAI had significantly prolonged TTP when treated with 6xR-CHOP + 2 R (not reached versus 52 months, p < 0.05), whereas max-SUVmanual failed to show an impact of additional rituximab. In the iPET-positive group, patients with high mean-SUVAI had a significantly longer TTP with (R-)CHOP than with the Burkitt protocol (14 versus 4 months, p < 0.01). Comprehensive iPET evaluation may provide new prognosticators in aggressive lymphoma. Additional application of rituximab was associated with prolonged TTP in iPET-negative patients with high mean-SUVAI. Comprehensive iPET interpretation could identify high-risk patients who benefit from study-specific interventions.
Collapse
Affiliation(s)
- Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany.
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany.
- West German Cancer Center, University Hospital Essen, Essen, Germany.
| | - David Kersting
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Patrick Sandach
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Justin Ferdinandus
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Matthias Weckesser
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Lale Umutlu
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Christine Hanoun
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andreas Hüttmann
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bastian von Tresckow
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Ulrich Dührsen
- German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen, Essen, Germany
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Schäfers
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
40
|
Salem AE, Shah HR, Covington MF, Koppula BR, Fine GC, Wiggins RH, Hoffman JM, Morton KA. PET-CT in Clinical Adult Oncology: I. Hematologic Malignancies. Cancers (Basel) 2022; 14:cancers14235941. [PMID: 36497423 PMCID: PMC9738711 DOI: 10.3390/cancers14235941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/28/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
PET-CT is an advanced imaging modality with many oncologic applications, including staging, assessment of response to therapy, restaging and evaluation of suspected recurrence. The goal of this 6-part series of review articles is to provide practical information to providers and imaging professionals regarding the best use of PET-CT for the more common adult malignancies. In the first article of this series, hematologic malignancies are addressed. The classification of these malignancies will be outlined, with the disclaimer that the classification of lymphomas is constantly evolving. Critical applications, potential pitfalls, and nuances of PET-CT imaging in hematologic malignancies and imaging features of the major categories of these tumors are addressed. Issues of clinical importance that must be reported by the imaging professionals are outlined. The focus of this article is on [18F] fluorodeoxyglucose (FDG), rather that research tracers or those requiring a local cyclotron. This information will serve as a resource for the appropriate role and limitations of PET-CT in the clinical management of patients with hematological malignancy for health care professionals caring for adult patients with hematologic malignancies. It also serves as a practical guide for imaging providers, including radiologists, nuclear medicine physicians and their trainees.
Collapse
Affiliation(s)
- Ahmed Ebada Salem
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
- Department of Radiodiagnosis and Intervention, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Harsh R. Shah
- Department of Medicine, Division of Hematology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84132, USA
| | - Matthew F. Covington
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Bhasker R. Koppula
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Gabriel C. Fine
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Richard H. Wiggins
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - John M. Hoffman
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Kathryn A. Morton
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, UT 84132, USA
- Intermountain Healthcare Hospitals, Murray, UT 84123, USA
- Correspondence: ; Tel.: +1-1801-581-7553
| |
Collapse
|
41
|
Baseline radiomics features and MYC rearrangement status predict progression in aggressive B-cell lymphoma. Blood Adv 2022; 7:214-223. [PMID: 36306337 PMCID: PMC9841040 DOI: 10.1182/bloodadvances.2022008629] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 01/21/2023] Open
Abstract
We investigated whether the outcome prediction of patients with aggressive B-cell lymphoma can be improved by combining clinical, molecular genotype, and radiomics features. MYC, BCL2, and BCL6 rearrangements were assessed using fluorescence in situ hybridization. Seventeen radiomics features were extracted from the baseline positron emission tomography-computed tomography of 323 patients, which included maximum standardized uptake value (SUVmax), SUVpeak, SUVmean, metabolic tumor volume (MTV), total lesion glycolysis, and 12 dissemination features pertaining to distance, differences in uptake and volume between lesions, respectively. Logistic regression with backward feature selection was used to predict progression after 2 years. The predictive value of (1) International Prognostic Index (IPI); (2) IPI plus MYC; (3) IPI, MYC, and MTV; (4) radiomics; and (5) MYC plus radiomics models were tested using the cross-validated area under the curve (CV-AUC) and positive predictive values (PPVs). IPI yielded a CV-AUC of 0.65 ± 0.07 with a PPV of 29.6%. The IPI plus MYC model yielded a CV-AUC of 0.68 ± 0.08. IPI, MYC, and MTV yielded a CV-AUC of 0.74 ± 0.08. The highest model performance of the radiomics model was observed for MTV combined with the maximum distance between the largest lesion and another lesion, the maximum difference in SUVpeak between 2 lesions, and the sum of distances between all lesions, yielding an improved CV-AUC of 0.77 ± 0.07. The same radiomics features were retained when adding MYC (CV-AUC, 0.77 ± 0.07). PPV was highest for the MYC plus radiomics model (50.0%) and increased by 20% compared with the IPI (29.6%). Adding radiomics features improved model performance and PPV and can, therefore, aid in identifying poor prognosis patients.
Collapse
|
42
|
Cabrera ME, Peña C, Leon P, Lois V, Rojas H, Vega V, Pizarro A, Calderon S, Rojas C, Aspillaga A, Gonzalez ML, Intriago M, Rojas B, Hales C, Oliva J, Romero M, Capurro M, Castillo JJ. Diffuse Large B-Cell Lymphoma in Chile: The Impact of Combined CHOP Plus Rituximab in the Public Health System. JCO Glob Oncol 2022; 8:e2200165. [PMID: 36351213 PMCID: PMC10166504 DOI: 10.1200/go.22.00165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
PURPOSE Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype. The purpose of this study was to evaluate the clinical features, prognostic factors, and results of DLBCL that was treated in the cancer centers of the public health system in Chile and compare cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) with rituximab with cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). PATIENTS AND METHODS Patients age > 15 years who were treated in 18 cancer centers in the country between 2001 and 2017 were included. The Kaplan-Meier method was used to calculate overall survival (OS), and Cox proportional hazard regression modeling was used to evaluate the effect of the addition of rituximab to CHOP on OS. RESULTS A total of 1,807 patients were evaluated. The median age at diagnosis was 62 (range, 15-95) years, with a female predominance (53%). Half of the patients were age ≥ 60 years. Serology for HIV infection was positive in 5% of cases (96 cases). International Prognostic Index scores were available for 90% of patients, of which 45% had low-risk, 25% low-intermediate-risk, 18% high-intermediate-risk, and 11% high-risk scores. CHOP was administered to 986 patients (55%; median follow-up, 13.2 years) and R-CHOP to 821 patients (45%; median follow-up, 8.4 years). R-CHOP was associated with superior OS compared with CHOP (5-year 66% v 48%, and 10-year 53% v 35%; P < .001). CONCLUSION Rituximab improved the survival of patients with DLBCL diagnosed and treated in Chile. The benefit was sustained over time, with curative rates of > 50%. This intervention shows that the inclusion of this biological drug justified the expenses incurred by the Ministry of Health in the National Lymphoma Protocols in Chile.
Collapse
Affiliation(s)
- María Elena Cabrera
- Hospital del Salvador, Titular Professor of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Pilar Leon
- Hospital Carlos van Buren, Valparaiso, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jorge J Castillo
- Division of Hematological Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
Choi JH, Lim I, Byun BH, Kim BI, Choi CW, Kang HJ, Shin DY, Lim SM. The role of 18F-FDG PET/CT in patients with diffuse large B-cell lymphoma after radioimmunotherapy using 131I-rituximab as consolidation therapy. PLoS One 2022; 17:e0273839. [PMID: 36156599 PMCID: PMC9512194 DOI: 10.1371/journal.pone.0273839] [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: 05/16/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the prognostic value of pretreatment 18F-FDG PET/CT after consolidation therapy of 131I-rituximab in patients with diffuse large B-cell lymphoma (DLBCL) who had acquired complete remission after receiving chemotherapy. Methods Patients who were diagnosed with DLBCL via histologic confirmation were retrospectively reviewed. All patients had achieved complete remission after 6 to 8 cycles of R-CHOP (rituximab, cyclophosphamide, vincristine, doxorubicin, and prednisolone) chemotherapy after which they underwent consolidation treatment with 131I-rituximab. 18F-FDG PET/CT scans were performed before R-CHOP for initial staging. The largest diameter of tumor, maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were obtained from pretreatment 18F-FDG PET/CT scans. Receiver-operating characteristic curves analysis was introduced for assessing the optimal criteria. Kaplan-Meier curve survival analysis was performed to evaluate both relapse free survival (RFS) and overall survival (OS). Results A total of 15 patients (12 males and 3 females) with a mean age of 56 (range, 30–73) years were enrolled. The median follow-up period of these patients was 73 months (range, 11–108 months). Four (27%) patients relapsed. Of them, three died during follow-up. Median values of the largest tumor size, highest SUVmax, MTV, and TLG were 5.3 cm (range, 2.0–16.4 cm), 20.2 (range, 11.1–67.4), 231.51 (range, 15–38.34), and 1277.95 (range, 238.37–10341.04), respectively. Patients with SUVmax less than or equal to 16.9 showed significantly worse RFS than patients with SUVmax greater than 16.9 (5-year RFS rate: 60% vs. 100%, p = 0.008). Patients with SUVmax less than or equal to 16.9 showed significantly worse OS than patients with SUVmax greater than 16.9 (5-year OS rate: 80% vs. 100% p = 0.042). Conclusion Higher SUVmax at pretreatment 18F-FDG PET/CT was associated with better relapse free survival and overall survival in DLBCL patients after consolidation therapy with 131I-rituximab. However, because this study has a small number of patients, a phase 3 study with a larger number of patients is needed for clinical application in the future.
Collapse
Affiliation(s)
- Joon Ho Choi
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Ilhan Lim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
- Department of Radiological & Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Korea
- * E-mail: (IL); (HJK)
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Byung Il Kim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Chang Woon Choi
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
| | - Hye Jin Kang
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
- * E-mail: (IL); (HJK)
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Republic of Korea
| |
Collapse
|
44
|
A tumor volume and performance status model to predict outcome before treatment in diffuse large B-cell lymphoma. Blood Adv 2022; 6:5995-6004. [PMID: 36044385 PMCID: PMC9691911 DOI: 10.1182/bloodadvances.2021006923] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 08/22/2022] [Indexed: 12/14/2022] Open
Abstract
Aggressive large B-cell lymphoma (LBCL) has variable outcomes. Current prognostic tools use factors for risk stratification that inadequately identify patients at high risk of refractory disease or relapse before initial treatment. A model associating 2 risk factors, total metabolic tumor volume (TMTV) >220 cm3 (determined by fluorine-18 fluorodeoxyglucose positron emission tomography coupled with computed tomography) and performance status (PS) ≥2, identified as prognostic in 301 older patients in the REMARC trial (#NCT01122472), was validated in 2174 patients of all ages treated in 2 clinical trials, PETAL (Positron Emission Tomography-Guided Therapy of Aggressive Non-Hodgkin Lymphomas; N = 510) and GOYA (N = 1315), and in real-world clinics (N = 349) across Europe and the United States. Three risk categories, low (no factors), intermediate (1 risk factor), and high (2 risk factors), significantly discriminated outcome in most of the series. Patients with 2 risk factors had worse outcomes than patients with no risk factors in the PETAL, GOYA, and real-world series. Patients with intermediate risk also had significantly worse outcomes than patients with no risk factors. The TMTV/Eastern Cooperative Oncology Group-PS combination outperformed the International Prognostic Index with a positive C-index for progression-free survival and overall survival in most series. The combination of high TMTV > 220 cm3 and ECOG-PS ≥ 2 is a simple clinical model to identify aggressive LBCL risk categories before treatment. This combination addresses the unmet need to better predict before treatment initiation for aggressive LBCL the patients likely to benefit the most or not at all from therapy.
Collapse
|
45
|
Lauer EM, Mutter J, Scherer F. Circulating tumor DNA in B-cell lymphoma: technical advances, clinical applications, and perspectives for translational research. Leukemia 2022; 36:2151-2164. [PMID: 35701522 PMCID: PMC9417989 DOI: 10.1038/s41375-022-01618-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/22/2022]
Abstract
Noninvasive disease monitoring and risk stratification by circulating tumor DNA (ctDNA) profiling has become a potential novel strategy for patient management in B-cell lymphoma. Emerging innovative therapeutic options and an unprecedented growth in our understanding of biological and molecular factors underlying lymphoma heterogeneity have fundamentally increased the need for precision-based tools facilitating personalized and accurate disease profiling and quantification. By capturing the entire mutational landscape of tumors, ctDNA assessment has some decisive advantages over conventional tissue biopsies, which usually target only one single tumor site. Due to its non- or minimal-invasive nature, serial and repeated ctDNA profiling provides a real-time picture of the genetic composition and facilitates quantification of tumor burden any time during the course of the disease. In this review, we present a comprehensive overview of technologies used for ctDNA detection and genotyping in B-cell lymphoma, focusing on pre-analytical and technical requirements, the advantages and limitations of various approaches, and highlight recent advances around improving sensitivity and suppressing technical errors. We broadly review potential applications of ctDNA in clinical practice and for translational research by describing how ctDNA might enhance lymphoma subtype classification, treatment response assessment, outcome prediction, and monitoring of measurable residual disease. We finally discuss how ctDNA could be implemented in prospective clinical trials as a novel surrogate endpoint and be utilized as a decision-making tool to guide lymphoma treatment in the future.
Collapse
Affiliation(s)
- Eliza M Lauer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jurik Mutter
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Florian Scherer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK) partner site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
46
|
Kiamanesh Z, Ayati N, Sadeghi R, Hawkes E, Lee ST, Scott AM. The value of FDG PET/CT imaging in outcome prediction and response assessment of lymphoma patients treated with immunotherapy: a meta-analysis and systematic review. Eur J Nucl Med Mol Imaging 2022; 49:4661-4676. [PMID: 35932329 DOI: 10.1007/s00259-022-05918-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/16/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Treatment strategies of lymphoid malignancies have been revolutionized by immunotherapy. Because of the inherent property of Hodgkin lymphoma and some subtypes of non-Hodgkin lymphoma as a highly FDG-avid tumor, functional 18F-FDG PET/CT imaging is already embedded in their routine care. Nevertheless, the question is whether it is still valuable in the context of these tumors being treated with immunotherapy. Herein, we will review the value of 18F-FDG PET/CT imaging lymphoid tumors treated with immunotherapy regimens. METHODS A comprehensive literature search of the PubMed database was conducted on the value of the 18F-FDG PET/CT for immunotherapy response monitoring of patients with malignant lymphoma. The articles were considered eligible if they met all of the following inclusion criteria: (a) clinical studies on patients with different types of malignant lymphoma, (b) treatment with anti-CD20 antibodies, immune checkpoint inhibitors or immune cell therapies, (c) and incorporated PET/CT with 18F-FDG as the PET tracer. RESULTS From the initial 1488 papers identified, 91 were ultimately included in our study. In anti-CD20 therapy, the highest pooled hazard ratios (HRs) of baseline, early, and late response monitoring parameters for progression-free survival (PFS) belong to metabolic tumor volume (MTV) (3.19 (95%CI: 2.36-4.30)), maximum standardized uptake value (SUVmax) (3.25 (95%CI: 2.08-5.08)), and Deauville score (DS) (3.73 (95%CI: 2.50-5.56)), respectively. These measurements for overall survival (OS) were MTV (4.39 (95%CI: 2.71-7.08)), DS (3.23 (95%CI: 1.87-5.58)), and DS (3.64 (95%CI: 1.40-9.43)), respectively. Early and late 18F-FDG PET/CT response assessment in immune checkpoint inhibitors (ICI) and immune cell therapy might be an effective tool for prediction of clinical outcome. CONCLUSION For anti-CD20 therapy of lymphoma, the MTV as a baseline 18F-FDG PET/CT-derived parameter has the highest HRs for PFS and OS. The DS as visual criteria in early and late response assessment has higher HRs for PFS and OS compared to the international harmonization project (IHP) visual criteria in anti-CD20 therapy. Early changes in 18F-FDG PET parameters may be predictive of response to ICIs and cell therapy in lymphoma patients.
Collapse
Affiliation(s)
- Zahra Kiamanesh
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narjess Ayati
- Department of Nuclear Medicine, Ultrasound & PET, Sydney Westmead Hospital, Sydney, NSW, Australia.,Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Victoria, Australia
| | - Ramin Sadeghi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Eliza Hawkes
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia.,Department of Medical Oncology & Clinical Haematology, Austin Health, Heidelberg, VIC, Australia.,School of Public Health & Preventative Medicine, Monash University, Melbourne, Australia
| | - Sze Ting Lee
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Victoria, Australia.,Department of Medicine, University of Melbourne, Victoria, Australia.,Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Road, Heidelberg, VIC, 3084, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Victoria, Australia. .,Department of Medicine, University of Melbourne, Victoria, Australia. .,Department of Molecular Imaging & Therapy, Austin Health, 145 Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
47
|
Mikhaeel NG, Heymans MW, Eertink JJ, de Vet HC, Boellaard R, Dührsen U, Ceriani L, Schmitz C, Wiegers SE, Hüttmann A, Lugtenburg PJ, Zucca E, Zwezerijnen GJ, Hoekstra OS, Zijlstra JM, Barrington SF. Proposed New Dynamic Prognostic Index for Diffuse Large B-Cell Lymphoma: International Metabolic Prognostic Index. J Clin Oncol 2022; 40:2352-2360. [PMID: 35357901 PMCID: PMC9287279 DOI: 10.1200/jco.21.02063] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/23/2022] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Baseline metabolic tumor volume (MTV) is a promising biomarker in diffuse large B-cell lymphoma (DLBCL). Our aims were to determine the best statistical relationship between MTV and survival and to compare MTV with the International Prognostic Index (IPI) and its individual components to derive the best prognostic model. METHODS PET scans and clinical data were included from five published studies in newly diagnosed diffuse large B-cell lymphoma. Transformations of MTV were compared with the primary end points of 3-year progression-free survival (PFS) and overall survival (OS) to derive the best relationship for further analyses. MTV was compared with IPI categories and individual components to derive the best model. Patients were grouped into three groups for survival analysis using Kaplan-Meier analysis; 10% at highest risk, 30% intermediate risk, and 60% lowest risk, corresponding with expected clinical outcome. Validation of the best model was performed using four studies as a test set and the fifth study for validation and repeated five times. RESULTS The best relationship for MTV and survival was a linear spline model with one knot located at the median MTV value of 307.9 cm3. MTV was a better predictor than IPI for PFS and OS. The best model combined MTV with age as continuous variables and individual stage as I-IV. The MTV-age-stage model performed better than IPI and was also better at defining a high-risk group (3-year PFS 46.3% v 58.0% and 3-year OS 51.5% v 66.4% for the new model and IPI, respectively). A regression formula was derived to estimate individual patient survival probabilities. CONCLUSION A new prognostic index is proposed using MTV, age, and stage, which outperforms IPI and enables individualized estimates of patient outcome.
Collapse
Affiliation(s)
- N. George Mikhaeel
- Department of Clinical Oncology, Guy's Cancer Centre and School of Cancer and Pharmaceutical Sciences, King's College London University, London, United Kingdom
| | - Martijn W. Heymans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Jakoba J. Eertink
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Henrica C.W. de Vet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Ronald Boellaard
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ulrich Dührsen
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Luca Ceriani
- Department of Oncology, IOSI—Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK—Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Christine Schmitz
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sanne E. Wiegers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Andreas Hüttmann
- Department of Hematology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Pieternella J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Emanuele Zucca
- Department of Oncology, IOSI—Oncology Institute of Southern Switzerland, Bellinzona; Università della Svizzera Italiana, Bellinzona, Switzerland
- SAKK—Swiss Group for Clinical Cancer Research, Bern, Switzerland
| | - Gerben J.C. Zwezerijnen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Otto S. Hoekstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Josée M. Zijlstra
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sally F. Barrington
- King's College London and Guy's and St Thomas' PET Centre, School of Biomedical Engineering and Imaging Sciences, King's Health Partners, Kings College London, London, United Kingdom
| |
Collapse
|
48
|
Hamid MS, Rutherford SC, Jang H, Kim S, Patel K, Bartlett NL, Malecek MK, Watkins MP, Maddocks KJ, Bond DA, Feldman TA, Magarelli G, Advani RH, Spinner MA, Evens AM, Shah M, Ahmed S, Stephens DM, Allen P, Tees MT, Karmali R, Cheson BD, Yazdy MS, Strouse C, Bailey NA, Pagel JM, Ramchandren R. Outcomes Among Classical Hodgkin Lymphoma Patients After an Interim PET Scan: A Real-World Experience. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e435-e442. [PMID: 35093285 DOI: 10.1016/j.clml.2021.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/17/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION The utility of dose escalation after positive positron emission tomography following 2 cycles of ABVD (PET2) for Hodgkin Lymphoma (HL) remains controversial. We describe the United States real-world practice patterns for PET2 positive patients. PATIENTS AND METHODS Data was collected from 15 sites on PET2 positive HL patients after receiving frontline treatment between January, 2015 and June, 2019. Descriptive analyses between those with therapy change and those continuing initial therapy were assessed. RESULTS A total of 129 patients were identified; 111 (86%) were treated with ABVD therapy and 18 (14%) with an alternate regimen. At PET2 assessment, 74.4% (96/129) had Deauville score (DS) 4 and 25.6% (33/129) had DS 5. Of the 66 limited stage (LS) patients with PET2 DS score of 4/5, 77.3% (51/66) continued initial therapy and 22.7% (15/66) changed to escalated therapy. The 12-month progression-free survival (PFS) for DS 4/5 LS patients was 67.0% (95% CI; 54.9-81.7) for patients without escalation compared with 51.4% (95% CI; 30.8-85.8) for those who escalated. Of the 63 DS 4/5 patients with advanced stage (AS) disease, 76.2% (48/63) continued initial therapy and 23.8% (15/63) changed to escalated therapy. The 12-month PFS for DS 4/5 AS patients was 38.3% (95% CI: 26.3%-55.7%) for patients without escalation compared with 57.1% (95% CI: 36.3-89.9) for those with escalation. CONCLUSION A minority of PET2 positive HL patients undergo therapy escalation and outcomes remain overall suboptimal. Improved prognostics markers and better therapeutics are required to improve outcomes for high-risk PET2 positive HL patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Nancy L Bartlett
- Washington University Medical University at St. Louis, St. Louis, MO
| | - Mary-Kate Malecek
- Washington University Medical University at St. Louis, St. Louis, MO
| | - Marcus P Watkins
- Washington University Medical University at St. Louis, St. Louis, MO
| | - Kami J Maddocks
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - David A Bond
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Tatyana A Feldman
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ
| | - Gabriela Magarelli
- John Theurer Cancer Center at Hackensack Meridian Health, Hackensack, NJ
| | | | | | - Andrew M Evens
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Mansi Shah
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | | | | | - Pamela Allen
- Winship Cancer Institute at Emory University, Atlanta, GA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Burggraaff CN, Eertink JJ, Lugtenburg PJ, Hoekstra OS, Arens AI, de Keizer B, Heymans MW, van der Holt B, Wiegers SE, Pieplenbosch S, Boellaard R, de Vet HC, Zijlstra JM. 18F-FDG PET Improves Baseline Clinical Predictors of Response in Diffuse Large B-Cell Lymphoma: The HOVON-84 Study. J Nucl Med 2022; 63:1001-1007. [PMID: 34675112 PMCID: PMC9258573 DOI: 10.2967/jnumed.121.262205] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 09/29/2021] [Indexed: 01/03/2023] Open
Abstract
We aimed to determine the added value of baseline metabolic tumor volume (MTV) and interim PET (I-PET) to the age-adjusted international prognostic index (aaIPI) to predict 2-y progression-free survival (PFS) in diffuse large B-cell lymphoma. Secondary objectives were to investigate optimal I-PET response criteria (using Deauville score [DS] or quantitative change in SUVmax [ΔSUVmax] between baseline and I-PET4 [observational I-PET scans after 4 cycles of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone administered in 2-wk intervals with intensified rituximab in the first 4 cycles [R(R)-CHOP14]). Methods: I-PET4 scans in the HOVON-84 (Hemato-Oncologie voor Volwassenen Nederland [Haemato Oncology Foundation for Adults in the Netherlands]) randomized clinical trial (EudraCT 2006-005174-42) were centrally reviewed using DS (cutoff, 4-5). Additionally, ΔSUVmax (prespecified cutoff, 70%) and baseline MTV were measured. Multivariable hazard ratio (HR), positive predictive value (PPV), and negative predictive value (NPV) were obtained for 2-y PFS. Results: In total, 513 I-PET4 scans were reviewed according to DS, and ΔSUVmax and baseline MTV were available for 367 and 296 patients. The NPV of I-PET ranged between 82% and 86% for all PET response criteria. Univariate HR and PPV were better for ΔSUVmax (4.8% and 53%, respectively) than for DS (3.1% and 38%, respectively). aaIPI and ΔSUVmax independently predicted 2-y PFS (HR, 3.2 and 5.0, respectively); adding MTV brought about a slight improvement. Low or low-intermediate aaIPI combined with a ΔSUVmax of more than 70% (37% of patients) yielded an NPV of 93%, and the combination of high-intermediate or high aaIPI and a ΔSUVmax of 70% or less yielded a PPV of 65%. Conclusion: In this study on diffuse large B-cell lymphoma, I-PET after 4 cycles of R(R)-CHOP14 added predictive value to aaIPI for 2-y PFS, and both were independent response biomarkers in a multivariable Cox model. We externally validated that ΔSUVmax outperformed DS in 2-y PFS prediction.
Collapse
Affiliation(s)
- Coreline N. Burggraaff
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jakoba J. Eertink
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieternella J. Lugtenburg
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Otto S. Hoekstra
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anne I.J. Arens
- Department of Radiology, Nuclear Medicine, and Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bart de Keizer
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martijn W. Heymans
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; and
| | - Bronno van der Holt
- Department of Hematology, HOVON Data Center, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sanne E. Wiegers
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Simone Pieplenbosch
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Henrica C.W. de Vet
- Department of Epidemiology and Data Science, Amsterdam UMC, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; and
| | - Josée M. Zijlstra
- Department of Hematology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
50
|
Ritter Z, Papp L, Zámbó K, Tóth Z, Dezső D, Veres DS, Máthé D, Budán F, Karádi É, Balikó A, Pajor L, Szomor Á, Schmidt E, Alizadeh H. Two-Year Event-Free Survival Prediction in DLBCL Patients Based on In Vivo Radiomics and Clinical Parameters. Front Oncol 2022; 12:820136. [PMID: 35756658 PMCID: PMC9216187 DOI: 10.3389/fonc.2022.820136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose For the identification of high-risk patients in diffuse large B-cell lymphoma (DLBCL), we investigated the prognostic significance of in vivo radiomics derived from baseline [18F]FDG PET/CT and clinical parameters. Methods Pre-treatment [18F]FDG PET/CT scans of 85 patients diagnosed with DLBCL were assessed. The scans were carried out in two clinical centers. Two-year event-free survival (EFS) was defined. After delineation of lymphoma lesions, conventional PET parameters and in vivo radiomics were extracted. For 2-year EFS prognosis assessment, the Center 1 dataset was utilized as the training set and underwent automated machine learning analysis. The dataset of Center 2 was utilized as an independent test set to validate the established predictive model built by the dataset of Center 1. Results The automated machine learning analysis of the Center 1 dataset revealed that the most important features for building 2-year EFS are as follows: max diameter, neighbor gray tone difference matrix (NGTDM) busyness, total lesion glycolysis, total metabolic tumor volume, and NGTDM coarseness. The predictive model built on the Center 1 dataset yielded 79% sensitivity, 83% specificity, 69% positive predictive value, 89% negative predictive value, and 0.85 AUC by evaluating the Center 2 dataset. Conclusion Based on our dual-center retrospective analysis, predicting 2-year EFS built on imaging features is feasible by utilizing high-performance automated machine learning.
Collapse
Affiliation(s)
- Zsombor Ritter
- Department of Medical Imaging, Medical School, University of Pécs, Pécs, Hungary
| | - László Papp
- Medical University of Vienna, Center for Medical Physics and Biomedical Engineering, Vienna, Austria
| | - Katalin Zámbó
- Department of Medical Imaging, Medical School, University of Pécs, Pécs, Hungary
| | - Zoltán Tóth
- University of Kaposvár, PET Medicopus Nonprofit Ltd., Kaposvár, Hungary
| | - Dániel Dezső
- Department of Medical Imaging, Medical School, University of Pécs, Pécs, Hungary
| | - Dániel Sándor Veres
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.,In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Budapest, Hungary
| | - Ferenc Budán
- Institute of Transdisciplinary Discoveries, Medical School, University of Pécs, Pécs, Hungary.,Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Éva Karádi
- Department of Hematology, University of Kaposvár, Kaposvár, Hungary
| | - Anett Balikó
- County Hospital Tolna, János Balassa Hospital, Szekszárd, Hungary
| | - László Pajor
- Department of Pathology, Medical School, University of Pécs, Pécs, Hungary
| | - Árpád Szomor
- 1st Department of Internal Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Erzsébet Schmidt
- Department of Medical Imaging, Medical School, University of Pécs, Pécs, Hungary
| | - Hussain Alizadeh
- 1st Department of Internal Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|