1
|
Hamova I, Maco M, Tkachenko A, Kupcova K, Velasova A, Trneny M, Mocikova H, Havranek O. Circulating tumor DNA as a powerful tool in diagnostics and treatment outcome prediction - focus on large B-cell lymphomas and follicular lymphomas. Expert Rev Mol Diagn 2025. [PMID: 40326242 DOI: 10.1080/14737159.2025.2500659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION Pathogenesis of large B-cell lymphomas (LBCL) and follicular lymphomas (FL) is a multistep process associated with development of diverse DNA alterations and consequent deregulation of critical cellular processes. Detection of tumor-associated mutations within non-tumor compartments (mainly plasma) is the basis of the 'liquid biopsy' concept. Apart from tumor mutational profiling, quantitative analysis of circulating tumor DNA (ctDNA) allows longitudinal assessment of tumor burden. ctDNA-based technologies provide a new tool for tumor diagnostics and treatment personalization. AREAS COVERED Our review provides a comprehensive overview and summary of available ctDNA studies in LBCL and FL. The accuracy of ctDNA based detection of lymphoma associated DNA alterations is correlated to the known LBCL and FL molecular landscape. Additionally, we summarized available evidence that supports and justifies the clinical use of ctDNA for lymphoma risk stratification, treatment response evaluation, and treatment response-adapted therapy. Lastly, we discuss other clinically important ctDNA applications: monitoring of lymphoma clonal evolution within resistance and/or relapse development and utilization of ctDNA for diagnostics in non-blood fluids and compartments (e.g. cerebrospinal fluid in primary CNS lymphomas). EXPERT OPINION Despite certain challenges including methodological standardization, ctDNA holds promise to soon become an integral part of lymphoma diagnostics and treatment management.
Collapse
Affiliation(s)
- Iva Hamova
- Biocev, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Maria Maco
- Department of Haematology, Fakultni nemocnice Kralovske Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Anton Tkachenko
- Biocev, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kristyna Kupcova
- Biocev, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Adriana Velasova
- Biocev, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marek Trneny
- First Department of Medicine - Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Heidi Mocikova
- Department of Haematology, Fakultni nemocnice Kralovske Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ondrej Havranek
- Biocev, First Faculty of Medicine, Charles University, Prague, Czech Republic
- First Department of Medicine - Hematology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
2
|
Alibrahim MN, Gloghini A, Carbone A. Classic Hodgkin lymphoma: Pathobiological features that impact emerging therapies. Blood Rev 2025; 71:101271. [PMID: 39904647 DOI: 10.1016/j.blre.2025.101271] [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: 11/28/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
Classic Hodgkin lymphoma (cHL) is defined by distinctive Hodgkin Reed-Sternberg (HRS) cells, which are CD30+/CD15+ multinucleated tumor cells lacking typical B-cell markers. These cells comprise <5 % of tumor mass but orchestrate an extensive immunosuppressive tumor microenvironment (TME). Classic HL was curable with radiation therapy and multi-agent chemotherapy. Despite high cure rates, treatment-related toxicities remain significant. The goals of multimodality therapy are to achieve a cure while minimizing treatment-associated toxicity. Advances in molecular insights into HRS cells have led to transformative therapies, including checkpoint inhibitors, antibody-drug conjugates like brentuximab vedotin, which have shown remarkable efficacy, especially in relapsed or refractory disease. However, challenges persist due to the heterogeneity of cHL, driven by the complex biology of HRS cells and their surrounding tumor microenvironment. Novel approaches such as single-cell RNA sequencing and circulating tumor DNA profiling provide promising strategies to address these challenges. This review examines the origin, morphology, phenotype, and genetic profiles of HRS cells, highlighting key pathobiological features, including biomarkers and Epstein-Barr Virus involvement. It also explores the biological mechanisms underlying HRS cell survival and evaluates standard and emerging therapies, offering insights into the rationale for novel treatment strategies.
Collapse
Affiliation(s)
| | - Annunziata Gloghini
- Department of Avanced Pathology, Fondazione IRCCS, Istituto Nazionale dei Tumori Milano, Italy.
| | - Antonino Carbone
- Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
3
|
Roschewski M, Longo DL, Armitage JO. Circulating Tumor DNA as Measurable Residual Disease in Aggressive B-Cell Lymphoma: A Narrative Review. JAMA Oncol 2025; 11:416-422. [PMID: 39913153 DOI: 10.1001/jamaoncol.2024.6144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Importance Achieving remission is the first step toward a cure in treating aggressive B-cell lymphomas. Radiographic imaging, such as fluorodeoxyglucose-positron emission tomography/computed tomography scans are the current standard to define remission at the end of therapy but lack specificity for lymphoma and cannot detect disease at the molecular level. Identifying measurable residual disease with ultrasensitive detection of circulating tumor DNA potentially offers the possibility of improving clinical outcomes. Observations Early studies of circulating tumor DNA in aggressive B-cell lymphomas showed a strong association with overall tumor burden, and baseline quantitative levels are associated with clinical outcomes after frontline chemotherapy. Next-generation sequencing methods that detect lymphoma-relevant genetic aberrations in circulating tumor DNA can also be used for noninvasive genotyping that strongly mirror tissue biopsies. Rapid changes in circulating tumor DNA dynamics after 1 or 2 cycles of frontline chemotherapy or within weeks of treatment with chimeric antigen receptor T-cell salvage therapy are also highly prognostic. Although serial monitoring of circulating tumor DNA can detect molecular relapse 3 to 6 months before clinical relapse, improved analytical thresholds are required to detect measurable residual disease at a singular point at the end of therapy. Modern advances in circulating tumor DNA methods now allow for the reliable detection of measurable residual disease, with an analytical detection threshold of 1 in 1 million cell-free DNA molecules. Conclusions and Relevance The results of this review suggest that modern ultrasensitive methods of detecting circulating tumor DNA may improve the current definition of remission in aggressive B-cell lymphomas. Incorporating circulating tumor DNA at the end of therapy assessment identifies patients who do not require surveillance monitoring and introduces paradigms of treating measurable residual disease within clinical trials. Practical barriers, including standardization of collection, availability, turnaround times, and cost, remain hurdles preventing widespread implementation into clinical practice.
Collapse
Affiliation(s)
| | - Dan L Longo
- Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
4
|
Fu L, Zhou X, Zhang X, Li X, Zhang F, Gu H, Wang X. Circulating tumor DNA in lymphoma: technologies and applications. J Hematol Oncol 2025; 18:29. [PMID: 40069858 PMCID: PMC11900646 DOI: 10.1186/s13045-025-01673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Lymphoma, a malignant tumor derived from lymphocytes and lymphoid tissues, presents with complex and heterogeneous clinical manifestations, requiring accurate patient classification for appropriate treatment. While invasive pathological examination of lymph nodes or lymphoid tissue remains the gold standard for lymphoma diagnosis, its utility is limited in cases of deep-seated tumors such as intraperitoneal and central nervous system lymphomas. In addition, biopsy procedures carry an inherent risk of complications. Computed tomography (CT) and positron emission tomography/computed tomography (PET/CT) imaging are essential for treatment assessment and monitoring, but lack the ability to detect early clonal evolution and minimal residual disease (MRD). Liquid biopsy-based analysis of circulating tumor DNA (ctDNA) offers a non-invasive alternative that allows for repeated sampling and overcomes the limitations of spatial heterogeneity and invasive biopsies. ctDNA provides genetic and epigenetic insights into lymphoma and serves as a dynamic, quantifiable biomarker for diagnosis, risk stratification, and treatment response. This review comprehensively summarizes common genetic variations in lymphoma and systematically evaluates ctDNA detection technologies, including PCR-based assays and next-generation sequencing (NGS). Applications of ctDNA detection in noninvasive genotyping, risk stratification, therapeutic response monitoring, and MRD detection are discussed across various lymphoma subtypes, including diffuse large B-cell lymphoma, Hodgkin lymphoma, follicular lymphoma, and T-cell lymphoma. By integrating recent research findings, the review highlights the role of ctDNA profiling in advancing precision medicine, enabling personalized therapeutic strategies, and improving clinical outcomes in lymphoma.
Collapse
Affiliation(s)
- Lina Fu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
| | - Xuerong Zhou
- Department of Hematology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Xiaoyu Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong Province, 250012, Jinan, China
| | - Xuhua Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
| | - Fan Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, Anhui Province, China.
| | - Xiaoxue Wang
- Department of Hematology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
5
|
Cottrell TR, Lotze MT, Ali A, Bifulco CB, Capitini CM, Chow LQM, Cillo AR, Collyar D, Cope L, Deutsch JS, Dubrovsky G, Gnjatic S, Goh D, Halabi S, Kohanbash G, Maecker HT, Maleki Vareki S, Mullin S, Seliger B, Taube J, Vos W, Yeong J, Anderson KG, Bruno TC, Chiuzan C, Diaz-Padilla I, Garrett-Mayer E, Glitza Oliva IC, Grandi P, Hill EG, Hobbs BP, Najjar YG, Pettit Nassi P, Simons VH, Subudhi SK, Sullivan RJ, Takimoto CH. Society for Immunotherapy of Cancer (SITC) consensus statement on essential biomarkers for immunotherapy clinical protocols. J Immunother Cancer 2025; 13:e010928. [PMID: 40054999 PMCID: PMC11891540 DOI: 10.1136/jitc-2024-010928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/05/2025] [Indexed: 03/12/2025] Open
Abstract
Immunotherapy of cancer is now an essential pillar of treatment for patients with many individual tumor types. Novel immune targets and technical advances are driving a rapid exploration of new treatment strategies incorporating immune agents in cancer clinical practice. Immunotherapies perturb a complex system of interactions among genomically unstable tumor cells, diverse cells within the tumor microenvironment including the systemic adaptive and innate immune cells. The drive to develop increasingly effective immunotherapy regimens is tempered by the risk of immune-related adverse events. Evidence-based biomarkers that measure the potential for therapeutic response and/or toxicity are critical to guide optimal patient care and contextualize the results of immunotherapy clinical trials. Responding to the lack of guidance on biomarker testing in early-phase immunotherapy clinical trials, we propose a definition and listing of essential biomarkers recommended for inclusion in all such protocols. These recommendations are based on consensus provided by the Society for Immunotherapy of Cancer (SITC) Clinical Immuno-Oncology Network (SCION) faculty with input from the SITC Pathology and Biomarker Committees and the Journal for ImmunoTherapy of Cancer readership. A consensus-based selection of essential biomarkers was conducted using a Delphi survey of SCION faculty. Regular updates to these recommendations are planned. The inaugural list of essential biomarkers includes complete blood count with differential to generate a neutrophil-to-lymphocyte ratio or systemic immune-inflammation index, serum lactate dehydrogenase and albumin, programmed death-ligand 1 immunohistochemistry, microsatellite stability assessment, and tumor mutational burden. Inclusion of these biomarkers across early-phase immunotherapy clinical trials will capture variation among trials, provide deeper insight into the novel and established therapies, and support improved patient selection and stratification for later-phase clinical trials.
Collapse
Affiliation(s)
- Tricia R Cottrell
- Queen's University Sinclair Cancer Research Institute, Kingston, Ontario, Canada
| | | | - Alaa Ali
- Stem Cell Transplant and Cellular Immunotherapy Program, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, Washington, DC, USA
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Christian M Capitini
- University of Wisconsin School of Medicine and Public Health and Carbone Cancer Center, Madison, Wisconsin, USA
| | | | - Anthony R Cillo
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Deborah Collyar
- Patient Advocates In Research (PAIR), Danville, California, USA
| | - Leslie Cope
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Susan Halabi
- Duke School of Medicine and Duke Cancer Institute, Durham, North Carolina, USA
| | - Gary Kohanbash
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Holden T Maecker
- Stanford University School of Medicine, Stanford, California, USA
| | - Saman Maleki Vareki
- Department of Oncology and Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Sarah Mullin
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Barbara Seliger
- Campus Brandenburg an der Havel, Brandenburg Medical School, Halle, Germany
| | - Janis Taube
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wim Vos
- Radiomics.bio, Liège, Belgium
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Kristin G Anderson
- Department of Microbiology, Immunology and Cancer Biology, Department of Obstetrics and Gynecology, Beirne B. Carter Center for Immunology Research and the University of Virginia Comprehensive Cancer Center, University of Virginia, Charlottesville, Virginia, USA
| | - Tullia C Bruno
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tumor Microenvironment Center, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Codruta Chiuzan
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | | | | | | | | | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Brian P Hobbs
- Dell Medical School, The University of Texas, Austin, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Sumit K Subudhi
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Needham, Massachusetts, USA
| | | |
Collapse
|
6
|
Strati P, Agajanian R, Lossos IS, Coleman M, Kridel R, Wood A, Lesley R, Wun C, Stephens DM. Acalabrutinib in combination with rituximab and lenalidomide in patients with relapsed or refractory follicular lymphoma: Results of the phase 1b open-label study (ACE-LY-003). Br J Haematol 2025; 206:887-898. [PMID: 39667721 PMCID: PMC11886943 DOI: 10.1111/bjh.19951] [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: 08/22/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Patients with relapsed/refractory (R/R) follicular lymphoma (FL) have limited effective treatment options. Bruton tyrosine kinase inhibitors (BTKis) increase the anti-tumoural phenotype of tumour-associated macrophages, providing rationale to combine them with rituximab and lenalidomide (R2). Acalabrutinib, a second-generation BTKi, has potential to improve R2 efficacy without increasing T-cell-mediated toxicity due to its lack of interleukin-2-inducible T-cell kinase inhibition. Here, we report safety and efficacy from a phase 1b dose-finding study (NCT02180711) evaluating acalabrutinib plus R2 in patients with R/R FL. Overall, 29 patients received acalabrutinib plus R2 (lenalidomide 15 mg, n = 8; lenalidomide 20 mg, n = 21). At a median acalabrutinib exposure of 21 months, the most common grade ≥3 treatment-emergent adverse event (TEAE) was neutropenia (37.9%). The incidence of grade ≥3 serious TEAEs was 37.5% and 52.4% in the lenalidomide 15-mg and 20-mg cohorts, respectively; overall, the most common were COVID-19 pneumonia, COVID-19 infection and pneumonia. Earlier treatment withholdings/reductions were observed in the 20-mg cohort. With a median follow-up of 34.1 months, the overall response rate was 75.9%. The complete response rate was 25.0% and 42.9% in the lenalidomide 15- and 20-mg cohorts, respectively. Due to acceptable toxicity and preliminary efficacy, the lenalidomide 20-mg dose was selected for further investigation.
Collapse
Affiliation(s)
- Paolo Strati
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Richy Agajanian
- The Oncology Institute of Hope and InnovationDowneyCaliforniaUSA
| | - Izidore S. Lossos
- Sylvester Comprehensive Cancer CenterUniversity of Miami–Miller School of MedicineMiamiFloridaUSA
| | - Morton Coleman
- Clinical Research Alliance/Weill Cornell MedicineNew YorkNew YorkUSA
| | | | | | | | | | - Deborah M. Stephens
- University of North Carolina Lineberger Cancer CenterChapel HillNorth CarolinaUSA
| |
Collapse
|
7
|
Navrkalova V, Mareckova A, Porc J, Hricko S, Hrabcakova V, Kissova J, Kundova S, Jarosova M, Pospisilova S, Kotaskova J, Janikova A. Advanced NGS analysis of cell-free tumor DNA supports clonal relation to primary high-grade B-cell lymphoma lesion and CNS relapse despite MRI negativity. Diagn Pathol 2025; 20:14. [PMID: 39905397 PMCID: PMC11792325 DOI: 10.1186/s13000-025-01609-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
High-grade B-cell lymphomas (HGBCLs) are aggressive blood cancers with a severe disease course, especially when the central nervous system (CNS) is involved. Standard histological examination depends on tissue availability and is currently supplemented with molecular tests, as the status of MYC, BCL2, or BCL6 gene rearrangements is required for proper lymphoma classification. This case report demonstrates the relevance of cerebrospinal fluid (CSF) cell-free DNA testing by integrative next-generation sequencing (NGS) panel. The benefit of this approach resided in tumor genotyping alongside the proof of CNS progression despite MRI negativity, revealing a clonal relationship with the primary tumor lesion. In addition, our strategy allowed us to classify the tumor as DLBCL/HGBL-MYC/BCL2 entity. In clinical practice, such a minimally invasive approach provides a more sensitive tool than standard imaging and cell analyzing techniques, enabling more accurate disease monitoring and relapse prediction in particular cases.
Collapse
MESH Headings
- Humans
- High-Throughput Nucleotide Sequencing/methods
- Magnetic Resonance Imaging
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/cerebrospinal fluid
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/cerebrospinal fluid
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/cerebrospinal fluid
- Middle Aged
- Male
- Female
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Central Nervous System Neoplasms/genetics
- Central Nervous System Neoplasms/pathology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/pathology
Collapse
Affiliation(s)
- Veronika Navrkalova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
- Center of Molecular Medicine, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
- Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic.
| | - Andrea Mareckova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jakub Porc
- Center of Molecular Medicine, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Samuel Hricko
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Viera Hrabcakova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jarmila Kissova
- Department of Clinical Hematology, University Hospital Brno, Brno, Czech Republic
| | - Sona Kundova
- Clinic of Radiology and Nuclear Medicine, University Hospital Brno, Brno, Czech Republic
| | - Marie Jarosova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Center of Molecular Medicine, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Jana Kotaskova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Center of Molecular Medicine, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Medical Genetics and Genomics, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Andrea Janikova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
8
|
Bisig B, Lefort K, Carras S, de Leval L. Clinical use of circulating tumor DNA analysis in patients with lymphoma. Hum Pathol 2025; 156:105679. [PMID: 39491629 DOI: 10.1016/j.humpath.2024.105679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The analysis of circulating tumor DNA (ctDNA) in liquid biopsy specimens has an established role for the detection of predictive molecular alterations and acquired resistance mutations in several tumors. The low-invasiveness of this approach allows for repeated sampling and dynamic monitoring of disease evolution. Originating from the entire body tumor bulk, plasma-derived ctDNA reflects intra- and interlesional genetic heterogeneity. In the management of lymphoma patients, ctDNA quantification at various timepoints of the patient's clinical history is emerging as a complementary tool that may improve risk stratification, assessment of treatment response and early relapse detection during follow-up, most prominently in patients with diffuse large B-cell lymphoma or classic Hodgkin lymphoma. While liquid biopsies have not yet entered standard-of-care treatment protocols in these settings, several trials have provided evidence that at least a subset of lymphoma patients may benefit from the introduction of liquid biopsies into daily clinical care. In parallel, continuous technological developments have enabled highly sensitive ctDNA assessment methods, which span from locus-specific techniques identifying single hotspot mutations, to sequencing panels and genome-wide approaches that explore broader genetic and epigenetic alterations. Here, we provide an overview of current methods and ongoing technical developments for ctDNA evaluation. We also summarize the most important data from a selection of clinical studies that have explored the clinical use of ctDNA in several lymphoma entities.
Collapse
Affiliation(s)
- Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Karine Lefort
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Sylvain Carras
- Institute for Advanced Biosciences (INSERM U1209, CNRS UMR 5309, UGA), Department of Molecular Biology and Department of Oncohematology, University Hospital Grenoble and University Grenoble Alpes, Grenoble, France
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
9
|
Lei S, Jia S, Takalkar S, Chang TC, Ma X, Szlachta K, Xu K, Cheng Z, Hui Y, Koo SC, Mead PE, Gao Q, Kumar P, Bailey CP, Sunny J, Pappo AS, Federico SM, Robinson GW, Gajjar A, Rubnitz JE, Jeha S, Pui CH, Inaba H, Wu G, Klco JM, Tatevossian RG, Mullighan CG. Genomic profiling of circulating tumor DNA for childhood cancers. Leukemia 2025; 39:420-430. [PMID: 39523434 DOI: 10.1038/s41375-024-02461-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The utility of circulating tumor DNA (ctDNA) analysis has not been well-established for disease detection and monitoring of childhood cancers, especially leukemias. We developed PeCan-Seq, a deep sequencing method targeting diverse somatic genomic variants in cell-free samples in childhood cancer. Plasma samples were collected at diagnosis from 233 children with hematologic, solid and brain tumors. All children with hematologic malignancy (n = 177) had detectable ctDNA at diagnosis. The median ctDNA fraction was 0.77, and 97% of 789 expected tumor variants were identified, including sequence mutations, copy number variations, and structural variations responsible for oncogenic fusions. In contrast, ctDNA was detected in 19 of 38 solid tumor patients and 1 of 18 brain tumor patients. Somatic variants from ctDNA were correlated with minimal residual disease levels as determined by flow cytometry in serial plasma samples from patients with B-cell acute lymphoblastic leukemia (B-ALL). We showcase multi-tumor detection by ctDNA analysis for a patient with concurrent B-ALL and neuroblastoma. In conclusion, PeCan-seq sensitively identified heterogeneous ctDNA alterations from 1 mL plasma for childhood hematologic malignancies and a subset of solid tumors. PeCan-seq provides a robust, non-invasive approach to augment comprehensive genomic profiling at diagnosis and mutation-specific detection during disease monitoring.
Collapse
Affiliation(s)
- Shaohua Lei
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sujuan Jia
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sunitha Takalkar
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Karol Szlachta
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ke Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhongshan Cheng
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yawei Hui
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul E Mead
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Qingsong Gao
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Priyadarshini Kumar
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Colin P Bailey
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jobin Sunny
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alberto S Pappo
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Giles W Robinson
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Amar Gajjar
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gang Wu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Ruth G Tatevossian
- Clinical Biomarkers Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center of Excellence for Leukemia Studies, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
10
|
Witzig TE, Taylor WR, Mahoney DW, Bamlet WR, Foote PH, Burger KN, Doering KA, Devens ME, Arndt JR, O'Connell MC, Berger CK, Novak AJ, Cerhan JR, Hennek J, Katerov S, Allawi HT, Jevremovic D, Dao LN, Graham RP, Kisiel JB. Blood Plasma Methylated DNA Markers in the Detection of Lymphoma: Discovery, Validation, and Clinical Pilot. Am J Hematol 2025; 100:218-228. [PMID: 39564730 PMCID: PMC11705204 DOI: 10.1002/ajh.27533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024]
Abstract
Lymphoma is one of the leading causes of cancer and cancer deaths and yet has not been amenable to population screening. The role of methylated DNA markers (MDMs) in the detection of lymphoma has not been extensively studied. We aimed to discover, validate, and test tissue-derived MDMs of lymphoma in archival plasma specimens. Reduced representation bisulfite sequencing (RRBS) was performed on a discovery set of frozen tissues. MDMs identified were converted to methylation-specific PCR assays and validated on independent formalin-fixed, paraffin-embedded (FFPE) tissues. Target enrichment long-probe quantitative-amplified signal (TELQAS) assays were developed and assayed in plasma-extracted, bisulfite-converted DNA from independent treatment-naïve lymphoma patients and healthy controls. Prediction of cancer status was modeled using random forest model with in silico cross-validation. After discovery and validation in tissue, 16 TELQAS assays (ZNF503, VWA5B1, HOXA9, GABRG3, ITGA5, MAX.chr17.7190, BNC1, CDK20, MAX.chr4.4069, TPBG, DNAH14, SYT6, CACNG8, FAM110B, ADRA1D, and NRN1) were selected for testing in plasma. These detected 78% (95% CI, 74%-82%) of lymphoma cases at 90% specificity. Excluding marginal zone and T-cell lymphomas, sensitivity increased to 84% (80%-88%). MDMs in plasma show promise to detect lymphoma and are candidates for inclusion in multi-cancer detection studies.
Collapse
Affiliation(s)
| | - William R. Taylor
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Douglas W. Mahoney
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - William R. Bamlet
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Patrick H. Foote
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Kelli N. Burger
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Karen A. Doering
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Mary E. Devens
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Jacquelyn R. Arndt
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Maria C. O'Connell
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Calise K. Berger
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| | - Anne J. Novak
- Division of HematologyMayo ClinicRochesterMinnesotaUSA
| | - James R. Cerhan
- Department of Qualitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Slava Katerov
- Exact Sciences Development CompanyMadisonWisconsinUSA
| | | | | | - Linda N. Dao
- Division of Anatomic PathologyMayo ClinicRochesterMinnesotaUSA
| | | | - John B. Kisiel
- Division of Gastroenterology and HepatologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
11
|
Ohmoto A, Fuji S. Spontaneous regression in mature T-cell non-Hodgkin lymphoma. Expert Rev Hematol 2025; 18:47-55. [PMID: 39645531 DOI: 10.1080/17474086.2024.2439469] [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: 03/23/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Spontaneous regression (SR) is observed in some patients with mature T-cell non-Hodgkin lymphoma (MTCL), including adult T-cell leukemia/lymphoma (ATL), although the incidence is rare. AREA COVERED We extracted 31 cases with MTCL who experienced SR based on a literature search and summarized the patient characteristics and clinical outcomes. EXPERT OPINION MTCL with SR included various subtypes, the most common being ATL (n = 17). Five of 24 cases (21%) maintained SR for more than 5 years, and the median duration of SR was 2 years. Sixteen of 31 cases (52%) experienced tumor relapse after SR. Two retrospective studies including ATL cases showed SR rates of 18% and 4%, respectively. Representative triggers are infection and surgical biopsies, and possible mechanisms include immunological mechanisms such as cross-reactivity of virus-specific T cells with tumor antigens. The diagnostic criteria for SR are not standardized among reports, and the clinical outcomes are not fully described. Practically, observation is the only accepted strategy after SR was achieved. In the era of molecular targeted therapy or immunotherapy, new strategies including maintenance therapy after SR could be discussed, although clinical data are lacking.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shigeo Fuji
- Department of Hematology, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
12
|
Bruscaggin A, Pini K, Rossi D. Detection of Circulating Tumor DNA in Lymphoma Patients. Methods Mol Biol 2025; 2865:475-490. [PMID: 39424738 DOI: 10.1007/978-1-0716-4188-0_21] [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: 10/21/2024]
Abstract
Liquid biopsy has become an opportunity in lymphoma diagnostics, since plasma circulating tumor DNA (ctDNA) is an easily accessible source of tumor DNA, which can be complementary to tissue biopsy. Through ctDNA, lymphomas can be molecularly characterized, tumor clonal evolution can be tracked, while monitoring minimal residual diseases during and after therapy. Here, we describe the methodology of cancer personalized profiling by deep sequencing (CAPP-seq) that we use for ctDNA qualification and quantification.
Collapse
Affiliation(s)
- Alessio Bruscaggin
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Katia Pini
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Davide Rossi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland.
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Faculty of Biomedical Science, Universita' della Svizzera italiana, Lugano, Switzerland.
| |
Collapse
|
13
|
Kou M, Deng Y. Circulating tumor DNA as a predictive biomarker for treatment response and survival in metastatic colorectal cancer. Int J Colorectal Dis 2024; 39:203. [PMID: 39681775 DOI: 10.1007/s00384-024-04785-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE To explore the potential of circulating tumor DNA (ctDNA) as a prognostic biomarker to predict treatment response and survival outcomes in patients with metastatic colorectal cancer (mCRC). METHODS A retrospective analysis was conducted on 134 patients with mCRC who were treated between January 2020 and December 2021. The patients were classified into ctDNA-negative and ctDNA-positive groups based on plasma ctDNA detection. Demographic, clinical, and laboratory parameters, treatment response, survival outcomes, and adverse events were recorded and analyzed. RESULTS No significant differences were observed in baseline characteristics between the two groups. Compared to the ctDNA-positive patients, ctDNA-negative patients exhibited superior outcomes, including a higher objective response rate (65.22% vs. 46.15%), disease control rate (81.16% vs. 63.08%), progression-free survival (8.24 ± 1.02 vs. 7.86 ± 0.91 months), overall survival (24.58 ± 3.58 vs. 23.27 ± 3.46 months), and 1-year survival rate (73.91% vs. 55.38%). The ctDNA-positive group had a significantly higher incidence of adverse events. Correlation analyses revealed significant associations between ctDNA status, tumor markers, treatment response, and survival outcomes. CONCLUSIONS ctDNA is a promising noninvasive biomarker for predicting treatment response, survival, and adverse events in mCRC, potentially guiding personalized therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Kou
- Department of Oncology, Maoming People's Hospital, Maoming City, Guangdong Province, China
| | - Ying Deng
- Department of Gastroenterology, Maoming People's Hospital, No.101 Weimin Road, Maonan District, Maoming City, Guangdong Province, 525000, China.
| |
Collapse
|
14
|
Ma L, Guo H, Zhao Y, Liu Z, Wang C, Bu J, Sun T, Wei J. Liquid biopsy in cancer current: status, challenges and future prospects. Signal Transduct Target Ther 2024; 9:336. [PMID: 39617822 PMCID: PMC11609310 DOI: 10.1038/s41392-024-02021-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/10/2024] [Accepted: 10/14/2024] [Indexed: 12/06/2024] Open
Abstract
Cancer has a high mortality rate across the globe, and tissue biopsy remains the gold standard for tumor diagnosis due to its high level of laboratory standardization, good consistency of results, relatively stable samples, and high accuracy of results. However, there are still many limitations and drawbacks in the application of tissue biopsy in tumor. The emergence of liquid biopsy provides new ideas for early diagnosis and prognosis of tumor. Compared with tissue biopsy, liquid biopsy has many advantages in the diagnosis and treatment of various types of cancer, including non-invasive, quickly and so on. Currently, the application of liquid biopsy in tumor detection has received widely attention. It is now undergoing rapid progress, and it holds significant potential for future applications. Around now, liquid biopsies encompass several components such as circulating tumor cells, circulating tumor DNA, exosomes, microRNA, circulating RNA, tumor platelets, and tumor endothelial cells. In addition, advances in the identification of liquid biopsy indicators have significantly enhanced the possibility of utilizing liquid biopsies in clinical settings. In this review, we will discuss the application, advantages and challenges of liquid biopsy in some common tumors from the perspective of diverse systems of tumors, and look forward to its future development prospects in the field of cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Huiling Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Yunxiang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhibo Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Chenran Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China
| | - Jiahao Bu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Clinical Laboratory of Henan province, Zhengzhou, Henan, China.
| | - Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
15
|
Jamal E, Poynton E, Elbogdady M, Shamaa S, Okosun J. Prospects for liquid biopsy approaches in lymphomas. Leuk Lymphoma 2024; 65:1923-1933. [PMID: 39126310 PMCID: PMC11627208 DOI: 10.1080/10428194.2024.2389210] [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: 06/26/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024]
Abstract
Analytes within liquid biopsies have emerged as promising alternatives to traditional tissue biopsies for various malignancies, including lymphomas. This review explores the clinical applications of one such liquid biopsy analyte, circulating tumor DNA (ctDNA) in different types of lymphoma, focusing on its role in diagnosis, disease monitoring, and relapse detection. Advancements in next-generation sequencing (NGS) and machine learning have enhanced ctDNA analysis, offering a multi-omic approach to understanding tumor genetics. In lymphoma, ctDNA provides insights into tumor heterogeneity, aids in genetic profiling, and predicts treatment response. Recent studies demonstrate the prognostic value of ctDNA and its potential to improve patient outcomes by facilitating early disease detection and personalized treatment strategies Despite these advancements, challenges remain in optimizing sample collection, processing, assay sensitivity, and overall consensus workflows in order to facilitate integration into routine clinical practice.
Collapse
Affiliation(s)
- Esraa Jamal
- Centre of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Clinical Haematology Unit, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Edward Poynton
- Centre of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Mohamed Elbogdady
- Clinical Haematology Unit, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Sameh Shamaa
- Clinical Haematology Unit, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Jessica Okosun
- Centre of Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
16
|
Li JY, Zuo LP, Xu J, Sun CY. Clinical applications of circulating tumor DNA in hematological malignancies: From past to the future. Blood Rev 2024; 68:101237. [PMID: 39261219 DOI: 10.1016/j.blre.2024.101237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Liquid biopsy, particularly circulating tumor DNA (ctDNA), has drawn a lot of attention as a non- or minimal-invasive detection approach for clinical applications in patients with cancer. Many hematological malignancies are well suited for serial and repeated ctDNA surveillance due to relatively high ctDNA concentrations and high loads of tumor-specific genetic and epigenetic abnormalities. Progress of detecting technology in recent years has improved sensitivity and specificity significantly, thus broadening and strengthening the potential utilities of ctDNA including early diagnosis, prognosis estimation, treatment response evaluation, minimal residual disease monitoring, targeted therapy selection, and immunotherapy surveillance. This manuscript reviews the detection methodologies, clinical application and future challenges of ctDNA in hematological malignancies, especially for lymphomas, myeloma and leukemias.
Collapse
Affiliation(s)
- Jun-Ying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Li-Ping Zuo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Jian Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China
| | - Chun-Yan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
17
|
Siy AB, Zhou M, Boncompagni ACA, Charville G, Poultsides G, Ganjoo KN. Monitoring Response Using Circulating Tumor DNA in Undifferentiated Pleomorphic Sarcoma: A Case Report. Cureus 2024; 16:e74837. [PMID: 39737274 PMCID: PMC11684465 DOI: 10.7759/cureus.74837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2024] [Indexed: 01/01/2025] Open
Abstract
Circulating tumor DNA (ctDNA) can be used to assess treatment response in patients with undifferentiated pleomorphic sarcoma (UPS). The importance of this is explored in our case of a 75-year-old man who was diagnosed with UPS of the right kidney. After a right nephrectomy and tumor resection, the patient was recovering well with initially undetectable, and then slightly elevated, circulating tumor DNA. Abdominal pain started shortly before a scheduled magnetic resonance imaging (MRI) which revealed a large mass in the resection bed invading the liver. The patient was treated with gemcitabine and docetaxel chemotherapy, and the ctDNA level rose dramatically before gradually decreasing and eventually becoming undetectable. At surgery, pathologic examination of the re-resection specimen revealed a complete pathological response. ctDNA monitoring may be a useful tool for early detection of response to chemotherapy in patients with UPS.
Collapse
Affiliation(s)
- Amanda B Siy
- Department of Medical Oncology, Stanford University, Stanford, USA
| | - Maggie Zhou
- Department of Hematology and Medical Oncology, Stanford University, Stanford, USA
| | | | | | | | - Kristen N Ganjoo
- Department of Medical Oncology, Stanford University, Stanford, USA
| |
Collapse
|
18
|
Hasanabadi S, Aghamiri SMR, Abin AA, Abdollahi H, Arabi H, Zaidi H. Enhancing Lymphoma Diagnosis, Treatment, and Follow-Up Using 18F-FDG PET/CT Imaging: Contribution of Artificial Intelligence and Radiomics Analysis. Cancers (Basel) 2024; 16:3511. [PMID: 39456604 PMCID: PMC11505665 DOI: 10.3390/cancers16203511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Lymphoma, encompassing a wide spectrum of immune system malignancies, presents significant complexities in its early detection, management, and prognosis assessment since it can mimic post-infectious/inflammatory diseases. The heterogeneous nature of lymphoma makes it challenging to definitively pinpoint valuable biomarkers for predicting tumor biology and selecting the most effective treatment strategies. Although molecular imaging modalities, such as positron emission tomography/computed tomography (PET/CT), specifically 18F-FDG PET/CT, hold significant importance in the diagnosis of lymphoma, prognostication, and assessment of treatment response, they still face significant challenges. Over the past few years, radiomics and artificial intelligence (AI) have surfaced as valuable tools for detecting subtle features within medical images that may not be easily discerned by visual assessment. The rapid expansion of AI and its application in medicine/radiomics is opening up new opportunities in the nuclear medicine field. Radiomics and AI capabilities seem to hold promise across various clinical scenarios related to lymphoma. Nevertheless, the need for more extensive prospective trials is evident to substantiate their reliability and standardize their applications. This review aims to provide a comprehensive perspective on the current literature regarding the application of AI and radiomics applied/extracted on/from 18F-FDG PET/CT in the management of lymphoma patients.
Collapse
Affiliation(s)
- Setareh Hasanabadi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Seyed Mahmud Reza Aghamiri
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Ahmad Ali Abin
- Faculty of Computer Science and Engineering, Shahid Beheshti University, Tehran 1983969411, Iran;
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, 500 Odense, Denmark
- University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| |
Collapse
|
19
|
Brahimllari O, Eloranta S, Georgii-Hemming P, Haider Z, Koch S, Krstic A, Skarp FP, Rosenquist R, Smedby KE, Taylan F, Thorvaldsdottir B, Wirta V, Wästerlid T, Boman M. Smart variant filtering - A blueprint solution for massively parallel sequencing-based variant analysis. Health Informatics J 2024; 30:14604582241290725. [PMID: 39394057 DOI: 10.1177/14604582241290725] [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: 10/13/2024]
Abstract
Massively parallel sequencing helps create new knowledge on genes, variants and their association with disease phenotype. This important technological advancement simultaneously makes clinical decision making, using genomic information for cancer patients, more complex. Currently, identifying actionable pathogenic variants with diagnostic, prognostic, or predictive impact requires substantial manual effort. Objective: The purpose is to design a solution for clinical diagnostics of lymphoma, specifically for systematic variant filtering and interpretation. Methods: A scoping review and demonstrations from specialists serve as a basis for a blueprint of a solution for massively parallel sequencing-based genetic diagnostics. Results: The solution uses machine learning methods to facilitate decision making in the diagnostic process. A validation round of interviews with specialists consolidated the blueprint and anchored it across all relevant expert disciplines. The scoping review identified four components of variant filtering solutions: algorithms and Artificial Intelligence (AI) applications, software, bioinformatics pipelines and variant filtering strategies. The blueprint describes the input, the AI model and the interface for dynamic browsing. Conclusion: An AI-augmented system is designed for predicting pathogenic variants. While such a system can be used to classify identified variants, diagnosticians should still evaluate the classification's accuracy, make corrections when necessary, and ultimately decide which variants are truly pathogenic.
Collapse
Affiliation(s)
- Orlinda Brahimllari
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Eloranta
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Zahra Haider
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sabine Koch
- Department of Learning, Informatics, Management and Ethics, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra Krstic
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | | | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Karin E Smedby
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Fulya Taylan
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Birna Thorvaldsdottir
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Valtteri Wirta
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell biology, Karolinska Institutet, Stockholm, 17177, Sweden
- Genomic Medicine Center Karolinska, Karolinska University Hospital, Stockholm, Sweden
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Division of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tove Wästerlid
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Boman
- MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Huang DZ, Zhang X, Rao J. [Progression and application of circulating tumor DNA in lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:878-882. [PMID: 39414617 PMCID: PMC11518914 DOI: 10.3760/cma.j.cn121090-20240528-00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 10/18/2024]
Abstract
Lymphomas are a highly heterogeneous group of tumors that are classified into several subtypes. The gold standard method for the molecular profiling of lymphoma is based on invasive lymph node or tissue biopsy. However, this method cannot accurately capture spatial tumor heterogeneity in each patient as well as systemic tumor invasion and tumor burden. Circulating tumor DNA (ctDNA) is an emerging and highly versatile biomarker that overcomes the basic limitations of imaging scanning and tissue biopsy; has the characteristics of being simple, rapid, and non-invasive; and has good specificity and high sensitivity. ctDNA testing has been applied to a variety of subtypes of lymphoma and has been used for somatic mutation genotyping, efficacy monitoring during treatment, detection of minimal residual disease, and the prediction of survival, which may help clinicians make better clinical decisions in the diagnosis and treatment of lymphoma patients. Furthermore, this study also aims to review the different methods of ctDNA analysis and describe the specific applications of ctDNA in different lymphoma subtypes.
Collapse
Affiliation(s)
- D Z Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Microenvironment, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400037, China
| | - X Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Microenvironment, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400037, China Jinfeng Laboratory, Chongqing 401329, China
| | - J Rao
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing Key Laboratory of Hematology and Microenvironment, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400037, China
| |
Collapse
|
21
|
Mishra M, Ahmed R, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent Advancements in the Application of Circulating Tumor DNA as Biomarkers for Early Detection of Cancers. ACS Biomater Sci Eng 2024; 10:4740-4756. [PMID: 38950521 PMCID: PMC11322919 DOI: 10.1021/acsbiomaterials.4c00606] [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: 03/30/2024] [Revised: 05/24/2024] [Accepted: 06/21/2024] [Indexed: 07/03/2024]
Abstract
Early detection of cancer is vital for increasing patient survivability chances. The three major techniques used to diagnose cancers are instrumental examination, tissue biopsy, and tumor biomarker detection. Circulating tumor DNA (ctDNA) has gained much attention in recent years due to advantages over traditional technology, such as high sensitivity, high specificity, and noninvasive nature. Through the mechanism of apoptosis, necrosis, and circulating exosome release in tumor cells, ctDNA can spread throughout the circulatory system and carry modifications such as methylations, mutations, gene rearrangements, and microsatellite instability. Traditional gene-detection technology struggles to achieve real-time, low-cost, and portable ctDNA measurement, whereas electrochemical biosensors offer low cost, high specificity alongside sensitivity, and portability for the detection of ctDNA. Therefore, this review focuses on describing the recent advancements in ctDNA biomarkers for various cancer types and biosensor developments for real-time, noninvasive, and rapid ctDNA detection. Further in the review, ctDNA sensors are also discussed in regards to their selections of probes for receptors based on the electrode surface recognition elements.
Collapse
Affiliation(s)
- Mahima Mishra
- Amity Institute
of Biotechnology, Amity University, Noida 201301, India
| | - Rubai Ahmed
- Department of Physiology, University of Gour Banga, Malda-732103, West Bengal, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, 281406 Uttar Pradesh, India
| | | | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda-732103, West Bengal, India
| | - Arindam Pramanik
- Amity Institute
of Biotechnology, Amity University, Noida 201301, India
- School of Medicine, University of Leeds, Leeds LS53RL, United Kingdom
| |
Collapse
|
22
|
Zhang S, Wang X, Yang Z, Ding M, Zhang M, Young KH, Zhang X. Minimal residual disease detection in lymphoma: methods, procedures and clinical significance. Front Immunol 2024; 15:1430070. [PMID: 39188727 PMCID: PMC11345172 DOI: 10.3389/fimmu.2024.1430070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Lymphoma is a highly heterogeneous lymphohematopoietic tumor. As our understanding of the biological and pathological characteristics of lymphoma improves, we are identifying an increasing number of lymphoma subtypes. Genotyping has enhanced our ability to diagnose, treat, and monitor the prognosis of lymphoma. Despite significant improvements in treatment effectiveness, traditional methods for assessing disease response and monitoring prognosis are imperfect, and there is no significant improvement in overall remission rates for lymphoma patients. Minimal Residual Disease (MRD) is often indicative of refractory disease or early relapse. For lymphoma patients, personalized MRD monitoring techniques offer an efficient means to estimate disease remission levels, predict early relapse risk, and assess the effectiveness of new drug regimens. In this review, we delve into the MRD procedures in lymphoma, including sample selection and requirements, detection methods and their limitations and advantages, result interpretation. Besides, we also introduce the clinical applications of MRD detection in lymphoma.
Collapse
Affiliation(s)
- Sijun Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Xiangyu Wang
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mengjie Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Ken H. Young
- Division of Hematopathology, Duke University Medicine Center, Duke Cancer Institute, Durham, NC, United States
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Bommier C, Maurer MJ, Lambert J. What clinicians should know about surrogate end points in hematologic malignancies. Blood 2024; 144:11-20. [PMID: 38603637 DOI: 10.1182/blood.2023022269] [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: 10/10/2023] [Revised: 03/14/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
ABSTRACT Use of surrogates as primary end points is commonplace in hematology/oncology clinical trials. As opposed to prognostic markers, surrogates are end points that can be measured early and yet can still capture the full effect of treatment, because it would be captured by the true outcome (eg, overall survival). We discuss the level of evidence of the most commonly used end points in hematology and share recommendations on how to apply and evaluate surrogate end points in research and clinical practice. Based on the statistical literature, this clinician-friendly review intends to build a bridge between clinicians and surrogacy specialists.
Collapse
Affiliation(s)
- Côme Bommier
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| | - Matthew John Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Jerome Lambert
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| |
Collapse
|
24
|
Banchi M, Cox MC, Bocci G. Metronomic chemotherapy in hematology: Lessons from preclinical and clinical studies to build a solid rationale for future schedules. Cancer Lett 2024; 591:216900. [PMID: 38636896 DOI: 10.1016/j.canlet.2024.216900] [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: 11/02/2023] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Metronomic chemotherapy (mCHEMO), based on frequent, regular administration of low, but pharmacologically active drug doses, optimizes antitumor efficacy by targeting multiple targets and reducing toxicity of antineoplastic drugs. This minireview will summarize preclinical and clinical studies on cytotoxic drugs given at weekly, daily, or at continuous metronomic schedules alone or in combination with novel targeted agents for hematological malignancies, including lymphoma, multiple myeloma, and leukemia. Most of the preclinical in vitro and in vivo studies have reported a significant benefit of both mCHEMO monotherapy and combinatorial regimens compared with chemotherapy at the maximum tolerated dose. However, the combination of mCHEMO with targeted drugs is still little explored in the hematologic clinical setting. Data obtained from preclinical studies on low dose metronomic chemotherapy in hematological malignancies clearly suggested the possibility to clinically investigate more tolerable and effective strategies for the treatment of patients with advanced hematological malignancies, or at least for those frail and elderly patients, who are not eligible or resistant to standard treatments.
Collapse
Affiliation(s)
- Marta Banchi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | | | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy.
| |
Collapse
|
25
|
Monick S, Rosenthal A. Circulating Tumor DNA as a Complementary Prognostic Biomarker during CAR-T Therapy in B-Cell Non-Hodgkin Lymphomas. Cancers (Basel) 2024; 16:1881. [PMID: 38791959 PMCID: PMC11120115 DOI: 10.3390/cancers16101881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The emergence of CD19-directed chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment paradigm for R/R B-cell NHLs. However, challenges persist in accurately evaluating treatment response and detecting early relapse, necessitating the exploration of novel biomarkers. Circulating tumor DNA (ctDNA) via liquid biopsy is a non-invasive tool for monitoring therapy efficacy and predicting treatment outcomes in B-NHL following CAR-T therapy. By overcoming the limitations of conventional imaging modalities, ctDNA assessments offer valuable insights into response dynamics, molecular mechanisms of resistance, and early detection of molecular relapse. Integration of ctDNA monitoring into clinical practice holds promise for personalized therapeutic strategies, guiding the development of novel targeted therapies, and enhancing patient outcomes. However, standardization of assay methodologies and consensus on clinical response metrics are imperative to unlock the full potential of ctDNA in the management of B-NHL. Prospective validation of ctDNA in clinical trials is necessary to establish its role as a complementary decision aid.
Collapse
Affiliation(s)
- Sarah Monick
- Department of Medicine, Mayo Clinic Arizona, Phoenix, AZ 85054, USA;
| | | |
Collapse
|
26
|
Sánchez-Beato M, Méndez M, Guirado M, Pedrosa L, Sequero S, Yanguas-Casás N, de la Cruz-Merino L, Gálvez L, Llanos M, García JF, Provencio M. A genetic profiling guideline to support diagnosis and clinical management of lymphomas. Clin Transl Oncol 2024; 26:1043-1062. [PMID: 37672206 PMCID: PMC11026206 DOI: 10.1007/s12094-023-03307-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023]
Abstract
The new lymphoma classifications (International Consensus Classification of Mature Lymphoid Neoplasms, and 5th World Health Organization Classification of Lymphoid Neoplasms) include genetics as an integral part of lymphoma diagnosis, allowing better lymphoma subclassification, patient risk stratification, and prediction of treatment response. Lymphomas are characterized by very few recurrent and disease-specific mutations, and most entities have a heterogenous genetic landscape with a long tail of recurrently mutated genes. Most of these occur at low frequencies, reflecting the clinical heterogeneity of lymphomas. Multiple studies have identified genetic markers that improve diagnostics and prognostication, and next-generation sequencing is becoming an essential tool in the clinical laboratory. This review provides a "next-generation sequencing" guide for lymphomas. It discusses the genetic alterations of the most frequent mature lymphoma entities with diagnostic, prognostic, and predictive potential and proposes targeted sequencing panels to detect mutations and copy-number alterations for B- and NK/T-cell lymphomas.
Collapse
Affiliation(s)
- Margarita Sánchez-Beato
- Servicio de Oncología Médica, Grupo de Investigación en Linfomas, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain.
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain.
| | - Miriam Méndez
- Servicio de Oncología Médica, Grupo de Investigación en Linfomas, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain
| | - María Guirado
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Hospital General Universitario de Elche, Alicante, Spain
| | - Lucía Pedrosa
- Servicio de Oncología Médica, Grupo de Investigación en Linfomas, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain
| | - Silvia Sequero
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Hospital Universitario San Cecilio, Granada, Spain
| | - Natalia Yanguas-Casás
- Servicio de Oncología Médica, Grupo de Investigación en Linfomas, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain
| | - Luis de la Cruz-Merino
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Facultad de Medicina, Hospital Universitario Virgen Macarena, Universidad de Sevilla, Instituto de Biomedicina de Sevilla (IBID)/CSIC, Seville, Spain
| | - Laura Gálvez
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Málaga, Spain
| | - Marta Llanos
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Hospital Universitario de Canarias, La Laguna, Sta. Cruz de Tenerife, Spain
| | - Juan Fernando García
- Servicio de Anatomía Patológica, Hospital MD Anderson Cancer Center, Madrid, Spain
| | - Mariano Provencio
- Servicio de Oncología Médica, Grupo de Investigación en Linfomas, Hospital Universitario Puerta de Hierro-Majadahonda, IDIPHISA, Madrid, Spain
- Grupo Oncológico para el Tratamiento y Estudio de los Linfomas-GOTEL, Madrid, Spain
- Servicio de Oncología Médica, Departamento de Medicina, Facultad de Medicina, Hospital Universitario Puerta de Hierro-Majadahonda, Universidad Autónoma de Madrid, IDIPHISA, Madrid, Spain
| |
Collapse
|
27
|
Li L, Shou L. Prognostic and clinicopathological significance of the Controlling Nutritional Status (CONUT) score in patients with lymphoma: a meta-analysis. BMJ Open 2024; 14:e078320. [PMID: 38453197 PMCID: PMC10921528 DOI: 10.1136/bmjopen-2023-078320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024] Open
Abstract
OBJECTIVES The role of the Controlling Nutritional Status (CONUT) scores in predicting the prognosis of lymphoma cases has been extensively explored, with no consistent results. The present meta-analysis focused on accurately evaluating whether CONUT could be used to predict the prognosis of lymphoma cases and its clinicopathological value. DESIGN The present meta-analysis was reported following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The prognostic significance of CONUT to overall survival (OS) and progression-free survival (PFS) in lymphoma was estimated by calculating pooled HRs with 95% CIs. The relationship between CONUT and clinicopathological characteristics was measured based on pooled ORs with 95% CIs. DATA SOURCES PubMed, Web of Science, Embase and Cochrane Library databases were comprehensively searched from inception through 24 March 2023. STATISTICAL METHODS Either a random-effects model or a fixed-effects model was selected depending on the level of heterogeneity among the included studies. RESULTS This meta-analysis enrolled seven articles, containing 2060 patients with lymphoma. According to the pooled analysis, a higher CONUT score significantly predicted poor OS (HR=1.94, 95% CI 1.46 to 2.57, p<0.001) as well as poorer PFS (HR=1.51, 95% CI 1.04 to 2.20, p=0.031). Furthermore, according to the combined analysis, a higher CONUT score was significantly associated with Ann Arbor stages III-IV (OR=3.75, 95% CI 2.96 to 4.75, p<0.001), an Eastern Cooperative Oncology Group performance status of 2-4 (OR=5.14, 95% CI 3.97 to 6.65, p<0.001), high-intermediate/high National Comprehensive Cancer Network International Prognostic Index (OR=8.05, 95% CI 5.11 to 12.66, p<0.001), B symptoms (OR=4.97, 95% CI 2.89 to 8.52, p<0.001), extranodal disease (OR=3.25, 95% CI 2.24 to 4.70, p<0.001), bone marrow involvement (OR=4.86, 95% CI 3.25 to 7.27, p<0.001) and elevated lactate dehydrogenase levels (OR=3.21, 95% CI 2.37 to 4.34, p<0.001). CONCLUSIONS According to our results, higher CONUT scores were significantly associated with poor OS and PFS in lymphoma.
Collapse
Affiliation(s)
- Lili Li
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| | - Lihong Shou
- Department of Hematology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| |
Collapse
|
28
|
Kelly KM, Friedberg JW. Classic Hodgkin Lymphoma in Adolescents and Young Adults. J Clin Oncol 2024; 42:653-664. [PMID: 37983570 DOI: 10.1200/jco.23.01799] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 08/28/2023] [Accepted: 09/20/2023] [Indexed: 11/22/2023] Open
Abstract
Hodgkin lymphoma (HL) represents one of the more common cancers occurring in adolescent and young adults (AYAs) age 15-39 years. Despite a generally high cure rate, age-related differences in HL biology and the optimal therapeutic approaches including supportive care and risks for long-term adverse effects in the AYA population remain understudied. After an overview of HL epidemiology and biology in the AYA population, this review will cover frontline pediatric and adult treatment approaches. Recently completed and ongoing studies will foster harmonization of risk group definition and trial eligibility criteria across the AYA spectrum, enabling more rapid progress. In addition to treatment approaches, an evolving holistic care approach to AYA HL will result in enhanced understanding of unique challenges, and continued improved short- and long-term outcome for these patients.
Collapse
Affiliation(s)
- Kara M Kelly
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center., Buffalo, NY
- Division of Pediatric Hematology/Oncology, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY
- Pediatric Hematology/Oncology, Oishei Children's Hospital, Buffalo, NY
| | | |
Collapse
|
29
|
Winter JN, Ferreri AJM. Predicting outcomes in CNS lymphoma with ctDNA. Blood 2024; 143:478-480. [PMID: 38329774 DOI: 10.1182/blood.2023023008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Affiliation(s)
| | - Andres J M Ferreri
- Istituto di Ricerca e Cura a Carattere Scientifico Ospedale San Raffaele
| |
Collapse
|
30
|
Calabretta E, di Trani M, Corrado F, Sollini M, Cristaldi V, Marino F, Terzi di Bergamo L, Bruscaggin A, Pirosa MC, Bramanti S, Chiti A, Rossi D, Carlo-Stella C. Baseline circulating tumour DNA and interim PET predict response in relapsed/refractory classical Hodgkin lymphoma. Br J Haematol 2024; 204:514-524. [PMID: 37853658 DOI: 10.1111/bjh.19162] [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: 08/13/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023]
Abstract
Reliable biomarkers for early identification of treatment failure in relapsed/refractory (r/r) classical Hodgkin lymphoma (cHL) are lacking. Circulating tumour DNA (ctDNA) profiling has emerged as a powerful predictive and prognostic tool in several haemopoietic and non-haemopoietic malignancies and may guide rational treatment choices in r/r cHL. To assess the predictive and prognostic value of ctDNA, we performed a retrospective analysis on 55 r/r cHL patients treated with the bendamustine, gemcitabine and vinorelbine (BEGEV) regimen and additionally evaluated the potential utility of integrating ctDNA with interim [18 F]-FDG positron emission tomography (iPET). Baseline ctDNA genotyping in r/r cHL mirrored gene mutations and pathways involved in newly diagnosed cHL. We found that baseline ctDNA quantification and serial ctDNA monitoring have prognostic value in r/r cHL receiving salvage chemotherapy. Lastly, integrating ctDNA quantification with iPET evaluation may improve the early identification of patients at high risk of failing standard salvage therapy, who may benefit from an early switch to immunotherapeutic agents. Collectively, our results support the implementation of non-invasive methods to detect minimal residual disease in recurrent cHL and justify its prospective evaluation in appropriately designed clinical trials.
Collapse
Affiliation(s)
- Eleonora Calabretta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martina di Trani
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Francesco Corrado
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Vanessa Cristaldi
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Fabrizio Marino
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lodovico Terzi di Bergamo
- Laboratory of Experimental Hematology, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Alessio Bruscaggin
- Laboratory of Experimental Hematology, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Maria Cristina Pirosa
- Laboratory of Experimental Hematology, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Stefania Bramanti
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Davide Rossi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Department of Oncology and Hematology, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
31
|
Fend F, van den Brand M, Groenen PJ, Quintanilla-Martinez L, Bagg A. Diagnostic and prognostic molecular pathology of lymphoid malignancies. Virchows Arch 2024; 484:195-214. [PMID: 37747559 PMCID: PMC10948535 DOI: 10.1007/s00428-023-03644-0] [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: 08/03/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/26/2023]
Abstract
With the explosion in knowledge about the molecular landscape of lymphoid malignancies and the increasing availability of high throughput techniques, molecular diagnostics in hematopathology has moved from isolated marker studies to a more comprehensive approach, integrating results of multiple genes analyzed with a variety of techniques on the DNA and RNA level. Although diagnosis of lymphoma still relies on the careful integration of clinical, morphological, phenotypic, and, if necessary molecular features, and only few entities are defined strictly by genetic features, genetic profiling has contributed profoundly to our current understanding of lymphomas and shaped the two current lymphoma classifications, the International Consensus Classification and the fifth edition of the WHO classification of lymphoid malignancies. In this review, the current state of the art of molecular diagnostics in lymphoproliferations is summarized, including clonality analysis, mutational studies, and gene expression profiling, with a focus on practical applications for diagnosis and prognostication. With consideration for differences in accessibility of high throughput techniques and cost limitations, we tried to distinguish between diagnostically relevant and in part disease-defining molecular features and optional, more extensive genetic profiling, which is usually restricted to clinical studies, patients with relapsed or refractory disease or specific therapeutic decisions. Although molecular diagnostics in lymphomas currently is primarily done for diagnosis and subclassification, prognostic stratification and predictive markers will gain importance in the near future.
Collapse
Affiliation(s)
- Falko Fend
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany.
| | - Michiel van den Brand
- Pathology-DNA, Location Rijnstate Hospital, Arnhem, the Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Patricia Jta Groenen
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image Guided and Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, Tübingen, Germany
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
32
|
Abstract
Lymphoid neoplasms represent a heterogeneous group of disease entities and subtypes with markedly different molecular and clinical features. Beyond genetic alterations, lymphoid tumors also show widespread epigenomic changes. These severely affect the levels and distribution of DNA methylation, histone modifications, chromatin accessibility, and three-dimensional genome interactions. DNA methylation stands out as a tracer of cell identity and memory, as B cell neoplasms show epigenetic imprints of their cellular origin and proliferative history, which can be quantified by an epigenetic mitotic clock. Chromatin-associated marks are informative to uncover altered regulatory regions and transcription factor networks contributing to the development of distinct lymphoid tumors. Tumor-intrinsic epigenetic and genetic aberrations cooperate and interact with microenvironmental cells to shape the transcriptome at different phases of lymphoma evolution, and intraclonal heterogeneity can now be characterized by single-cell profiling. Finally, epigenetics offers multiple clinical applications, including powerful diagnostic and prognostic biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Martí Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain;
| | - José Ignacio Martín-Subero
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain;
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
33
|
Yu D, Wang J, Zheng B, Yuan M, Gu D, Chen R, Chen X. Comprehensive genomic profiling of small bowel adenocarcinoma by tissue and plasma biopsy. Genomics 2024; 116:110766. [PMID: 38141930 DOI: 10.1016/j.ygeno.2023.110766] [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: 08/21/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Small bowel adenocarcinoma (SBA) is a rare and aggressive malignancy with limited treatment options and poor prognosis. The molecular landscape and immunological characteristics of SBA are poorly understood. Here, we performed comprehensive mutation profiling of tissue and plasma biopsies from 143 and 42 patients with SBA. Analysis showed that SBA had a distinct mutation spectrum from left- and right-sided colorectal carcinoma. Plasma biopsy had high concordance with tissue biopsy for single nucleotide variants and structural variants, but low concordance for copy number variations, which showed that plasma biopsy can be an alternative to tissue biopsy. Moreover, we analyzed the association of TMB with clinical and molecular features, and found that TMB was significantly higher in tumors with DNA damage response alterations. Our findings provide valuable insights into the molecular and immunological features of SBA and demonstrate the potential of plasma biopsy as a non-invasive method for SBA diagnosis and treatment.
Collapse
Affiliation(s)
- Dan Yu
- The Second Xiangya Hospital of Central South University, Hunan 410011, China
| | - Jianzheng Wang
- Department of Oncology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer & Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou 450008, China
| | - Bo Zheng
- Geneplus-Beijing, Beijing 102206, China.
| | | | - Dejian Gu
- Geneplus-Beijing, Beijing 102206, China
| | | | - Xiaobing Chen
- Department of Oncology, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan Engineering Research Center of Precision Therapy of Gastrointestinal Cancer & Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou 450008, China.
| |
Collapse
|
34
|
Kim JJ, Kim HM, Kim H, Kim SJ, Lee ST, Choi JR, Shin S, Hwang DY. Circulating Tumor DNA Reflects Histologic and Clinical Characteristics of Various Lymphoma Subtypes. Cancer Res Treat 2024; 56:314-323. [PMID: 37475138 PMCID: PMC10789961 DOI: 10.4143/crt.2023.667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/15/2023] [Indexed: 07/22/2023] Open
Abstract
PURPOSE We designed and evaluated the clinical performance of a plasma circulating tumor DNA (ctDNA) panel of 112 genes in various subtypes of lymphoma. MATERIALS AND METHODS Targeted deep sequencing with an error-corrected algorithm was performed in ctDNA from plasma samples that were collected before treatment in 42 lymphoma patients. Blood buffy coat was utilized as a germline control. We evaluated the targeted gene panel using mutation detection concordance on the plasma samples with matched tissue samples analyzed the mutation profiles of the ctDNA. RESULTS Next-generation sequencing analysis using matched tissue samples was available for 18 of the 42 patients. At least one mutation was detected in the majority of matched tissue biopsy samples (88.9%) and plasma samples (83.3%). A considerable number of mutations (40.4%) that were detected in the tissue samples were also found in the matched plasma samples. Majority of patients (21/42) were diffuse large B cell lymphoma patients. The overall detection rate of ctDNA in patients was 85.7% (36/42). The frequently mutated genes included PIM1, TET2, BCL2, KMT2D, KLHL6, HIST1H1E, and IRF8. A cutoff concentration (4,506 pg/mL) of ctDNA provided 88.9% sensitivity and 82.1% specificity to predict ctDNA mutation detection. The ctDNA concentration correlated with elevated lactate dehydrogenase level and the disease stage. CONCLUSION Our design panel can detect many actionable gene mutations, including those at low frequency. Therefore, liquid biopsy can be applied clinically in the evaluation of lymphoma patients, especially in aggressive lymphoma patients.
Collapse
Affiliation(s)
- Jin Ju Kim
- Department of Laboratory Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hye Min Kim
- Department of Pathology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Hongkyung Kim
- Department of Laboratory Medicine, Chung-Ang University Gwangmyung Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Korea
| | - Soo-Jeong Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Doh Yu Hwang
- Division of Hematology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Dean EA, Kimmel GJ, Frank MJ, Bukhari A, Hossain NM, Jain MD, Dahiya S, Miklos DB, Altrock PM, Locke FL. Circulating tumor DNA adds specificity to PET after axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv 2023; 7:4608-4618. [PMID: 37126659 PMCID: PMC10448428 DOI: 10.1182/bloodadvances.2022009426] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/07/2023] [Accepted: 03/30/2023] [Indexed: 05/03/2023] Open
Abstract
We examined the meaning of metabolically active lesions on 1-month restaging nuclear imaging of patients with relapsed/refractory large B-cell lymphoma receiving axicabtagene ciloleucel (axi-cel) by assessing the relationship between total metabolic tumor volume (MTV) on positron emission tomography (PET) scans and circulating tumor DNA (ctDNA) in the plasma. In this prospective multicenter sample collection study, MTV was retrospectively calculated via commercial software at baseline, 1, and 3 months after chimeric antigen receptor (CAR) T-cell therapy; ctDNA was available before and after axi-cel administration. Spearman correlation coefficient (rs) was used to study the relationship between the variables, and a mathematical model was constructed to describe tumor dynamics 1 month after CAR T-cell therapy. The median time between baseline scan and axi-cel infusion was 33 days (range, 1-137 days) for all 57 patients. For 41 of the patients with imaging within 33 days of axi-cel or imaging before that time but no bridging therapy, the correlation at baseline became stronger (rs, 0.61; P < .0001) compared with all patients (rs, 0.38; P = .004). Excluding patients in complete remission with no measurable residual disease, ctDNA and MTV at 1 month did not correlate (rs, 0.28; P = .11) but correlated at 3 months (rs, 0.79; P = .0007). Modeling of tumor dynamics, which incorporated ctDNA and inflammation as part of MTV, recapitulated the outcomes of patients with positive radiologic 1-month scans. Our results suggested that nonprogressing hypermetabolic lesions on 1-month PET represent ongoing treatment responses, and their composition may be elucidated by concurrently examining the ctDNA.
Collapse
Affiliation(s)
- Erin A. Dean
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Institute, Tampa, FL
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL
| | - Gregory J. Kimmel
- Department of Integrated Mathematical Oncology, Moffitt Research Institute, Tampa, FL
| | - Matthew J. Frank
- Division of Blood and Stem Cell Transplantation, Department of Medicine, Stanford University, Stanford, CA
| | - Ali Bukhari
- Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
- Division of Hematology and Oncology, Department of Internal Medicine, Wright-Patterson Medical Center, Wright-Patterson Air Force Base, OH
| | - Nasheed M. Hossain
- Cell Therapy and Transplant Program, Division of Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Institute, Tampa, FL
| | - Saurabh Dahiya
- Division of Blood and Stem Cell Transplantation, Department of Medicine, Stanford University, Stanford, CA
- Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| | - David B. Miklos
- Division of Blood and Stem Cell Transplantation, Department of Medicine, Stanford University, Stanford, CA
| | - Philipp M. Altrock
- Department of Evolutionary Theory, Max Planck Institute for Evolutionary Biology, Ploen, Germany
| | - Frederick L. Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer and Research Institute, Tampa, FL
| |
Collapse
|
36
|
Alderuccio JP, Lossos IS. Enhancing prognostication and personalizing treatment of extranodal marginal zone lymphoma. Expert Rev Hematol 2023; 16:333-348. [PMID: 37086394 PMCID: PMC10183153 DOI: 10.1080/17474086.2023.2206557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/20/2023] [Indexed: 04/23/2023]
Abstract
INTRODUCTION Extranodal marginal zone lymphoma (EMZL) of mucosa-associated lymphoid tissue is an indolent lymphoma originating from marginal zone B-cells and associated with chronic inflammation. EMZL demonstrates distinct genomic alterations according to the primary extranodal site of disease but commonly affects signaling pathways including NF-ĸB, B-cell receptor, and NOTCH. Treatment with radiation therapy is commonly implemented in localized diseases, and multiple agents are available for patients with advanced-stage diseases in need of therapy. Bendamustine with rituximab is a frontline platform associated with high efficacy. AREAS COVERED Clinical features, diagnosis, genomics, models enabling risk stratification, treatment options, and future directions. EXPERT OPINION The lack of consistent genotyping profile in EMZL precludes the development of tissue and circulatory biomarkers for the diagnosis, risk stratification, and monitoring of minimal residual disease. Furthermore, the biological heterogeneity observed in extranodal sites associated with overall limited genomic data prevents the testing of druggable pathways aiming for a personalized treatment approach. Future clinical trials should focus on EMZL considering the unique clinical characteristics in the eligibility criteria and response assessment to better inform efficacy of novel agents and delineate sequences of therapies.
Collapse
Affiliation(s)
| | - Izidore S. Lossos
- Department of Medicine, Division of Hematology
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
37
|
Talotta D, Almasri M, Cosentino C, Gaidano G, Moia R. Liquid biopsy in hematological malignancies: current and future applications. Front Oncol 2023; 13:1164517. [PMID: 37152045 PMCID: PMC10157039 DOI: 10.3389/fonc.2023.1164517] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
The assessment of the cancer mutational profile is crucial for patient management, stratification, and therapeutic decisions. At present, in hematological malignancies with a solid mass, such as lymphomas, tumor genomic profiling is generally performed on the tissue biopsy, but the tumor may harbor genetic lesions that are unique to other anatomical compartments. The analysis of circulating tumor DNA (ctDNA) on the liquid biopsy is an emerging approach that allows genotyping and monitoring of the disease during therapy and follow-up. This review presents the different methods for ctDNA analysis and describes the application of liquid biopsy in different hematological malignancies. In diffuse large B-cell lymphoma (DLBCL) and Hodgkin lymphoma (HL), ctDNA analysis on the liquid biopsy recapitulates the mutational profile of the tissue biopsy and can identify mutations otherwise absent on the tissue biopsy. In addition, changes in the ctDNA amount after one or two courses of chemotherapy significantly predict patient outcomes. ctDNA analysis has also been tested in myeloid neoplasms with promising results. In addition to mutational analysis, liquid biopsy also carries potential future applications of ctDNA, including the analysis of ctDNA fragmentation and epigenetic patterns. On these grounds, several clinical trials aiming at incorporating ctDNA analysis for treatment tailoring are currently ongoing in hematological malignancies.
Collapse
Affiliation(s)
| | | | | | | | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
38
|
Liang X, Hu R, Li Q, Wang C, Liu Y. Prognostic factors for diffuse large B-cell lymphoma: clinical and biological factors in the rituximab era. Exp Hematol 2023:S0301-472X(23)00071-1. [PMID: 36933759 DOI: 10.1016/j.exphem.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023]
Affiliation(s)
- Xiping Liang
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing, China
| | - Renzhi Hu
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing, China
| | - Qiying Li
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing, China
| | - Chaoyu Wang
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing, China
| | - Yao Liu
- Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing, China.
| |
Collapse
|
39
|
Assanto GM, Del Giudice I, Della Starza I, Soscia R, Cavalli M, Cola M, Bellomarino V, Di Trani M, Guarini A, Foà R. Research Topic: Measurable Residual Disease in Hematologic Malignancies. Can digital droplet PCR improve measurable residual disease monitoring in chronic lymphoid malignancies? Front Oncol 2023; 13:1152467. [PMID: 36998457 PMCID: PMC10043164 DOI: 10.3389/fonc.2023.1152467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/01/2023] [Indexed: 03/17/2023] Open
Abstract
Minimal/measurable residual disease (MRD) monitoring is progressively changing the management of hematologic malignancies. The possibility of detecting the persistence/reappearance of disease in patients in apparent clinical remission offers a refined risk stratification and a treatment decision making tool. Several molecular techniques are employed to monitor MRD, from conventional real-time quantitative polymerase chain reaction (RQ-PCR) to next generation sequencing and digital droplet PCR (ddPCR), in different tissues or compartments through the detection of fusion genes, immunoglobulin and T-cell receptor gene rearrangements or disease-specific mutations. RQ-PCR is still the gold standard for MRD analysis despite some limitations. ddPCR, considered the third-generation PCR, yields a direct, absolute, and accurate detection and quantification of low-abundance nucleic acids. In the setting of MRD monitoring it carries the major advantage of not requiring a reference standard curve built with the diagnostic sample dilution and of allowing to reduce the number of samples below the quantitative range. At present, the broad use of ddPCR to monitor MRD in the clinical practice is limited by the lack of international guidelines. Its application within clinical trials is nonetheless progressively growing both in acute lymphoblastic leukemia as well as in chronic lymphocytic leukemia and non-Hodgkin lymphomas. The aim of this review is to summarize the accumulating data on the use of ddPCR for MRD monitoring in chronic lymphoid malignancies and to highlight how this new technique is likely to enter into the clinical practice.
Collapse
Affiliation(s)
| | - Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- *Correspondence: Ilaria Del Giudice, ; Robin Foà,
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- Gruppo Italiano Malattie Ematologiche dell'Adulto (GIMEMA), Fondazione GIMEMA Franco Mandelli Onlus, Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Mattia Cola
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Vittorio Bellomarino
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Mariangela Di Trani
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
- *Correspondence: Ilaria Del Giudice, ; Robin Foà,
| |
Collapse
|
40
|
Jiménez-Ubieto A, Poza M, Martin-Muñoz A, Ruiz-Heredia Y, Dorado S, Figaredo G, Rosa-Rosa JM, Rodriguez A, Barcena C, Navamuel LP, Carrillo J, Sanchez R, Rufian L, Juárez A, Rodriguez M, Wang C, de Toledo P, Grande C, Mollejo M, Casado LF, Calbacho M, Baumann T, Rapado I, Gallardo M, Sarandeses P, Ayala R, Martínez-López J, Barrio S. Real-life disease monitoring in follicular lymphoma patients using liquid biopsy ultra-deep sequencing and PET/CT. Leukemia 2023; 37:659-669. [PMID: 36596983 DOI: 10.1038/s41375-022-01803-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/09/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023]
Abstract
In the present study, we screened 84 Follicular Lymphoma patients for somatic mutations suitable as liquid biopsy MRD biomarkers using a targeted next-generation sequencing (NGS) panel. We found trackable mutations in 95% of the lymph node samples and 80% of the liquid biopsy baseline samples. Then, we used an ultra-deep sequencing approach with 2 · 10-4 sensitivity (LiqBio-MRD) to track those mutations on 151 follow-up liquid biopsy samples from 54 treated patients. Positive LiqBio-MRD at first-line therapy correlated with a higher risk of progression both at the interim evaluation (HRINT 11.0, 95% CI 2.10-57.7, p = 0.005) and at the end of treatment (HREOT, HR 19.1, 95% CI 4.10-89.4, p < 0.001). Similar results were observed by PET/CT Deauville score, with a median PFS of 19 months vs. NR (p < 0.001) at the interim and 13 months vs. NR (p < 0.001) at EOT. LiqBio-MRD and PET/CT combined identified the patients that progressed in less than two years with 88% sensitivity and 100% specificity. Our results demonstrate that LiqBio-MRD is a robust and non-invasive approach, complementary to metabolic imaging, for identifying FL patients at high risk of failure during the treatment and should be considered in future response-adapted clinical trials.
Collapse
Affiliation(s)
- Ana Jiménez-Ubieto
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.
| | - María Poza
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | | | - Yanira Ruiz-Heredia
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,Altum sequencing Co., Madrid, Spain
| | - Sara Dorado
- Altum sequencing Co., Madrid, Spain.,Computational Science Department, Carlos III University, Madrid, Spain
| | | | - Juan Manuel Rosa-Rosa
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Antonia Rodriguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Carmen Barcena
- Hospital Universitario 12 de Octubre, Departamento de Anatomía Patológica, Madrid, Spain
| | | | | | - Ricardo Sanchez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,Altum sequencing Co., Madrid, Spain
| | - Laura Rufian
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,Altum sequencing Co., Madrid, Spain
| | - Alexandra Juárez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,Altum sequencing Co., Madrid, Spain
| | - Margarita Rodriguez
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,Altum sequencing Co., Madrid, Spain
| | - Chongwu Wang
- Hosea Precision Medical Technology Co., Ltd., Weihai, Shangdong, China
| | - Paula de Toledo
- Computational Science Department, Carlos III University, Madrid, Spain
| | | | | | | | - María Calbacho
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Tycho Baumann
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Inmaculada Rapado
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Miguel Gallardo
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain.,H12O-CNIO Haematological Malignancies Clinical Research Unit, CNIO, Madrid, Spain
| | - Pilar Sarandeses
- Hospital Universitario 12 de Octubre, Departamento de Medicina Nuclear, Madrid, Spain
| | - Rosa Ayala
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Joaquín Martínez-López
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain
| | - Santiago Barrio
- Department of Hematology, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), CNIO, CIBERONC, Madrid, Spain. .,Altum sequencing Co., Madrid, Spain.
| |
Collapse
|
41
|
Chokr N, Gomez-Arteaga A. Measurable Residual Disease After CAR T-Cell Therapy. Semin Hematol 2023; 60:34-41. [PMID: 37080709 DOI: 10.1053/j.seminhematol.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/21/2023]
Abstract
Testing for measurable residual disease (MRD) provides important prognostic and predictive implications on survival and management of many hematologic diseases. Among the many clinical uses of MRD is post-therapy response assessment and risk stratification. With the integration of precision medicine in routine clinical care and the development of novel and innovative therapies resulting in deeper responses, it is necessary to refine the role of MRD, standardize available methodologies and define its role as a surrogate endpoint for relapse and time-to-next treatment in clinical studies. Chimeric Antigen Receptor (CAR) T-cell therapy is an approved treatment for various hematologic malignancies. Even though it produces high rates of remission, the durability of response is still a consideration as almost 40% to 50% of patients eventually relapse. MRD testing as a prognostic and surrogate marker is being explored in patients after CAR T-cell therapy to predict early relapse. In this chapter, we review the various tools available for MRD detection and monitoring post-CAR T-cell therapy. We later discuss disease-specific MRD assessment and its application in recent studies in the post-CAR T setting.
Collapse
|
42
|
Gong H, Tang B, Li T, Li J, Tang L, Ding C. The added prognostic values of baseline PET dissemination parameter in patients with angioimmunoblastic T-cell lymphoma. EJHAEM 2023; 4:67-77. [PMID: 36819177 PMCID: PMC9928789 DOI: 10.1002/jha2.610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2022]
Abstract
To explore the prognostic values of baseline 2-deoxy-2-[18F] fluoro-D-glucose (FDG) positron emission tomography/computed tomography (PET/CT) dissemination parameter in angioimmunoblastic T-cell lymphoma (AITL) and its added values to total metabolic tumour volume (TMTV). Eighty-one AITL patients with at least two FDG-avid lesions in baseline PET/CT were retrospectively included. PET parameters including TMTV and the distance between the two lesions that are the furthest apart (Dmax) were obtained. Univariate Cox analysis showed that both Dmax and TMTV were risk factors for progression-free survival (PFS) and overall survival (OS). Multivariate Cox analysis models of different combinations showed that high Dmax (> 65.7 cm) could independently predict both PFS and OS, while high TMTV (>456.6 cm3) was only significant for OS. A concise PET model based on TMTV and Dmax can effectively risk-stratify patients. PFS and OS rates were significantly lower in patients with high Dmax and high TMTV than in patients with low Dmax and low TMTV (3-year PFS rate: 15.0% vs. 48.7%, p = 0.001; 3-year OS rate: 27.6% vs. 79.0%, p < 0.001). Dmax can directly reflect the disease dissemination characteristic and has a significant prognostic value for FDG-avid AITL patients. It has the potential to be introduced into new risk stratification models for tailored treatment.
Collapse
Affiliation(s)
- Huanyu Gong
- Department of Nuclear MedicineJiangsu Province HospitalThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Bo Tang
- Department of RadiologyShuyang Hospital of Traditional Chinese MedicineSuqianChina
| | - Tiannv Li
- Department of Nuclear MedicineJiangsu Province HospitalThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jianyong Li
- Department of HematologyJiangsu Province HospitalThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Lijun Tang
- Department of Nuclear MedicineJiangsu Province HospitalThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Chongyang Ding
- Department of Nuclear MedicineJiangsu Province HospitalThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Recent advances have been made in circulating tumor DNA (ctDNA), the method to minimally invasive detect lymphoma sensitively with tumor-derived DNA in the blood of patients with lymphomas. This article discusses these various methods of ctDNA detection and the clinical context in which they have been applied to for a variety of lymphoma subtypes. RECENT FINDINGS ctDNA has been applied to a variety of subtypes of lymphoma and has been used in the context of genotyping somatic mutations and classification of disease, monitoring of response during treatment, detecting minimal residual disease even with radiographic remission, and predicting relapse and long-term survival outcomes. There are a variety of techniques used to measure ctDNA including digital polymerase chain reaction and next-generation sequencing techniques including high-throughput variable-diversity-joining rearrangement sequencing, high-throughput sequencing of somatic mutations, and Cancer Personalized Profiling by deep sequencing. While the greatest data has been generated in diffuse large B cell lymphoma, there have been studies utilizing application of ctDNA in follicular lymphoma, mantle cell lymphoma, Hodgkin's lymphoma, peripheral T cell lymphoma, and primary CNS lymphoma among others. ctDNA is an emerging biomarker in lymphoma that can minimally invasively provide further genotypic information, diagnostic clarification, and treatment prognostication by detection of minimal residual disease even without radiographic evidence of disease. Future studies are needed to standardize the use of ctDNA and translate its use clinically for the management of lymphoma patients.
Collapse
Affiliation(s)
| | - Jasmine Zain
- City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
44
|
de Leval L, Alizadeh AA, Bergsagel PL, Campo E, Davies A, Dogan A, Fitzgibbon J, Horwitz SM, Melnick AM, Morice WG, Morin RD, Nadel B, Pileri SA, Rosenquist R, Rossi D, Salaverria I, Steidl C, Treon SP, Zelenetz AD, Advani RH, Allen CE, Ansell SM, Chan WC, Cook JR, Cook LB, d’Amore F, Dirnhofer S, Dreyling M, Dunleavy K, Feldman AL, Fend F, Gaulard P, Ghia P, Gribben JG, Hermine O, Hodson DJ, Hsi ED, Inghirami G, Jaffe ES, Karube K, Kataoka K, Klapper W, Kim WS, King RL, Ko YH, LaCasce AS, Lenz G, Martin-Subero JI, Piris MA, Pittaluga S, Pasqualucci L, Quintanilla-Martinez L, Rodig SJ, Rosenwald A, Salles GA, San-Miguel J, Savage KJ, Sehn LH, Semenzato G, Staudt LM, Swerdlow SH, Tam CS, Trotman J, Vose JM, Weigert O, Wilson WH, Winter JN, Wu CJ, Zinzani PL, Zucca E, Bagg A, Scott DW. Genomic profiling for clinical decision making in lymphoid neoplasms. Blood 2022; 140:2193-2227. [PMID: 36001803 PMCID: PMC9837456 DOI: 10.1182/blood.2022015854] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/15/2022] [Indexed: 01/28/2023] Open
Abstract
With the introduction of large-scale molecular profiling methods and high-throughput sequencing technologies, the genomic features of most lymphoid neoplasms have been characterized at an unprecedented scale. Although the principles for the classification and diagnosis of these disorders, founded on a multidimensional definition of disease entities, have been consolidated over the past 25 years, novel genomic data have markedly enhanced our understanding of lymphomagenesis and enriched the description of disease entities at the molecular level. Yet, the current diagnosis of lymphoid tumors is largely based on morphological assessment and immunophenotyping, with only few entities being defined by genomic criteria. This paper, which accompanies the International Consensus Classification of mature lymphoid neoplasms, will address how established assays and newly developed technologies for molecular testing already complement clinical diagnoses and provide a novel lens on disease classification. More specifically, their contributions to diagnosis refinement, risk stratification, and therapy prediction will be considered for the main categories of lymphoid neoplasms. The potential of whole-genome sequencing, circulating tumor DNA analyses, single-cell analyses, and epigenetic profiling will be discussed because these will likely become important future tools for implementing precision medicine approaches in clinical decision making for patients with lymphoid malignancies.
Collapse
Affiliation(s)
- Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Ash A. Alizadeh
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA
| | - P. Leif Bergsagel
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Phoenix, AZ
| | - Elias Campo
- Haematopathology Section, Hospital Clínic, Institut d'Investigaciones Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Andrew Davies
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jude Fitzgibbon
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Steven M. Horwitz
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ari M. Melnick
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - William G. Morice
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Ryan D. Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
- BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada
| | - Bertrand Nadel
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Stefano A. Pileri
- Haematopathology Division, IRCCS, Istituto Europeo di Oncologia, IEO, Milan, Italy
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Solna, Sweden
| | - Davide Rossi
- Institute of Oncology Research and Oncology Institute of Southern Switzerland, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Itziar Salaverria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | | | - Andrew D. Zelenetz
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Ranjana H. Advani
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
| | - Carl E. Allen
- Division of Pediatric Hematology-Oncology, Baylor College of Medicine, Houston, TX
| | | | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - James R. Cook
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Lucy B. Cook
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Francesco d’Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Stefan Dirnhofer
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Kieron Dunleavy
- Division of Hematology and Oncology, Georgetown Lombardi Comprehensive Cancer Centre, Georgetown University Hospital, Washington, DC
| | - Andrew L. Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Falko Fend
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Philippe Gaulard
- Department of Pathology, University Hospital Henri Mondor, AP-HP, Créteil, France
- Faculty of Medicine, IMRB, INSERM U955, University of Paris-Est Créteil, Créteil, France
| | - Paolo Ghia
- Università Vita-Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - John G. Gribben
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Olivier Hermine
- Service D’hématologie, Hôpital Universitaire Necker, Université René Descartes, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Daniel J. Hodson
- Wellcome MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Eric D. Hsi
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Elaine S. Jaffe
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kennosuke Karube
- Department of Pathology and Laboratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Toyko, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Wolfram Klapper
- Hematopathology Section and Lymph Node Registry, Department of Pathology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Won Seog Kim
- Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, South Korea
| | - Rebecca L. King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Young H. Ko
- Department of Pathology, Cheju Halla General Hospital, Jeju, Korea
| | | | - Georg Lenz
- Department of Medicine A, Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - José I. Martin-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Miguel A. Piris
- Department of Pathology, Jiménez Díaz Foundation University Hospital, CIBERONC, Madrid, Spain
| | - Stefania Pittaluga
- Hematopathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, NY
- Department of Pathology & Cell Biology, Columbia University, New York, NY
- The Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Scott J. Rodig
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | | | - Gilles A. Salles
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jesus San-Miguel
- Clínica Universidad de Navarra, Navarra, Cancer Center of University of Navarra, Cima Universidad de NavarraI, Instituto de Investigacion Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Céncer, Pamplona, Spain
| | - Kerry J. Savage
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | - Laurie H. Sehn
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| | - Gianpietro Semenzato
- Department of Medicine, University of Padua and Veneto Institute of Molecular Medicine, Padova, Italy
| | - Louis M. Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Steven H. Swerdlow
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Judith Trotman
- Haematology Department, Concord Repatriation General Hospital, Sydney, Australia
| | - Julie M. Vose
- Department of Internal Medicine, Division of Hematology-Oncology, University of Nebraska Medical Center, Omaha, NE
| | - Oliver Weigert
- Department of Medicine III, LMU Hospital, Munich, Germany
| | - Wyndham H. Wilson
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jane N. Winter
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Pier L. Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna Istitudo di Ematologia “Seràgnoli” and Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Università di Bologna, Bologna, Italy
| | - Emanuele Zucca
- Institute of Oncology Research and Oncology Institute of Southern Switzerland, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - David W. Scott
- Centre for Lymphoid Cancer, BC Cancer and University of British Columbia, Vancouver, Canada
| |
Collapse
|
45
|
Tumor-intrinsic causes of CAR-T failure. Blood 2022; 140:414-415. [PMID: 35925642 PMCID: PMC9353152 DOI: 10.1182/blood.2022016851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/11/2022] [Indexed: 11/20/2022] Open
|
46
|
Risk assessment with low pass whole genome sequencing of cell free DNA before CD19 CAR T-cells for large B-cell lymphoma. Blood 2022; 140:504-515. [PMID: 35512184 PMCID: PMC9353148 DOI: 10.1182/blood.2022015601] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/21/2022] [Indexed: 12/03/2022] Open
Abstract
FCS, defined by lpWGS of cfDNA, is associated with inferior outcomes in patients with LBCL treated with CAR T-cell therapy. FCS, LDH, and number of extranodal sites significantly stratified outcomes in a large single center cohort of CAR T cell–treated patients.
Patients with relapsed or refractory large B-cell lymphomas (rrLBCL) can achieve long-term remission after CD19 chimeric antigen receptor T-cell therapy (CART19). However, more than half of recipients will experience treatment failure. Thus, approaches are needed to identify high-risk patients who may benefit from alternative or consolidative therapy. We evaluated low-pass whole-genome sequencing (lpWGS) of cell-free DNA (cfDNA) before CART19 as a new approach for risk stratification. We performed lpWGS on pretreatment plasma samples from 122 patients at time of leukapheresis who received standard-of-care CART19 for rrLBCL to define DNA copy number alterations (CNAs). In multivariable selection, high focal CNA score (FCS) denoting genomic instability was the most significant pretreatment variable associated with inferior 3-month complete response rates (28% vs 56%, P = .0029), progression-free survival (PFS; P = .0007; hazard ratio, 2.11), and overall survival (OS; P = .0026; hazard ratio, 2.10). We identified 34 unique focal CNAs in 108 (89%) patients; of these, deletion 10q23.3 leading to loss of FAS death receptor was the most highly associated with poor outcomes, leading to inferior PFS (P < .0001; hazard ratio, 3.49) and OS (P = .0027; hazard ratio, 2.68). By combining FCS with traditional markers of increased tumor bulk (elevated lactate dehydrogenase and >1 extranodal site), we built a simple risk model that could reliably risk stratify patients. Thus, lpWGS of cfDNA is a minimally invasive assay that could rapidly identify high-risk patients and may guide patient selection for and targeted therapies to evaluate in future clinical trials.
Collapse
|
47
|
Lisson CS, Lisson CG, Achilles S, Mezger MF, Wolf D, Schmidt SA, Thaiss WM, Bloehdorn J, Beer AJ, Stilgenbauer S, Beer M, Götz M. Longitudinal CT Imaging to Explore the Predictive Power of 3D Radiomic Tumour Heterogeneity in Precise Imaging of Mantle Cell Lymphoma (MCL). Cancers (Basel) 2022; 14:393. [PMID: 35053554 PMCID: PMC8773890 DOI: 10.3390/cancers14020393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 02/06/2023] Open
Abstract
The study's primary aim is to evaluate the predictive performance of CT-derived 3D radiomics for MCL risk stratification. The secondary objective is to search for radiomic features associated with sustained remission. Included were 70 patients: 31 MCL patients and 39 control subjects with normal axillary lymph nodes followed over five years. Radiomic analysis of all targets (n = 745) was performed and features selected using the Mann Whitney U test; the discriminative power of identifying "high-risk MCL" was evaluated by receiver operating characteristics (ROC). The four radiomic features, "Uniformity", "Entropy", "Skewness" and "Difference Entropy" showed predictive significance for relapse (p < 0.05)-in contrast to the routine size measurements, which showed no relevant difference. The best prognostication for relapse achieved the feature "Uniformity" (AUC-ROC-curve 0.87; optimal cut-off ≤0.0159 to predict relapse with 87% sensitivity, 65% specificity, 69% accuracy). Several radiomic features, including the parameter "Short Axis," were associated with sustained remission. CT-derived 3D radiomics improves the predictive estimation of MCL patients; in combination with the ability to identify potential radiomic features that are characteristic for sustained remission, it may assist physicians in the clinical management of MCL.
Collapse
Affiliation(s)
- Catharina Silvia Lisson
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Christoph Gerhard Lisson
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Sherin Achilles
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Marc Fabian Mezger
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Visual Computing Group, Institute of Media Informatics, Ulm University, James-Franck-Ring, 89081 Ulm, Germany
| | - Daniel Wolf
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Visual Computing Group, Institute of Media Informatics, Ulm University, James-Franck-Ring, 89081 Ulm, Germany
| | - Stefan Andreas Schmidt
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Wolfgang M Thaiss
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Johannes Bloehdorn
- Department of Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ambros J Beer
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Translational Imaging "From Molecule to Man" (MoMan), Department of Internal Medicine II, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- i2SouI-Innovative Imaging in Surgical Oncology Ulm, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Comprehensive Cancer Center Ulm (CCCU), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Meinrad Beer
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Personalized Medicine (ZPM), University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Center for Translational Imaging "From Molecule to Man" (MoMan), Department of Internal Medicine II, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- i2SouI-Innovative Imaging in Surgical Oncology Ulm, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Michael Götz
- Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- Artificial Intelligence in Experimental Radiology (XAIRAD), Department of Diagnostic and Interventional Radiology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany
- German Cancer Research Center (DKFZ), Division Medical Image Computing, 69120 Heidelberg, Germany
| |
Collapse
|