1
|
Rudà R, Pellerino A, Soffietti R. Blood and cerebrospinal fluid biomarkers in neuro-oncology. Curr Opin Neurol 2024; 37:693-701. [PMID: 39329301 DOI: 10.1097/wco.0000000000001317] [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: 09/28/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the value of blood and CSF biomarkers in primary CNS tumors. RECENT FINDINGS Several analytes can be assessed with liquid biopsy techniques, including circulating tumor cells, circulating cell-free tumor DNA, circulating cell-free RNA, circulating proteins and metabolites, extracellular vesicles and tumor-educated platelets. Among diffuse gliomas of the adult, ctDNA in blood or CSF has represented the most used analyte, with the detection of molecular alterations such as MGMT promoter, PTEN, EGFRVIII, TERT promoter mutation and IDH R132H mutation. In general, CSF is enriched for ctDNA as compared with plasma. The use of MRI-guided focused ultrasounds to disrupt the blood-brain barrier could enhance the level of biomarkers in both blood and CSF. The detection of MYD88 L265P mutation with digital droplet PCR and the detection of ctDNA with next generation sequencing represent the best tools to diagnose and monitoring CNS lymphomas under treatment. In meningiomas, the low concentration of ctDNA is a limiting factor for the detection of driver mutations, such as NF2, AKTs, SMO, KLF4, TRAF7, SMARCB1, SMARCE1, PTEN, and TERT; an alternative approach could be the isolation of ctDNA through circulating extracellular vesicles. Liquid biopsies are being used extensively for diagnosis and surveillance of diffuse midline gliomas, in particular with the detection of the driver mutation H3K27M. Last, specific methylome patterns in CSF may allow the distinction of glioblastomas from CNS lymphomas or meningiomas. SUMMARY This review summarizes the current knowledge and future perspectives of liquid biopsy of blood and CSF for diagnosis and monitoring of primary CNS tumors.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University and City of Health and Science Hospital
| | - Alessia Pellerino
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University and City of Health and Science Hospital
| | | |
Collapse
|
2
|
Doubrovinskaia S, Egert A, Karschnia P, Scheffler GT, Traub BL, Galluzzo D, Huttner A, Fulbright RK, Baehring JM, Kaulen LD. Factors influencing timely diagnosis in neurolymphomatosis. J Neurooncol 2024; 170:309-317. [PMID: 39115616 DOI: 10.1007/s11060-024-04792-2] [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: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Neurolymphomatosis refers to infiltration of the peripheral nervous system (PNS) by non-Hodgkin lymphoma (NHL). Diagnostic intervals in neurolymphomatosis and factors delaying diagnosis have not been evaluated. We therefore aimed to analyze diagnostic intervals in a large cohort. METHODS The quality control database at Yale Cancer Center, Section of Neuro-Oncology, was searched for neurolymphomatosis cases diagnosed between 2001 and 2021. Univariate analyses were performed to identify parameters influencing diagnostic intervals. RESULTS We identified 22 neurolymphomatosis cases including 7 with primary and 15 with secondary disease, which occurred a median (range: 4-144) of 16 months after initial NHL diagnosis. Patients typically presented with painful polyneuropathy (73%), that was asymmetrical and rapidly progressive. Diagnosis was based on PNS biopsy (50%) or integration of neuroimaging findings (50%) with NHL history and diagnostic cerebrospinal fluid examinations. Median interval from symptom onset to diagnosis was 3 months (range: 1-12). Secondary neurolymphomatosis compared to primary disease (median 2 vs. 6 months, p = 0.02), and cases with rapidly-progressive asymmetrical neuropathy as opposed to other presentations (median 2 vs. 6 months; p < 0.001) were diagnosed earlier. Upfront conventional CT compared to other modalities (median 2 vs. 5 months p = 0.04) and nerve root localization as opposed to other disease sites (median 1.5 vs. 4 months; p = 0.04) delayed diagnosis. CONCLUSIONS NL type and localization, neuropathy course and distribution, and imaging modality selected for initial evaluation influence diagnostic intervals in neurolymphomatosis. Knowledge of this rare entity is critical for early suspicion, and diagnosis.
Collapse
Affiliation(s)
- Sofia Doubrovinskaia
- Department of Neurology, Yale School of Medicine, New Haven, USA
- Department of Neurology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
| | - Antonia Egert
- Department of Neurology, Yale School of Medicine, New Haven, USA
| | - Philipp Karschnia
- Department of Neurology, Yale School of Medicine, New Haven, USA
- Department of Neurosurgery, Ludwig Maximilian University (LMU), Munich, Germany
| | - Georg T Scheffler
- Department of Neurology, Yale School of Medicine, New Haven, USA
- Department of Neurosurgery, Ludwig Maximilian University (LMU), Munich, Germany
| | - Benjamin-Leon Traub
- Department of Neurology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Galluzzo
- Department of Neurology, Yale School of Medicine, New Haven, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, USA
| | - Robert K Fulbright
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Joachim M Baehring
- Department of Neurology, Yale School of Medicine, New Haven, USA.
- Department of Neurosurgery, Yale School of Medicine, New Haven, USA.
| | - Leon D Kaulen
- Department of Neurology, Yale School of Medicine, New Haven, USA.
- Department of Neurology, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany.
- Clinical Cooperation Unit Neuro-Oncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
3
|
Frontzek F, Renaud L, Dührsen U, Poeschel V, Bernard S, Chartier L, Ketterer N, Récher C, Fitoussi O, Held G, Casasnovas O, Haioun C, Mounier N, Tilly H, Morschhauser F, Le Gouill S, Karsten IE, Duns G, Steidl C, Scott DW, Klapper W, Rosenwald A, Ott G, Molina T, Lenz G, Ziepert M, Altmann B, Thieblemont C, Schmitz N. Identification, risk factors, and clinical course of CNS relapse in DLBCL patients across 19 prospective phase 2 and 3 trials-a LYSA and GLA/ DSHNHL collaboration. Leukemia 2024; 38:2225-2234. [PMID: 39152324 DOI: 10.1038/s41375-024-02371-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024]
Abstract
Progression or relapse in the central nervous system (CNS) remains a rare but mostly fatal event for patients with diffuse large B-cell lymphoma (DLBCL). In a retrospective analysis of 5189 patients treated within 19 prospective German and French phase 2/3 trials, we identified 159 patients experiencing a CNS event (relapse: 62%, progression: 38%). Intracerebral, meningeal, intraspinal, or combined involvement was reported in 44%, 31%, 3%, and 22% of patients, respectively. 62 of 155 evaluable patients (40%) showed concurrent systemic progression/ relapse. 82% of all CNS events occurred within two years after study inclusion or randomization. 87% of patients showed extranodal involvement outside the CNS. Patients generally had poor outcomes with a median overall survival (OS) of 3.4 months (95% CI 2.9-4.2) and a 2-year OS of 15% (10-22%). Outcomes did not differ depending on the site or time point of CNS events. Patients with isolated CNS events demonstrated significantly better OS (p = 0.023). Twenty-five patients were consolidated with autologous or allogeneic stem cell transplantation and achieved a 3-year OS of 36% (20-66%). This large study including more than 5000 DLBCL patients highlights the unmet medical need to improve the outcome of DLBCL patients suffering from CNS relapse.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/therapy
- Male
- Central Nervous System Neoplasms/therapy
- Central Nervous System Neoplasms/secondary
- Central Nervous System Neoplasms/mortality
- Central Nervous System Neoplasms/pathology
- Middle Aged
- Female
- Adult
- Aged
- Risk Factors
- Prospective Studies
- Young Adult
- Retrospective Studies
- Neoplasm Recurrence, Local/pathology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Disease Progression
- Aged, 80 and over
- Clinical Trials, Phase III as Topic
- Prognosis
- Recurrence
- Clinical Trials, Phase II as Topic
Collapse
Affiliation(s)
- Fabian Frontzek
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada.
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany.
| | - Loïc Renaud
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | - Ulrich Dührsen
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Viola Poeschel
- Department of Internal Medicine 1 (Oncology, Hematology, Clinical Immunology and Rheumatology), Saarland University Medical School, Homburg/Saar, Germany
| | - Sophie Bernard
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | | | - Nicolas Ketterer
- Centre d'Oncologie-Hématologie, Clinique Bois-Cerf, Lausanne, Switzerland
| | - Christian Récher
- Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Olivier Fitoussi
- Oncologie-Hematologie, Polyclinique Bordeaux Nord Aquitaine, Bordeaux, France
| | - Gerhard Held
- Department for Hematology and Oncology, Westpfalz - Klinikum Kaiserslautern, Kaiserslautern, Germany
| | - Olivier Casasnovas
- Service d'Hématologie Clinique, Centre Hospitalier Universitaire Dijon, Dijon, France
| | - Corinne Haioun
- Lymphoid malignancies unit-APHP, Hopital Henri Mondor, Creteil, France
| | | | - Hervé Tilly
- INSERM U1245, Centre Henri Becquerel, Rouen, France
| | | | | | - Imke E Karsten
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany
| | - Gerben Duns
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David W Scott
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
- Division of Medical Oncology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | | | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart, Germany
| | - Thierry Molina
- Universite de Paris, Assistance Publique-Hôpitaux de Paris, Hopital Necker, Anatomo-pathologie, Paris, France
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany
| | - Marita Ziepert
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Bettina Altmann
- Institute for Medical Informatics, Statistics, and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Catherine Thieblemont
- Université de Paris; Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Hemato-oncologie, Paris, France
| | - Norbert Schmitz
- Department of Medicine A, Hematology, Oncology, and Pneumonology, University Hospital of Münster, Münster, Germany.
| |
Collapse
|
4
|
Tolley ER, Lewinter C, Pedersen LM, Nielsen TH. Efficacy of intravenous high-dose methotrexate in preventing relapse to the central nervous system in R-CHOP(-like)-treated, high-risk, diffuse large B-cell lymphoma patients and its effect on mortality: a systematic review and meta-analysis. Haematologica 2024; 109:3327-3337. [PMID: 38497149 PMCID: PMC11443377 DOI: 10.3324/haematol.2023.284281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
Abstract
Central nervous system (CNS) relapse in patients with diffuse large B-cell lymphoma (DLBCL) carries a dismal prognosis and most clinical guidelines recommend CNS prophylaxis to patients deemed at high risk of CNS relapse. However, results from observational studies investigating the effect of CNS prophylaxis have yielded conflicting results. The aims of this study were to evaluate: (i) whether addition of prophylactic intravenous high-dose methotrexate (HD-MTX) reduces the risk of CNS relapse in high-risk DLBCL patients treated with R-CHOP or similar, and (ii) whether HD-MTX prophylaxis confers an overall survival benefit, irrespective of CNS relapse. We performed a systematic search of MEDLINE/PubMed and EMBASE for data on DLBCL patients at high risk of CNS relapse treated with R-CHOP or similar who received HD-MTX as an intervention and a comparator arm of patients who did not receive prophylaxis and/or intrathecal prophylaxis. A risk of bias was estimated using the ROBINS-I tool and the quality of the evidence was assessed by the GRADE approach. Finally, a meta- analysis based on the systematic review was conducted. A total of 1,812 studies were screened. No randomized controlled trials were identified. Seven observational studies comprising 1,661 patients met the inclusion criteria. We found a statistically non-significant relative risk of 0.54 (95% confidence interval: 0.27-1.07) of CNS relapse for patients receiving HD-MTX versus controls. The meta-analysis investigating mortality demonstrated a relative risk of death of 0.70 (95% confidence interval: 0.44-1.11) for patients treated with HD-MTX versus controls. The overall risk of bias was adjudged as "serious" and the quality of the evidence was rated as "low". In conclusion, our data indicate that HD-MTX does not prevent or, at best, only slightly reduces the risk of CNS relapse and confers no survival benefit.
Collapse
Affiliation(s)
- Elisabeth R Tolley
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen
| | | | - Lars M Pedersen
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen
| | - Torsten Holm Nielsen
- Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Hematology, Zealand University Hospital, Roskilde, Denmark; Danish Medicines Agency, Copenhagen.
| |
Collapse
|
5
|
Villa D. Central nervous system prophylaxis: it's time to start from scratch. Haematologica 2024; 109:3103-3104. [PMID: 38654662 PMCID: PMC11443389 DOI: 10.3324/haematol.2024.285425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Indexed: 04/26/2024] Open
Affiliation(s)
- Diego Villa
- BC Cancer Centre for Lymphoid Cancer and University of British Columbia, Vancouver.
| |
Collapse
|
6
|
Li JY, Zuo LP, Xu J, Sun CY. Clinical applications of circulating tumor DNA in hematological malignancies: From past to the future. Blood Rev 2024:101237. [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] [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
|
7
|
Gupta M, Bradley JD, Massaad E, Burns EJ, Georgantas NZ, Maron GE, Batten JM, Gallagher A, Thierauf J, Nayyar N, Gordon A, Jones SS, Pisapia M, Sun Y, Jones PS, Barker FG, Curry WT, Gupta R, Romero JM, Wang N, Brastianos PK, Martinez-Lage M, Tateishi K, Forst DA, Nahed BV, Batchelor TT, Ritterhouse LL, Iser F, Kessler T, Jordan JT, Dietrich J, Meyerson M, Cahill DP, Lennerz JK, Carter BS, Shankar GM. Rapid tumor DNA analysis of cerebrospinal fluid accelerates treatment of central nervous system lymphoma. Blood 2024; 144:1093-1100. [PMID: 38776489 PMCID: PMC11406186 DOI: 10.1182/blood.2024023832] [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: 01/03/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
ABSTRACT Delays and risks associated with neurosurgical biopsies preclude timely diagnosis and treatment of central nervous system (CNS) lymphoma and other CNS neoplasms. We prospectively integrated targeted rapid genotyping of cerebrospinal fluid (CSF) into the evaluation of 70 patients with CNS lesions of unknown cause. Participants underwent genotyping of CSF-derived DNA using a quantitative polymerase chain reaction-based approach for parallel detection of single-nucleotide variants in the MYD88, TERT promoter, IDH1, IDH2, BRAF, and H3F3A genes within 80 minutes of sample acquisition. Canonical mutations were detected in 42% of patients with neoplasms, including cases of primary and secondary CNS lymphoma, glioblastoma, IDH-mutant brainstem glioma, and H3K27M-mutant diffuse midline glioma. Genotyping results eliminated the need for surgical biopsies in 7 of 33 cases (21.2%) of newly diagnosed neoplasms, resulting in significantly accelerated initiation of disease-directed treatment (median, 3 vs 12 days; P = .027). This assay was then implemented in a Clinical Laboratory Improvement Amendments environment, with 2-day median turnaround for diagnosis of CNS lymphoma from 66 patients across 4 clinical sites. Our study prospectively demonstrates that targeted rapid CSF genotyping influences oncologic management for suspected CNS tumors.
Collapse
Affiliation(s)
- Mihir Gupta
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
- Department of Neurosurgery, University of California San Diego, La Jolla, CA
| | - Joseph D. Bradley
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Elie Massaad
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Evan J. Burns
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | | | - Garrett E. Maron
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Julie M. Batten
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Aidan Gallagher
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Julia Thierauf
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- Department of Otorhinolaryngology, Head and Neck Surgery, Experimental Head and Neck Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Naema Nayyar
- Cancer Center, Massachusetts General Hospital, Boston, MA
| | - Amanda Gordon
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - SooAe S. Jones
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Michelle Pisapia
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Ying Sun
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Pamela S. Jones
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Fred G. Barker
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - William T. Curry
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Rajiv Gupta
- Department of Neuroradiology, Massachusetts General Hospital, Boston, MA
| | - Javier M. Romero
- Department of Neuroradiology, Massachusetts General Hospital, Boston, MA
| | - Nancy Wang
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Priscilla K. Brastianos
- Cancer Center, Massachusetts General Hospital, Boston, MA
- Department of Neurology, Massachusetts General Hospital, Boston, MA
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA
| | - Maria Martinez-Lage
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA
| | - Kensuke Tateishi
- Department of Neurosurgery, Yokohama City University, Yokohama, Japan
| | | | - Brian V. Nahed
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Tracy T. Batchelor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA
| | | | - Florian Iser
- Department of Neurology and Neuro-Oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Kessler
- Department of Neurology and Neuro-Oncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Justin T. Jordan
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jorg Dietrich
- Cancer Center, Massachusetts General Hospital, Boston, MA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel P. Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | | | - Bob S. Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| | - Ganesh M. Shankar
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
8
|
Corcoran SR, Phelan JD, Choi J, Shevchenko G, Fenner RE, Yu X, Scheich S, Hsiao T, Morris VM, Papachristou EK, Kishore K, D'Santos CS, Ji Y, Pittaluga S, Wright GW, Urlaub H, Pan KT, Oellerich T, Muppidi J, Hodson DJ, Staudt LM. Molecular Determinants of Sensitivity to Polatuzumab Vedotin in Diffuse Large B-Cell Lymphoma. Cancer Discov 2024; 14:1653-1674. [PMID: 38683128 DOI: 10.1158/2159-8290.cd-23-0802] [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: 07/15/2023] [Revised: 03/12/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Polatuzumab vedotin (Pola-V) is an antibody-drug conjugate directed to the CD79B subunit of the B-cell receptor (BCR). When combined with conventional immunochemotherapy, Pola-V improves outcomes in diffuse large B-cell lymphoma (DLBCL). To identify determinants of Pola-V sensitivity, we used CRISPR-Cas9 screening for genes that modulated Pola-V toxicity for lymphomas or the surface expression of its target, CD79B. Our results reveal the striking impact of CD79B glycosylation on Pola-V epitope availability on the lymphoma cell surface and on Pola-V toxicity. Genetic, pharmacological, and enzymatic approaches that remove sialic acid from N-linked glycans enhanced lymphoma killing by Pola-V. Pola-V toxicity was also modulated by KLHL6, an E3 ubiquitin ligase that is recurrently inactivated in germinal center derived lymphomas. We reveal how KLHL6 targets CD79B for degradation in normal and malignant germinal center B cells, thereby determining expression of the surface BCR complex. Our findings suggest precision medicine strategies to optimize Pola-V as a lymphoma therapeutic. Significance: These findings unravel the molecular basis of response heterogeneity to Pola-V and identify approaches that might be deployed therapeutically to enhance the efficacy of CD79B-specific tumor killing. In addition, they reveal a novel post-translational mechanism used by normal and malignant germinal center B cells to regulate expression of the BCR. See related commentary by Leveille, p. 1577 See related article by Meriranta et al.
Collapse
Affiliation(s)
- Sean R Corcoran
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts
| | - James D Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jaewoo Choi
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Galina Shevchenko
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Rachel E Fenner
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Xin Yu
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Sebastian Scheich
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Tony Hsiao
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Vivian M Morris
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | | | - Kamal Kishore
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Clive S D'Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Yanlong Ji
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, Maryland
| | - George W Wright
- Biometrics Research Program, National Cancer Institute, NIH, Bethesda, Maryland
| | - Henning Urlaub
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Kuan-Ting Pan
- University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Thomas Oellerich
- University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Daniel J Hodson
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
9
|
Hench J, Hultschig C, Bratic Hench I, Sadasivan H, Yaldizli Ö, Hutter G, Dirnhofer S, Tzankov A, Frank S. Rapid brain lymphoma diagnostics through nanopore sequencing of cytology-negative cerebrospinal fluid. Acta Neuropathol 2024; 148:36. [PMID: 39225884 PMCID: PMC11371845 DOI: 10.1007/s00401-024-02793-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Affiliation(s)
- J Hench
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland.
| | - C Hultschig
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - I Bratic Hench
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - H Sadasivan
- AI Group, AMD and Paul G Allen School of CSE, University of Washington, Seattle, USA
| | - Ö Yaldizli
- Dept. of Neurology, University Hospital Basel, Basel, Switzerland
| | - G Hutter
- Dept. of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - S Dirnhofer
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - A Tzankov
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - S Frank
- Institute for Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
10
|
Lv J, Xu LX, Li ZX, Lin L, Wu CF, Quan TQ, Zhen ZC, Li WF, Tang LL, Mao YP, Chen L, Guo R, Zhang LL, Ai XL, Wu SY, Hao MY, Wei D, Li JB, Ma J, Chen YP, Zhou GQ, Sun Y. Longitudinal on-treatment circulating tumor DNA as a biomarker for real-time dynamic risk monitoring in cancer patients: The EP-SEASON study. Cancer Cell 2024; 42:1401-1414.e4. [PMID: 39059389 DOI: 10.1016/j.ccell.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Recurrence risks of cancer patient can change during treatment as a result of treatment-related tumor evolution. However, biomarkers that can monitor these changes are lacking. Here, we investigated whether tracking circulating tumor DNA (ctDNA) dynamics through liquid biopsy can inform real-time recurrence risk. Nasopharyngeal carcinoma (NPC) provides an ideal model where cell-free Epstein-Barr virus (EBV) DNA (cfEBV DNA), a ctDNA, can be sensitively detected. We conducted the EP-SEASON study (NCT03855020) and prospectively recruited 1,000 NPC patients undergoing per-protocol cfEBV DNA assessments at 11 time points and receiving sequential chemo-radiotherapy. Longitudinal cfEBV DNA displayed distinct patterns during neoadjuvant chemotherapy and radiotherapy. Despite the prognostic significance of cfEBV DNA at each time point, real-time recurrence risks changed in sync with cfEBV DNA dynamics. Furthermore, we identified phenotypes of whole-course ctDNA changing dynamics associated with different survival outcomes. In conclusion, tracking longitudinal on-treatment ctDNA can forecast real-time recurrence risk, facilitating risk-adapted, individualized patient management.
Collapse
Affiliation(s)
- Jiawei Lv
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ling-Xin Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Zhi-Xuan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Li Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Chen-Fei Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ting-Qiu Quan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Zi-Cheng Zhen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Wen-Fei Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ling-Long Tang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Yan-Ping Mao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Lei Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Rui Guo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Lu-Lu Zhang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin-Lei Ai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Shi-Yue Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Meng-Yu Hao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Denghui Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ji-Bin Li
- Clinical Trial Centre, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun Ma
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Yu-Pei Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Guan-Qun Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Ying Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| |
Collapse
|
11
|
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
|
12
|
Wilson MR, Cwynarski K, Eyre TA, Smith J, Chaganti S, Fox CP, McKay P. Central nervous system prophylaxis in large B-cell lymphoma: A British Society for Haematology Good Practice Paper. Br J Haematol 2024. [PMID: 39128894 DOI: 10.1111/bjh.19686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
This Good Practice Paper provides recommendations for the baseline investigation, risk stratification and use of prophylactic interventions for patients with large B-cell lymphoma at risk of central nervous system relapse. Recent evidence which has questioned the role of high-dose methotrexate in this clinical scenario is discussed in detail.
Collapse
Affiliation(s)
| | | | - Toby A Eyre
- Oxford University Hospitals NHS Trust, Churchill Cancer Centre, Oxford, UK
| | | | - Sridhar Chaganti
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | | | - Pamela McKay
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| |
Collapse
|
13
|
Fürstenau M, Giza A, Weiss J, Kleinert F, Robrecht S, Franzen F, Stumpf J, Langerbeins P, Al-Sawaf O, Simon F, Fink AM, Schneider C, Tausch E, Schetelig J, Dreger P, Böttcher S, Fischer K, Kreuzer KA, Ritgen M, Schilhabel A, Brüggemann M, Stilgenbauer S, Eichhorst B, Hallek M, Cramer P. Acalabrutinib, venetoclax, and obinutuzumab in relapsed/refractory CLL: final efficacy and ctDNA analysis of the CLL2-BAAG trial. Blood 2024; 144:272-282. [PMID: 38620072 DOI: 10.1182/blood.2023022730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/17/2024] Open
Abstract
ABSTRACT The phase 2 CLL2-BAAG trial tested the measurable residual disease (MRD)-guided triple combination of acalabrutinib, venetoclax, and obinutuzumab after optional bendamustine debulking in 45 patients with relapsed/refractory chronic lymphocytic leukemia (CLL). MRD was measured by flow cytometry (FCM; undetectable MRD <10-4) in peripheral blood (PB) and circulating tumor DNA (ctDNA) using digital droplet polymerase chain reaction of variable-diversity-joining (VDJ) rearrangements and CLL-related mutations in plasma. The median number of previous treatments was 1 (range, 1-4); 18 patients (40%) had received a Bruton tyrosine kinase inhibitor (BTKi) and/or venetoclax before inclusion, 14 of 44 (31.8%) had TP53 aberrations, and 34 (75.6%) had unmutated immunoglobulin heavy-chain variable region genes. With a median observation time of 36.3 months and all patients off-treatment for a median of 21.9 months, uMRD <10-4 in PB was achieved in 42 of the 45 patients (93.3%) at any time point, including 17 of 18 (94.4%) previously exposed to venetoclax/BTKi and 13 of 14 (92.9%) with TP53 aberrations. The estimated 3-year progression-free and overall survival rates were 85.0% and 93.8%, respectively. Overall, 585 paired FCM/ctDNA samples were analyzed and 18 MRD recurrences (5 with and 13 without clinical progression) occurred after the end of treatment. Twelve samples were first detected by ctDNA, 3 by FCM, and 3 synchronously. In conclusion, time-limited MRD-guided acalabrutinib, venetoclax, and obinutuzumab achieved deep remissions in almost all patients with relapsed/refractory CLL. The addition of ctDNA-based analyses to FCM MRD assessment seems to improve early detection of relapses. This trial was registered at www.clinicaltrials.gov as #NCT03787264.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Sulfonamides/administration & dosage
- Sulfonamides/therapeutic use
- Aged
- Middle Aged
- Female
- Male
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/blood
- Pyrazines/administration & dosage
- Pyrazines/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Neoplasm, Residual
- Benzamides/administration & dosage
- Benzamides/therapeutic use
- Adult
- Recurrence
Collapse
Affiliation(s)
- Moritz Fürstenau
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Adam Giza
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Jonathan Weiss
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Fanni Kleinert
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Sandra Robrecht
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Fabian Franzen
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Janina Stumpf
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Petra Langerbeins
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Othman Al-Sawaf
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Florian Simon
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Anna-Maria Fink
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Christof Schneider
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Eugen Tausch
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Johannes Schetelig
- Department I of Internal Medicine, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Peter Dreger
- Department V of Internal Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Böttcher
- Department III of Internal Medicine, University Hospital Rostock, Rostock, Germany
| | - Kirsten Fischer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Karl-Anton Kreuzer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Matthias Ritgen
- Department II of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Anke Schilhabel
- Department II of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Monika Brüggemann
- Department II of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stephan Stilgenbauer
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Barbara Eichhorst
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| | - Paula Cramer
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, German Chronic Lymphocytic Leukemia Study Group, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
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
|
15
|
Calimeri T, Anzalone N, Cangi MG, Fiore P, Gagliardi F, Miserocchi E, Ponzoni M, Ferreri AJM. Molecular diagnosis of primary CNS lymphoma in 2024 using MYD88 Leu265Pro and IL-10. Lancet Haematol 2024; 11:e540-e549. [PMID: 38937027 DOI: 10.1016/s2352-3026(24)00104-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 06/29/2024]
Abstract
Early diagnosis is crucial for the successful treatment of primary CNS lymphoma (PCNSL), a rapidly progressing tumour. Suspicion raised on brain MRI must be confirmed by a histopathological diagnosis of a tumour specimen collected by stereotactic biopsy. In rare cases, cerebrospinal fluid (CSF) or vitreous humour might aid in providing a cytological diagnosis. Several disease-related, patient-related, and treatment-related factors affect the timing and accuracy of diagnosis and patient outcome. Some molecules detected in CSF, aqueous and vitreous humour, and peripheral blood were proposed as diagnostic biomarkers for PCNSL; however, detection methods for most of these molecules are not yet standardised, have a long turnaround time, are expensive, and have little reproducibility among labs. By contrast, the MYD88Leu265Pro somatic hotspot mutation, revealed by PCR-based assay, is currently and reliably used during the diagnosis of some lymphomas, and IL-10, measured by enzyme-linked immunosorbent assay, is routinely used to diagnose and monitor different common metabolic and immunological diseases. Several independent studies have shown that MYD88Leu265Pro and IL-10 can be easily assessed in peripheral blood, plasma, aqueous and vitreous humour, and CSF of patients with PCNSL with substantial sensitivity and specificity, especially when evaluated in combination. In this Viewpoint, evidence supporting the routine use of MYD88Leu265Pro and IL-10 in diagnosing PCNSL is considered, and some examples of the frequent difficulties found in the diagnosis of PCNSL are provided, highlighting the role and indications of these two biomarkers to improve the timely recognition of this aggressive tumour.
Collapse
Affiliation(s)
| | - Nicoletta Anzalone
- Neuroradiology Unit, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | | | - Paolo Fiore
- Lymphoma Unit, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Elisabetta Miserocchi
- Ophthalmological Unit, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Maurilio Ponzoni
- Pathology Unit, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Andrés J M Ferreri
- Lymphoma Unit, IRCCS Ospedale San Raffaele, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
16
|
Liang JH, Wu YF, Shen HR, Li Y, Liang JH, Gao R, Hua W, Shang CY, Du KX, Xing TY, Zhang XY, Wang CX, Zhu LQ, Shao YW, Li JY, Wu JZ, Yin H, Wang L, Xu W. Clinical implications of CSF-ctDNA positivity in newly diagnosed diffuse large B cell lymphoma. Leukemia 2024; 38:1541-1552. [PMID: 38750139 DOI: 10.1038/s41375-024-02279-7] [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: 01/28/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 07/03/2024]
Abstract
The clinical implications of CSF-ctDNA positivity in newly diagnosed diffuse large B cell lymphoma (ND-DLBCL) remains largely unexplored. One hundred ND-DLBCL patients were consecutively enrolled as training cohort and another 26 ND-DLBCL patients were prospectively enrolled in validation cohort. CSF-ctDNA positivity (CSF(+)) was identified in 25 patients (25.0%) in the training cohort and 7 patients (26.9%) in the validation cohort, extremely higher than CNS involvement rate detected by conventional methods. Patients with mutations of CARD11, JAK2, ID3, and PLCG2 were more predominant with CSF(+) while FAT4 mutations were negatively correlated with CSF(+). The downregulation of PI3K-AKT signaling, focal adhesion, actin cytoskeleton, and tight junction pathways were enriched in CSF(+) ND-DLBCL. Furthermore, pretreatment CSF(+) was significantly associated with poor outcomes. Three risk factors, including high CSF protein level, high plasma ctDNA burden, and involvement of high-risk sites were used to predict the risk of CSF(+) in ND-DLBCL. The sensitivity and specificity of pretreatment CSF-ctDNA to predict CNS relapse were 100% and 77.3%. Taken together, we firstly present the prevalence and the genomic and transcriptomic landscape for CSF-ctDNA(+) DLBCL and highlight the importance of CSF-ctDNA as a noninvasive biomarker in detecting and monitoring of CSF infiltration and predicting CNS relapse in DLBCL.
Collapse
Affiliation(s)
- Jin-Hua Liang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Yi-Fan Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Hao-Rui Shen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Yue Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jun-Heng Liang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Rui Gao
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Wei Hua
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Chun-Yu Shang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Kai-Xin Du
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Tong-Yao Xing
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Xin-Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Chen-Xuan Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Liu-Qing Zhu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yang W Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Jian-Yong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Jia-Zhu Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Hua Yin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China
| | - Wei Xu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210029, China.
| |
Collapse
|
17
|
Matsuda R, Maeoka R, Morimoto T, Nakazawa T, Morisaki Y, Yokoyama S, Kotsugi M, Takeshima Y, Yamada S, Nishimura F, Park YS, Nakagawa I. Pre-treatment systemic inflammation response index and systemic immune inflammation in patients with primary central nerve system lymphoma as a useful prognostic indicator. J Neurooncol 2024; 168:487-494. [PMID: 38658464 DOI: 10.1007/s11060-024-04692-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE The systemic inflammation response index (SIRI) and systemic immune-inflammation index (SII) are based on neutrophil, monocyte, platelet, and lymphocyte counts. The SIRI and SII are used to predict the survival of patients with malignant tumors. It is well known that the inflammatory immune response is closely related to cancer occurrence and progression. In the present study, we evaluated the potential prognostic significance of SIRI and SII in patients with primary central nervous system lymphoma (PCNSL). METHODS Fifty-eight consecutive patients were enrolled in this study between November 2006 and May 2022. Among the 58 patients, 47 patients with sufficient blood test data and follow-up were analyzed. The patients with steroid intake at the time point of the blood test and higher C-reactive protein were excluded. RESULTS The median follow-up and survival times were 31 and 36 months, respectively. The optimal cutoff SIRI value was based on the receiver operating characteristic curve (ROC) for overall survival (OS) and stratified patients into low (< 1.43 × 109/L, n = 22) and high (≥ 1.43 × 109/L, n = 25) SIRI groups. The optimal cutoff SII value based on the ROC for OS stratified patients into low (< 694.9, n = 28) and high (≥ 694.9, n = 19) SII groups. A low SIRI value was associated with longer OS (p = 0.006). Furthermore, a low SII value was associated with longer OS (p = 0.044). The prognostic factors associated with prolonged survival in univariate analysis using the Cox proportional hazard model were age < 65 years, low SIRI, and low SII. The multivariate analysis demonstrated that age < 65 years and low SIRI independently predicted longer OS. CONCLUSION Simple, less expensive, and routinely ordered preoperative blood count assessments such as SIRI and SII predict the OS of patients with PCNSL. This study demonstrated that PCNSL is associated with pre-treatment systemic immune-inflammation states.
Collapse
Affiliation(s)
- Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan.
| | - Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yudai Morisaki
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Shohei Yokoyama
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Masashi Kotsugi
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Yasuhiro Takeshima
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Young-Soo Park
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8522, Japan
| |
Collapse
|
18
|
Herzi D, Le Garff-Tavernier M, Sourdeau E, Choquet S, Soussain C, Nichelli L, Mathon B, Mokhtari K, Laurenge A, Alentorn A, Boussen I, Alcantara M, Hoang-Xuan K, Houillier C. Prognostic Value of CSF IL-10 at Early Assessment of Induction Chemotherapy in Primary CNS Lymphomas: A LOC Network Study. Neurology 2024; 102:e209527. [PMID: 38830184 DOI: 10.1212/wnl.0000000000209527] [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: 06/05/2024] Open
Abstract
OBJECTIVES Despite a high response rate at the first evaluation during induction chemotherapy, the risk of early relapse remains high and unpredictable in primary CNS lymphomas (PCSNLs). We aimed to assess the prognostic value of early IL-10 levels in CSF (e-IL-10) after 2 months of induction chemotherapy. METHODS We retrospectively selected from the LOC (Lymphomes Oculo-Cérébraux) network database patients with PCSNLs who had complete or partial response at the 2-month evaluation of a high-dose methotrexate-based first-line chemotherapy for whom e-IL-10 was available. RESULTS Thirty patients (median age: 62 years, brain involvement in 30/30, CSF involvement in 10/30, median baseline CSF IL-10: 27.5 pg/mL) met the selection criteria. e-IL-10 was undetectable in 22 patients and detectable in 8 patients. At the end of induction treatment, 7 of 8 and 4 of 22 of the patients with detectable and undetectable e-IL-10 had experienced progressive disease, respectively (p = 0.001, OR: 26.8, 95% CI 2-1,478). The median progression-free survival times were 5.8 months (95% CI 2.8-8.8) and 28.7 months (95% CI 13.4-43.9) in the groups with detectable and undetectable e-IL-10, respectively (p < 0.001). DISCUSSION Our results suggest that despite an objective response, the persistence of detectable e-IL-10 is associated with a high risk of early relapse in PCNSL. A closer follow-up of such patients is warranted.
Collapse
Affiliation(s)
- Dora Herzi
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Magali Le Garff-Tavernier
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Elise Sourdeau
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Sylvain Choquet
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Carole Soussain
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Lucia Nichelli
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Bertrand Mathon
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Karima Mokhtari
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Alice Laurenge
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Agusti Alentorn
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Ines Boussen
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Marion Alcantara
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Khê Hoang-Xuan
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| | - Caroline Houillier
- From the Departments of Neurooncology (D.H., A.L., A.A., K.H.-X., C.H.), Biological Hematology (M.L.G.-T., E.S.), Clinical Hematology (S.C., I.B.), Neuro-radiology (L.N.), Neurosurgery (B.M.), and Neuropathology (K.M.), Hôpital Pitié-Salpêtrière, Paris; and Clinical Hematology (C.S., M.A.) Institut Curie, Saint Cloud, France
| |
Collapse
|
19
|
Nayak L, Bettegowda C, Scherer F, Galldiks N, Ahluwalia M, Baraniskin A, von Baumgarten L, Bromberg JEC, Ferreri AJM, Grommes C, Hoang-Xuan K, Kühn J, Rubenstein JL, Rudà R, Weller M, Chang SM, van den Bent MJ, Wen PY, Soffietti R. Liquid biopsy for improving diagnosis and monitoring of CNS lymphomas: A RANO review. Neuro Oncol 2024; 26:993-1011. [PMID: 38598668 PMCID: PMC11145457 DOI: 10.1093/neuonc/noae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND The utility of liquid biopsies is well documented in several extracranial and intracranial (brain/leptomeningeal metastases, gliomas) tumors. METHODS The RANO (Response Assessment in Neuro-Oncology) group has set up a multidisciplinary Task Force to critically review the role of blood and cerebrospinal fluid (CSF)-liquid biopsy in CNS lymphomas, with a main focus on primary central nervous system lymphomas (PCNSL). RESULTS Several clinical applications are suggested: diagnosis of PCNSL in critical settings (elderly or frail patients, deep locations, and steroid responsiveness), definition of minimal residual disease, early indication of tumor response or relapse following treatments, and prediction of outcome. CONCLUSIONS Thus far, no clinically validated circulating biomarkers for managing both primary and secondary CNS lymphomas exist. There is need of standardization of biofluid collection, choice of analytes, and type of technique to perform the molecular analysis. The various assays should be evaluated through well-organized central testing within clinical trials.
Collapse
Affiliation(s)
- Lakshmi Nayak
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Florian Scherer
- Department of Medicine I, Faculty of Medicine, Medical Center—University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Norbert Galldiks
- Department of Neurology, University of Cologne, Medical Faculty and University Hospital Cologne, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), and Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany
| | - Manmeet Ahluwalia
- Rose and Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland OH and Miami Cancer Institute, Baptist Health South Florida, International University, Miami, Florida, USA
| | - Alexander Baraniskin
- Department of Hematology, Oncology and Palliative Care, Evangelisches Krankenhaus Hamm, Hamm, Germany
| | - Louisa von Baumgarten
- Department of Neurosurgery, Ludwig-Maximilians—University of Munich, Munich, Germany
- German Cancer Consortium, Partner Site Munich, Munich, Germany
| | | | - Andrés J M Ferreri
- Università Vita-Salute San Raffaele and IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Christian Grommes
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Khê Hoang-Xuan
- APHP, Department of Neuro-oncology, Groupe Hospitalier Pitié-Salpêtrière; Sorbonne Université, Paris Brain Institute ICM, Paris, France
| | - Julia Kühn
- Department of Medicine I, Faculty of Medicine, Medical Center University of Freiburg, University of Freiburg, Freiburg, Germany
| | - James L Rubenstein
- UCSF Hematology/Oncology, Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Susan M Chang
- Department of Neurosurgery and Division of Neuro-Oncology, University of California, San Francisco, California, USA
| | | | - Patrick Y Wen
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Riccardo Soffietti
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| |
Collapse
|
20
|
Zhang X, Zhao Z, Wang R, Chen H, Zheng X, Liu L, Lan L, Li P, Wu S, Cao Q, Luo R, Hu W, Lyu S, Zhang Z, Xie D, Ye Y, Wang Y, Cai M. A multicenter proof-of-concept study on deep learning-based intraoperative discrimination of primary central nervous system lymphoma. Nat Commun 2024; 15:3768. [PMID: 38704409 PMCID: PMC11069536 DOI: 10.1038/s41467-024-48171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
Accurate intraoperative differentiation of primary central nervous system lymphoma (PCNSL) remains pivotal in guiding neurosurgical decisions. However, distinguishing PCNSL from other lesions, notably glioma, through frozen sections challenges pathologists. Here we sought to develop and validate a deep learning model capable of precisely distinguishing PCNSL from non-PCNSL lesions, especially glioma, using hematoxylin and eosin (H&E)-stained frozen whole-slide images. Also, we compared its performance against pathologists of varying expertise. Additionally, a human-machine fusion approach integrated both model and pathologic diagnostics. In external cohorts, LGNet achieved AUROCs of 0.965 and 0.972 in distinguishing PCNSL from glioma and AUROCs of 0.981 and 0.993 in differentiating PCNSL from non-PCNSL lesions. Outperforming several pathologists, LGNet significantly improved diagnostic performance, further augmented to some extent by fusion approach. LGNet's proficiency in frozen section analysis and its synergy with pathologists indicate its valuable role in intraoperative diagnosis, particularly in discriminating PCNSL from glioma, alongside other lesions.
Collapse
Affiliation(s)
- Xinke Zhang
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zihan Zhao
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ruixuan Wang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haohua Chen
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xueyi Zheng
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lili Liu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lilong Lan
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Peng Li
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shuyang Wu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qinghua Cao
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Rongzhen Luo
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Shanshan Lyu
- Department of Pathology, Guangdong Provincial People's Hospital, Guangzhou, 510080, China
| | - Zhengyu Zhang
- Department of Pathology, Nanfang Hospital, Soutern Medical University, Guangzhou, 510515, China
| | - Dan Xie
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Soutern Medical University, Guangzhou, 510515, China.
| | - Yu Wang
- Department of Pathology, Zhujiang Hospital, Soutern Medical University, Guangzhou, 510280, China.
| | - Muyan Cai
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
21
|
Cherng HJJ, Herrera A. Circulating Tumor DNA in Diffuse Large B-Cell Lymphoma: from Bench to Bedside? Curr Treat Options Oncol 2024; 25:659-678. [PMID: 38656685 DOI: 10.1007/s11864-024-01201-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
OPINION STATEMENT Diffuse large B-cell lymphoma (DLBCL) is a curable disease with variable outcomes due to underlying heterogeneous clinical and molecular features-features that are insufficiently characterized with our current tools. Due to these limitations, treatment largely remains a "one-size-fits-all" approach. Circulating tumor DNA (ctDNA) is a novel biomarker in cancers that is increasingly utilized for risk stratification and response assessment. ctDNA is readily detectable from the plasma of patients with DLBCL but has not yet been incorporated into clinical care to guide treatment. Here, we describe how ctDNA sequencing represents a promising technology in development to personalize the care of patients with DLBCL. We will review the different types of ctDNA assays being studied and the rapidly growing body of evidence supporting the utility of ctDNA in different treatment settings in DLBCL. Risk stratification by estimation of tumor burden and liquid genotyping, molecular response assessment during treatment, and monitoring for measurable residual disease (MRD) to identify therapy resistance and predict clinical relapse are all potential applications of ctDNA. It is time for clinical trials in DLBCL to utilize ctDNA as an integral biomarker for patient selection, response-adapted designs, and surrogate endpoints. As more ctDNA assays become commercially available for routine use, clinicians should consider liquid biopsy when treatment response is equivocal on imaging. Incorporating MRD may also guide decision-making if patients experience severe treatment toxicities. Though important barriers remain, we believe that ctDNA will soon be ready to transition from bench to bedside to individualize treatment for our patients with DLBCL.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/blood
- Lymphoma, Large B-Cell, Diffuse/genetics
- Humans
- Circulating Tumor DNA/blood
- Biomarkers, Tumor/blood
- Liquid Biopsy/methods
- Disease Management
- Translational Research, Biomedical
- Precision Medicine/methods
- Prognosis
- Clinical Decision-Making
- Disease Susceptibility
Collapse
Affiliation(s)
- Hua-Jay J Cherng
- Lymphoma Service, Division of Hematology & Oncology, Columbia University Irving Medical Center, 177 Fort Washington Avenue, 6GN-Rm 435, New York, NY, 10032, USA.
| | - Alex Herrera
- Division of Lymphoma, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
22
|
Natsumeda M, Shibuma S, Takahashi H, On J, Mouri Y, Tomikawa K, Fujiwara H, Watanabe J, Tsukamoto Y, Okada M, Takeda R, Shimizu H, Takizawa J, Kakita A, Oishi M. Recent advances in liquid biopsy of central nervous system lymphomas: case presentations and review of the literature. Brain Tumor Pathol 2024; 41:85-91. [PMID: 38597999 DOI: 10.1007/s10014-024-00483-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
Surgical biopsy is the gold standard for diagnosing central nervous system (CNS) lymphomas. However, reliable liquid biopsy methods for diagnosing CNS lymphomas have quickly developed and have been implicated in clinical decision-making. In the current report, we introduce two patients for whom liquid biopsy was essential for diagnosing CNS lymphomas and discuss the rapidly growing applications of this technology.
Collapse
Affiliation(s)
- Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan.
- Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Satoshi Shibuma
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Haruhiko Takahashi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jotaro On
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoshihiro Mouri
- Department of Neurosurgery, Niigata Prefectural Central Hospital, Joetsu, Japan
| | - Kaoru Tomikawa
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hidemoto Fujiwara
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jun Watanabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Rui Takeda
- Department of Hematology, Endocrinology and Metabolism, Faculty of Medicine, Niigata University, Niigata, Japan
| | - Hiroshi Shimizu
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jun Takizawa
- Department of Hematology, Endocrinology and Metabolism, Faculty of Medicine, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
23
|
Zeremski V, Adolph L, Beer S, Berisha M, Jacobs B, Kahl C, Koenecke C, Kropf S, Panse J, Petersen J, Schmidt-Hieber M, Schneider J, Vucinic V, Walter J, Weigert O, Witte HM, Mougiakakos D. Relevance of different prognostic scores in primary CNS lymphoma in the era of intensified treatment regimens: A retrospective, multicenter analysis of 174 patients. Eur J Haematol 2024; 112:641-649. [PMID: 38164819 DOI: 10.1111/ejh.14159] [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: 11/06/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVES Treatment intensification (including consolidative high-dose chemotherapy with autologous stem cell transplantation [HDT-ASCT]) significantly improved outcome in primary central nervous system lymphoma (PCNSL) patients. METHODS We conducted a multicenter, retrospective analysis of newly diagnosed PCNSL patients, treated with intensified treatment regimens. The following scores were evaluated in terms of overall survival (OS) and progression-free survival (PFS): Memorial Sloan-Kettering Cancer Center (MSKCC), International Extranodal Lymphoma Study Group (IELSG), and three-factor (3F) prognostic score. Further, all scores were comparatively investigated for model quality and concordance. RESULTS Altogether, 174 PCNSL patients were included. One hundred and five patients (60.3%) underwent HDT-ASCT. Two-year OS and 2-year PFS for the entire population were 73.3% and 48.5%, respectively. The MSKCC (p = .003) and 3F score (p < .001), but not the IELSG score (p = .06), had the discriminatory power to identify different risk groups for OS. In regard to concordance, the 3F score (C-index [0.71]) outperformed both the MSKCC (C-index [0.64]) and IELSG (C-index [0.53]) score. Moreover, the superiority of the 3F score was shown for PFS, successfully stratifying patients in three risk groups, which also resulted in the highest C-index (0.66). CONCLUSION The comparative analysis of established PCNSL risk scores affirm the clinical utility of the 3F score stratifying the widest prognostic spectrum among PCNSL patients treated with intensified treatment approaches.
Collapse
Affiliation(s)
- Vanja Zeremski
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Louisa Adolph
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Sina Beer
- Department of Hematology and Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Mirjeta Berisha
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Benedikt Jacobs
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| | - Christoph Kahl
- Department of Hematology, Oncology and Palliative Care, Klinikum Magdeburg, Magdeburg, Germany
- Department of Hematology, Oncology, and Palliative Care, University Medical Center, University of Rostock, Rostock, Germany
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Siegfried Kropf
- Department of Biometry and Medical Informatics, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Jens Panse
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Judith Petersen
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Martin Schmidt-Hieber
- Clinic of Hematology, Oncology, Pneumology and Nephrology, Carl-Thiem-Hospital Cottbus, Cottbus, Germany
| | - Jessica Schneider
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Vladan Vucinic
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, Leipzig University Medical Center, Leipzig, Germany
| | - Jeanette Walter
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Center for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf (ABCD), Aachen, Germany
| | - Oliver Weigert
- Department of Internal Medicine III, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Hanno M Witte
- Department of Hematology and Oncology, Federal Armed Hospital Ulm, Ulm, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology and Oncology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Department of Internal Medicine 5, Hematology and Clinical Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
24
|
Li W, Huang Y, Xiao M, Zhao J, Du S, Wang Z, Hu S, Yang L, Cai J. PBRM1 presents a potential ctDNA marker to monitor response to neoadjuvant chemotherapy in cervical cancer. iScience 2024; 27:109160. [PMID: 38414861 PMCID: PMC10897912 DOI: 10.1016/j.isci.2024.109160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/06/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024] Open
Abstract
Neoadjuvant chemotherapy (NACT) is a therapeutic option for locally advanced cervical cancer (LACC) patients. This study was aimed to identify potential liquid biopsy biomarkers to monitor the NACT response. Through targeted next-generation sequencing (NGS) analysis of circulating tumor DNA (ctDNA) and tumor tissue DNA (ttDNA) taken from LACC patients undergoing platinum-based NACT, 64 genes with mutations emerge during NACT in the non-responders but none in the responders. Among them, the PBRM1, SETD2, and ROS1 mutations were frequently detected in the ctDNA and ttDNA of the non-responders, and mutant PBRM1 was associated with poorer survival of patients. In vitro, PBRM1 knockdown promoted resistance to cisplatin through boosting STAT3 signaling in cervical cancer cells, while it sensitized tumor cells to poly-ADP-ribose-polymerase inhibitor olaparib. These findings suggest that mutant PBRM1 is a potential ctDNA marker of emerging resistance to NACT and of increased sensitivity to olaparib, which warrants further clinical validation.
Collapse
Affiliation(s)
- Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhui Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Man Xiao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shi Du
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sha Hu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lu Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
25
|
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
|
26
|
Heger JM, Mattlener J, Schneider J, Gödel P, Sieg N, Ullrich F, Lewis R, Bucaciuc-Mracica T, Schwarz RF, Rueß D, Ruge MI, Montesinos-Rongen M, Deckert M, Blau T, Kutsch N, Balke-Want H, Weiss J, Becker K, Reinhardt HC, Hallek M, Borchmann P, von Tresckow B, Borchmann S. Entirely noninvasive outcome prediction in central nervous system lymphomas using circulating tumor DNA. Blood 2024; 143:522-534. [PMID: 37946299 DOI: 10.1182/blood.2023022020] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT State-of-the-art response assessment of central nervous system lymphoma (CNSL) by magnetic resonance imaging is challenging and an insufficient predictor of treatment outcomes. Accordingly, the development of novel risk stratification strategies in CNSL is a high unmet medical need. We applied ultrasensitive circulating tumor DNA (ctDNA) sequencing to 146 plasma and cerebrospinal fluid (CSF) samples from 67 patients, aiming to develop an entirely noninvasive dynamic risk model considering clinical and molecular features of CNSL. Our ultrasensitive method allowed for the detection of CNSL-derived mutations in plasma ctDNA with high concordance to CSF and tumor tissue. Undetectable plasma ctDNA at baseline was associated with favorable outcomes. We tracked tumor-specific mutations in plasma-derived ctDNA over time and developed a novel CNSL biomarker based on this information: peripheral residual disease (PRD). Persistence of PRD after treatment was highly predictive of relapse. Integrating established baseline clinical risk factors with assessment of radiographic response and PRD during treatment resulted in the development and independent validation of a novel tool for risk stratification: molecular prognostic index for CNSL (MOP-C). MOP-C proved to be highly predictive of outcomes in patients with CNSL (failure-free survival hazard ratio per risk group of 6.60; 95% confidence interval, 3.12-13.97; P < .0001) and is publicly available at www.mop-c.com. Our results highlight the role of ctDNA sequencing in CNSL. MOP-C has the potential to improve the current standard of clinical risk stratification and radiographic response assessment in patients with CNSL, ultimately paving the way toward individualized treatment.
Collapse
Affiliation(s)
- Jan-Michel Heger
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
| | - Julia Mattlener
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- German Hodgkin Study Group, Cologne, Germany
| | - Jessica Schneider
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
| | - Philipp Gödel
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
| | - Noëlle Sieg
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
| | - Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung partner site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- CCCE, Essen, Germany
| | - Richard Lewis
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
| | - Teodora Bucaciuc-Mracica
- Institute for Computational Cancer Biology, Center for Integrated Oncology, Cancer Research Center Cologne Essen, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Roland F Schwarz
- Institute for Computational Cancer Biology, Center for Integrated Oncology, Cancer Research Center Cologne Essen, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | - Daniel Rueß
- Department of Stereotaxy and Functional Neurosurgery, Center of Neurosurgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Maximilian I Ruge
- Department of Stereotaxy and Functional Neurosurgery, Center of Neurosurgery, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Manuel Montesinos-Rongen
- Institute of Neuropathology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Martina Deckert
- Institute of Neuropathology, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Tobias Blau
- Institute of Neuropathology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Nadine Kutsch
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
| | - Hyatt Balke-Want
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
- Stanford Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Jonathan Weiss
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
| | - Kerstin Becker
- West German Genome Center, University of Cologne, Cologne, Germany
| | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung partner site Essen), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- CCCE, Essen, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
| | - Peter Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- German Hodgkin Study Group, Cologne, Germany
| | | | - Sven Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany
- Cancer Center Cologne Essen (CCCE), Cologne, Germany
- Cologne Lymphoma Working Group, Cologne, Germany
- German Hodgkin Study Group, Cologne, Germany
| |
Collapse
|
27
|
Lauer EM, Riegler E, Mutter JA, Alig SK, Bleul S, Kuehn J, Ranganathan L, Klingler C, Demerath T, Würtemberger U, Rau A, Weiß J, Eisenblaetter M, Bamberg F, Prinz M, Finke J, Duyster J, Illerhaus G, Diehn M, Alizadeh AA, Schorb E, Reinacher PC, Scherer F. Improved early outcome prediction by MRI-based 3D tumor volume assessment in patients with CNS lymphomas. Neuro Oncol 2024; 26:374-386. [PMID: 37713267 PMCID: PMC10836777 DOI: 10.1093/neuonc/noad177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Central nervous system lymphomas (CNSL) display remarkable clinical heterogeneity, yet accurate prediction of outcomes remains challenging. The IPCG criteria are widely used in routine practice for the assessment of treatment response. However, the value of the IPCG criteria for ultimate outcome prediction is largely unclear, mainly due to the uncertainty in delineating complete from partial responses during and after treatment. METHODS We explored various MRI features including semi-automated 3D tumor volume measurements at different disease milestones and their association with survival in 93 CNSL patients undergoing curative-intent treatment. RESULTS At diagnosis, patients with more than 3 lymphoma lesions, periventricular involvement, and high 3D tumor volumes showed significantly unfavorable PFS and OS. At first interim MRI during treatment, the IPCG criteria failed to discriminate outcomes in responding patients. Therefore, we randomized these patients into training and validation cohorts to investigate whether 3D tumor volumetry could improve outcome prediction. We identified a 3D tumor volume reduction of ≥97% as the optimal threshold for risk stratification (=3D early response, 3D_ER). Applied to the validation cohort, patients achieving 3D_ER had significantly superior outcomes. In multivariate analyses, 3D_ER was independently prognostic of PFS and OS. Finally, we leveraged prognostic information from 3D MRI features and circulating biomarkers to build a composite metric that further improved outcome prediction in CNSL. CONCLUSIONS We developed semi-automated 3D tumor volume measurements as strong and independent early predictors of clinical outcomes in CNSL patients. These radiologic features could help improve risk stratification and help guide future treatment approaches.
Collapse
Affiliation(s)
- Eliza M Lauer
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ella Riegler
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jurik A Mutter
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Divisions of Oncology and Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | | | - Sabine Bleul
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Kuehn
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lavanya Ranganathan
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Klingler
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Theo Demerath
- Department of Neuroradiology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Urs Würtemberger
- Department of Neuroradiology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Alexander Rau
- Department of Neuroradiology, Medical Center, University of Freiburg, Freiburg, Germany
- Department of Radiology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jakob Weiß
- Department of Radiology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michel Eisenblaetter
- Department of Radiology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Fabian Bamberg
- Department of Radiology, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Jürgen Finke
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gerald Illerhaus
- Department of Hematology/Oncology and Palliative Care, Klinikum Stuttgart, Stuttgart, Germany
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford School of Medicine, Stanford, CA, USA
| | - Ash A Alizadeh
- Divisions of Oncology and Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Elisabeth Schorb
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter C Reinacher
- Department of Stereotactic and Functional Neurosurgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Fraunhofer Institute for Laser Technology (ILT), Aachen, Germany
| | - Florian Scherer
- Department of Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Cite Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
Roschewski M, Hodson DJ. Diffuse large B-cell lymphoma involving the central nervous system: biologic rationale for targeted therapy. Haematologica 2024; 109:388-400. [PMID: 37706315 PMCID: PMC10828633 DOI: 10.3324/haematol.2021.278613] [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: 05/03/2023] [Accepted: 09/04/2023] [Indexed: 09/15/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive B-cell lymphoma curable even in advanced stages. DLBCL involving the central nervous system (CNS) is more difficult to cure and fewer treatment options exist. Primary CNS lymphoma (PCNSL) refers to aggressive lymphomas confined to the CNS, and are almost always DLBCL. Standard approaches for PCNSL use high-dose methotrexate-based combinations as induction therapy and younger patients often receive dose-intensive consolidation. However, dose-intensive therapies are not suitable for all patients, and older patients have fewer effective treatment options. Patients with relapsed or chemotherapy-refractory disease have a very poor prognosis. Secondary CNS lymphoma (SCNSL) describes aggressive lymphomas involving the CNS at initial presentation or relapses within the CNS after treatment for systemic DLBCL. Isolated CNS relapse is often managed as PCNSL, but patients with synchronous involvement of DLBCL in both the periphery and the CNS pose a unique clinical challenge. Insights into the molecular circuitry of DLBCL have identified distinct genetic subtypes including cases with a predilection for CNS invasion. PCNSL and subsets of SCNSL are characterized by chronically activated B-cell receptor and NFκB signaling along with genetic evidence of immune evasion which may be exploited therapeutically. Improved mechanistic understanding of targetable pathways underpinning CNS lymphomas has led to numerous clinical trials testing targeted agent combinations and immunotherapy approaches with promising early results. Biologically rational strategies may further improve the cure rate of CNS lymphomas, either by overcoming intrinsic or acquired treatment resistance and/or by being broadly applicable to patients of all ages.
Collapse
Affiliation(s)
- Mark Roschewski
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer nstitute, Bethesda, MD, 20892.
| | - Daniel J Hodson
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge.
| |
Collapse
|
29
|
D'Angelo CR. Diagnostic, Pathologic, and Therapeutic Considerations for Primary CNS Lymphoma. JCO Oncol Pract 2024; 20:195-202. [PMID: 37967301 DOI: 10.1200/op.23.00294] [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: 05/12/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/17/2023] Open
Abstract
Primary CNS lymphoma (PCNSL) is a rare lymphoma representing 3% of CNS malignancies. The diagnosis is complicated by the unique risks associated with brain biopsy, and the treatment is similarly complicated by the restriction of effective therapeutics able to cross the blood-brain barrier. Currently, the majority of individuals diagnosed with this disease are immunocompetent although immune deficiency related to HIV or immunosuppressive therapy remains an important risk factor. Improvements in both frontline therapy and consolidation options, including the use of hematopoietic stem-cell transplantation, have translated to improved survival. Unfortunately, patients experiencing relapsed or refractory disease often fare poorly. Here, we review key clinical, pathologic, and therapeutic aspects of PCNSL and highlight challenging clinical scenarios that may be encountered by the treating oncologist.
Collapse
|
30
|
Mussetti A, Fabbri N, Sureda A. CAR T-cell therapy in aggressive lymphomas-identifying prognostic and predictive markers. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:357-363. [PMID: 38066912 PMCID: PMC10727003 DOI: 10.1182/hematology.2023000436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
We discuss different pre-infusion, post-infusion and post-CAR T-cell relapse prognostic factors influencing the outcomes of anti-CD19 CAR T-cell therapy in patients with relapsed or refractory large B-cell lymphomas. Despite the overall positive results of anti-CD19 CAR T-cell therapy, a significant percentage of patients relapse. We summarize the efforts made to identify predictive factors for response and durable remissions and survival. In the pre-infusion setting, the patient-related factors discussed include Eastern Cooperative Oncology Group performance status, age, and comorbidities. Disease-related factors like tumor burden, histology, and biological features are also considered. In addition, inflammation-related factors and CAR T-cell product-related factors are considered. After CAR T-cell infusion, factors such as disease response assessed by 18FDG-PET/CT scan, liquid biopsy monitoring, and CAR T-cell expansion become crucial in predicting survival outcomes. Response to 18FDG-PET/CT scan is a widely used test for confirming response and predicting survival. Liquid biopsy, in combination with 18FDG-PET/CT scan, has shown potential in predicting outcomes. CAR T-cell expansion and persistence have shown mixed effects on survival, with some studies indicating their association with response. In the setting of post-CAR T-cell relapse, prognostic factors include refractory disease, time of relapse, and elevated lactate dehydrogenase levels at CAR T-cell infusion. Enrollment in clinical trials is crucial for improving outcomes in these patients. Overall, we discuss a comprehensive overview of prognostic factors that can influence the outcomes of anti-CD19 CAR T-cell therapy in patients with relapsed or refractory large B-cell lymphomas, highlighting the need for personalized approaches in treatment decision-making.
Collapse
Affiliation(s)
- Alberto Mussetti
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Nicole Fabbri
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Anna Sureda
- Department of Hematology, Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Medicine Department, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Foster JB, Koptyra MP, Bagley SJ. Recent Developments in Blood Biomarkers in Neuro-oncology. Curr Neurol Neurosci Rep 2023; 23:857-867. [PMID: 37943477 DOI: 10.1007/s11910-023-01321-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 11/10/2023]
Abstract
PURPOSE OF REVIEW Given the invasive and high-risk nature of brain surgery, the need for non-invasive biomarkers obtained from the peripheral blood is greatest in tumors of the central nervous system (CNS). In this comprehensive review, we highlight recent advances in blood biomarker development for adult and pediatric brain tumors. RECENT FINDINGS We summarize recent blood biomarker development for CNS tumors across multiple key analytes, including peripheral blood mononuclear cells, cell-free DNA, cell-free RNA, proteomics, circulating tumor cells, and tumor-educated platelets. We also discuss methods for enhancing blood biomarker detection through transient opening of the blood-brain barrier. Although blood-based biomarkers are not yet used in routine neuro-oncology practice, this field is advancing rapidly and holds great promise for improved and non-invasive management of patients with brain tumors. Prospective and adequately powered studies are needed to confirm the clinical utility of any blood biomarker prior to widespread clinical implementation.
Collapse
Affiliation(s)
- Jessica B Foster
- Division of Oncology, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mateusz P Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephen J Bagley
- Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, 10th Floor Perelman Center, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
Cuzzo B, Lipsky A, Cherng HJJ. Measurable Residual Disease Monitoring in Lymphoma. Curr Hematol Malig Rep 2023; 18:292-304. [PMID: 37930608 DOI: 10.1007/s11899-023-00715-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/07/2023]
Abstract
PURPOSE OF REVIEW The utility of analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and disease in the bone marrow as an adjunctive tool in caring for hematologic cancer patients is expanding. This holds true for lymphoma where these biomarkers are being explored as a means of genotyping and quantifying disease. Regarding the latter, they can be used to monitor measurable residual disease (MRD) during and after treatment. This holds potential for aiding clinical decisions amidst treatment, detecting earlier relapse, and improving prognostication. Here, we review the evidence to support these applications in a variety of lymphoma subtypes. RECENT FINDINGS Numerous clinical trials across a variety of lymphomas have demonstrated value in MRD monitoring. MRD monitoring is often prognostic for progression free survival (PFS) and even overall survival (OS) at several time points in a disease course, particularly when utilizing serial measurements. With regards to tailoring treatment, there are a growing number of trials examining MRD-adaptive treatment strategies to intensify or de-escalate treatment to individualize care. Lastly, MRD monitoring has been utilized successfully in detecting earlier relapse when compared to more standard methods of clinical surveillance such as radiographic assessment. Although not routinely implemented into clinical practice, MRD monitoring in lymphoma is helping shape the future landscape of this disease by aiding in prognostication, guiding therapy, and detecting earlier relapse. Steps to standardize and further examine this technology prospectively are being taken to bring MRD monitoring to the forefront of the field.
Collapse
Affiliation(s)
- Brian Cuzzo
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Andrew Lipsky
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Hua-Jay J Cherng
- Columbia University Medical Center, 161 Fort Washington Ave, New York, NY, 10032, USA.
| |
Collapse
|
33
|
Alderuccio JP, Nayak L, Cwynarski K. How I treat secondary CNS involvement by aggressive lymphomas. Blood 2023; 142:1771-1783. [PMID: 37702537 PMCID: PMC10862244 DOI: 10.1182/blood.2023020168] [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: 05/15/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023] Open
Abstract
Secondary central nervous system (CNS) lymphoma (SCNSL) is a rare but clinically challenging scenario with historically disappointing outcomes. SCNSL refers to lymphoma that has spread into the CNS concurrently with systemic disease or CNS relapse during or after frontline immunochemotherapy, presenting with or without systemic lymphoma. Diffuse large B-cell lymphoma (DLBCL) denotes the most common entity, but an increased incidence is observed in other histologies, such as Burkitt lymphoma and mantle-cell lymphoma. The incidence, timing in disease course, location, evidence supporting the use of CNS prophylaxis, and treatment pathways vary according to histology. No randomized data exist to delineate the best treatment approaches with current recommendations based on retrospective and single-arm studies. However, a regimen comprising immunochemotherapy, incorporating agents that cross the blood-brain barrier, followed by thiotepa-containing conditioning and autologous stem-cell transplant outlined in the international MARIETTA study demonstrated improvement in outcomes, representing a major accomplishment in the care of patients with DLBCL with SCNSL. Anti-CD19 chimeric antigen receptor T cell denotes a paradigm shift in the treatment of patients with systemic aggressive lymphomas, with emerging data also demonstrating efficacy without higher neurotoxicity in those with SCNSL. In this manuscript we discuss 5 clinical scenarios and review the evidence supporting our recommendations.
Collapse
Affiliation(s)
- Juan Pablo Alderuccio
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Kate Cwynarski
- Department of Haematology, University College London Hospital, London, United Kingdom
| |
Collapse
|
34
|
Pouyiourou M, Kraft BN, Wohlfromm T, Stahl M, Kubuschok B, Löffler H, Hacker UT, Hübner G, Weiss L, Bitzer M, Ernst T, Schütt P, Hielscher T, Delorme S, Kirchner M, Kazdal D, Ball M, Kluck K, Stenzinger A, Bochtler T, Krämer A. Nivolumab and ipilimumab in recurrent or refractory cancer of unknown primary: a phase II trial. Nat Commun 2023; 14:6761. [PMID: 37875494 PMCID: PMC10598029 DOI: 10.1038/s41467-023-42400-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
Cancer of unknown primary has a dismal prognosis, especially following failure of platinum-based chemotherapy. 10-20% of patients have a high tumor mutational burden (TMB), which predicts response to immunotherapy in many cancer types. In this prospective, non-randomized, open-label, multicenter Phase II trial (EudraCT 2018-004562-33; NCT04131621), patients relapsed or refractory after platinum-based chemotherapy received nivolumab and ipilimumab following TMBhigh vs. TMBlow stratification. Progression-free survival (PFS) represented the primary endpoint; overall survival (OS), response rates, duration of clinical benefit and safety were the secondary endpoints. The trial was prematurely terminated in March 2021 before reaching the preplanned sample size (n = 194). Among 31 evaluable patients, 16% had a high TMB ( > 12 mutations/Mb). Overall response rate was 16% (95% CI 6-34%), with 7.7% (95% CI 1-25%) vs. 60% (95% CI 15-95%) in TMBlow and TMBhigh, respectively. Although the primary endpoint was not met, high TMB was associated with better median PFS (18.3 vs. 2.4 months) and OS (18.3 vs. 3.6 months). Severe immune-related adverse events were reported in 29% of cases. Assessing on-treatment dynamics of circulating tumor DNA using combined targeted hotspot mutation and shallow whole genome sequencing as part of a predefined exploratory analysis identified patients benefiting from immunotherapy irrespective of initial radiologic response.
Collapse
Affiliation(s)
- Maria Pouyiourou
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | - Bianca N Kraft
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Timothy Wohlfromm
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Michael Stahl
- Department of Medical Oncology, Evangelische Kliniken Essen-Mitte, Essen, Germany
| | - Boris Kubuschok
- Department of Internal Medicine II, Augsburg University Medical Center and Bavarian Cancer Research Center (BZKF), Partner Cite Augsburg, Augsburg, Germany
| | - Harald Löffler
- Department of Internal Medicine III, Marienhospital Stuttgart, Stuttgart, Germany
| | - Ulrich T Hacker
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Gerdt Hübner
- Department of Internal Medicine III, Ameos Krankenhausgesellschaft Ostholstein, Eutin, Germany
| | - Lena Weiss
- Department of Internal Medicine, Comprehensive Cancer Center, University of Munich, Munich, Germany
| | - Michael Bitzer
- Department of Gastroenterology, Hepatology and Infectiology, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Ernst
- Department of Internal Medicine II, Jena University Hospital, Jena, Germany
| | | | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Delorme
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Kirchner
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), University of Heidelberg, Heidelberg, Germany
| | - Daniel Kazdal
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), University of Heidelberg, Heidelberg, Germany
| | - Markus Ball
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), University of Heidelberg, Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), University of Heidelberg, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Center for Personalized Medicine (ZPM), University of Heidelberg, Heidelberg, Germany
| | - Tilmann Bochtler
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
35
|
Rozenblum L, Galanaud D, Houillier C, Soussain C, Baptiste A, Belin L, Edeline V, Naggara P, Soret M, Causse-Lemercier V, Willems L, Choquet S, Ursu R, Hoang-Xuan K, Kas A. [18F]FDG PET-MRI provides survival biomarkers in primary central nervous system lymphoma in the elderly: an ancillary study from the BLOCAGE trial of the LOC network. Eur J Nucl Med Mol Imaging 2023; 50:3684-3696. [PMID: 37462774 DOI: 10.1007/s00259-023-06334-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE Primary central nervous system lymphoma (PCNSL) incidence is rising among elderly patients, presenting challenges due to poor prognosis and treatment-related toxicity risks. This study explores the potential of combining [18F]fluorodeoxyglucose ([18F]FDG) PET scans and multimodal MRI for improving management in elderly patients with de novo PCNSL. METHODS Immunocompetent patients over 60 years with de novo PCNSL were prospectively enrolled in a multicentric study between January 2016 and April 2021. Patients underwent brain [18F]FDG PET-MRI before receiving high-dose methotrexate-based chemotherapy. Relationships between extracted PET (metabolic tumor volume (MTV), sum of MTV for up to five lesions (sumMTV), metabolic imaging lymphoma aggressiveness score (MILAS)) and MRI parameters (tumor contrast-enhancement size, cerebral blood volume (CBV), cerebral blood flow (CBF), apparent diffusion coefficient (ADC)) and treatment response and outcomes were analyzed. RESULTS Of 54 newly diagnosed diffuse large B-cell PCNSL patients, 52 had positive PET and MRI with highly [18F]FDG-avid and contrast-enhanced disease (SUVmax: 27.7 [22.8-36]). High [18F]FDG uptake and metabolic volume were significantly associated with low ADCmean values and high CBF at baseline. Among patients, 69% achieved an objective response at the end of induction therapy, while 17 were progressive. Higher cerebellar SUVmean and lower sumMTV at diagnosis were significant predictors of complete response: 6.4 [5.7-7.7] vs 5.4 [4.5-6.6] (p = 0.04) and 5.5 [2.1-13.3] vs 15.9 [4.2-19.5] (p = 0.01), respectively. Two-year overall survival (OS) was 71%, with a median progression-free survival (PFS) of 29.6 months and a median follow-up of 37 months. Larger tumor volumes on PET or enhanced T1-weighted MRI were significant predictors of poorer OS, while a high MILAS score at diagnosis was associated with early death (< 1 year). CONCLUSION Baseline cerebellar metabolism and sumMTV may predict response to end of chemotherapy in PCNSL. Tumor volume and MILAS at baseline are strong prognostic factors.
Collapse
Affiliation(s)
- Laura Rozenblum
- Department of Nuclear Medicine, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France.
- Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris, France.
| | - Damien Galanaud
- Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris, France
- Department of Neuroradiology, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Caroline Houillier
- Deparrment of Neurology 2 Mazarin, APHP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, Paris, France
| | - Carole Soussain
- Department of Hematology, Institut Curie, Site Saint-Cloud and INSERM U932 Institut Curie, Université PSL, 75005, Paris, France
| | - Amandine Baptiste
- Department of Public Health, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière - Charles Foix, Paris, France
| | - Lisa Belin
- Department of Public Health, Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, Hôpitaux Universitaires Pitié-Salpêtrière - Charles Foix, Paris, France
| | | | - Philippe Naggara
- Department of Nuclear Medicine, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Marine Soret
- Department of Nuclear Medicine, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris, France
| | - Valérie Causse-Lemercier
- Department of Nuclear Medicine, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Lise Willems
- Department of Hematology, Cochin Hospital, APHP, Paris, France
| | - Sylvain Choquet
- Department of Hematology, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Renata Ursu
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, Service de Neurologie, Paris, France
| | - Khê Hoang-Xuan
- Deparrment of Neurology 2 Mazarin, APHP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, Paris, France
| | - Aurélie Kas
- Department of Nuclear Medicine, Groupe Hospitalier Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
- Sorbonne Université, INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, LIB, Paris, France
| |
Collapse
|
36
|
Xian RR, Ambinder RF. Cell-Free Circulating Tumor DNA and Epstein-Barr Virus DNA for Early Diagnosis of Epstein-Barr Virus-Associated Cancers. J Clin Oncol 2023; 41:4290-4292. [PMID: 37478392 DOI: 10.1200/jco.23.00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 04/11/2023] [Indexed: 07/23/2023] Open
Affiliation(s)
- Rena R Xian
- Department of Pathology, Johns Hopkins University, School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Richard F Ambinder
- Department of Oncology, Johns Hopkins University, School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
37
|
Bobillo S, Wilson MR, Cwynarski K. Controversies in central nervous system prophylaxis of high-risk diffuse large B-cell lymphoma. Curr Opin Oncol 2023; 35:382-388. [PMID: 37551947 DOI: 10.1097/cco.0000000000000973] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
PURPOSE OF REVIEW Central nervous system (CNS) relapse in patients with diffuse large B-cell lymphoma (DLBCL) is an uncommon but devastating complication with an overall survival of less than 6 months. This article will review the recent updates on CNS prophylaxis including new potential advances in the identification of high-risk patients. RECENT FINDINGS The identification of patients at a high risk of CNS relapse is based on clinical and biological features has improved over recent years; however, the of different CNS prophylaxis strategies including intrathecal chemotherapy and high-dose methotrexate have been recently questioned in several large retrospective studies. The analysis of cell-free circulating tumor DNA (ctDNA) in the cerebrospinal fluid has been shown to identify patients with a high risk of CNS involvement and work is ongoing to identify how this can be used as a prognostic biomarker. SUMMARY Recent clinical retrospective data have questioned the effectiveness of intrathecal and high-dose methotrexate in the prevention of CNS relapse in high-risk DLBCL patients. The role of more sensitive methods to detect CNS involvement and the benefit of novel therapies in CNS relapse prevention are currently under evaluation.
Collapse
Affiliation(s)
- Sabela Bobillo
- Department of Haematology, Vall d'Hebron Institute of Oncology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Matthew R Wilson
- Department of Haematology, Beatson West of Scotland Cancer Centre, Glasgow
| | - Kate Cwynarski
- Department of Haematology, University College London Hospital, London, UK
| |
Collapse
|
38
|
Calimeri T, Steidl C, Fiore P, Ferreri AJM. New hopes in relapsed refractory primary central nervous system lymphoma. Curr Opin Oncol 2023; 35:364-372. [PMID: 37551946 DOI: 10.1097/cco.0000000000000980] [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: 08/09/2023]
Abstract
PURPOSE OF REVIEW Patients with relapsed/refractory primary central nervous system lymphoma (rrPCNSL) have poor prognosis, with a median survival after relapse of 6.8 months. In this review, we discuss the evolving landscape and the possible future directions related to this important unmet clinical need. RECENT FINDINGS The modern two-phase approach for newly diagnosed PCNSL based on an induction using high-dose methotrexate (HD-MTX) combinations and a subsequent consolidation, has significantly improved the outcome in this setting. However, this strategy is able to cure more or less 50% of patients. rrPCNSL patients have a very poor prognosis with a reported 5-year overall survival of 18%. Late relapses (after third year) and use of high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT) represent important factors associated with a better outcome in this setting. On the basis of the growing acquisition of knowledge on the molecular characteristics of PCNSL, the use of non-chemotherapeutic drugs such as bruton tyrosine kinase inhibitors (BTK-is), immunomodulatory drugs (IMiDs) and immune checkpoint blockers (ICBs) is increasing in the last years along with the introduction of novel approaches (CAR-T cells and blood--brain barrier disruption). However, despite high responses in some cases, durations are often short, translating in outcome results still unsatisfactory. SUMMARY Treatment of rrPCNSL patients is challenging. As no standard of care exist in this setting, it is of paramount importance to acquire new knowledge related to this condition and start multidisciplinary collaboration in order to improve pts outcome.
Collapse
Affiliation(s)
| | | | - Paolo Fiore
- Lymphoma Unit, IRCCS San Raffaele Scientific Institute
- University 'Vita-Salute San Raffaele', Milan, Italy
| | | |
Collapse
|
39
|
Foerster AK, Lauer EM, Scherer F. Clinical applications of circulating tumor DNA in central nervous system lymphoma. Semin Hematol 2023; 60:150-156. [PMID: 37442670 DOI: 10.1053/j.seminhematol.2023.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Detection and characterization of circulating tumor DNA (ctDNA) in body fluids have the potential to revolutionize management of patients with lymphoma. Minimal access to malignant DNA through a simple blood draw or lumbar puncture is particularly appealing for CNS lymphomas (CNSL), which cannot be easily or repeatedly sampled without invasive surgeries. Profiling of ctDNA provides a real-time snapshot of the genetic composition in patients with CNSL and enables ultrasensitive quantification of lymphoma burden at any given time point during the course of the disease. Here, we broadly review technical challenges of ctDNA identification in CNSL, recent advances of innovative liquid biopsy technologies, potential clinical applications of ctDNA and how it may improve CNSL risk stratification, outcome prediction, and monitoring of measurable residual disease. Finally, we discuss clinical trials and scenarios in which ctDNA could be implemented to guide risk-adapted and personalized treatment decisions.
Collapse
Affiliation(s)
- Anna Katharina Foerster
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Eliza M Lauer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Florian Scherer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK) partner site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
40
|
Sworder BJ, Kurtz DM. Cell-free DNA in large B-cell lymphoma: MRD and beyond. Semin Hematol 2023; 60:142-149. [PMID: 37474409 PMCID: PMC10528139 DOI: 10.1053/j.seminhematol.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/31/2023] [Accepted: 06/24/2023] [Indexed: 07/22/2023]
Abstract
Large B-cell lymphomas (LBCLs) are a strikingly diverse set of diseases, including clinical, biological, and molecular heterogeneity. Despite a wealth of information resolving this heterogeneity in the research setting, applying molecular features routinely in the clinic remains challenging. The advent of circulating tumor DNA (ctDNA) liquid biopsies promises to unlock additional molecular information in the clinic, including mutational genotyping, molecular classification, and minimal residual disease detection. Here, we examine the technologies, applications, and studies exploring the utility of ctDNA in LBCLs.
Collapse
Affiliation(s)
- Brian J Sworder
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
| | - David M Kurtz
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA; Stanford Cancer Institute, Stanford University, Stanford, CA.
| |
Collapse
|
41
|
Meriranta L, Pitkänen E, Leppä S. Blood has never been thicker: Cell-free DNA fragmentomics in the liquid biopsy toolbox of B-cell lymphomas. Semin Hematol 2023; 60:132-141. [PMID: 37455222 DOI: 10.1053/j.seminhematol.2023.06.006] [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: 03/20/2023] [Revised: 05/30/2023] [Accepted: 06/24/2023] [Indexed: 07/18/2023]
Abstract
Liquid biopsies utilizing plasma circulating tumor DNA (ctDNA) are anticipated to revolutionize decision-making in cancer care. In the field of lymphomas, ctDNA-based blood tests represent the forefront of clinically applicable tools to harness decades of genomic research for disease profiling, quantification, and detection. More recently, the discovery of nonrandom fragmentation patterns in cell-free DNA (cfDNA) has opened another avenue of liquid biopsy research beyond mutational interrogation of ctDNA. Through examination of structural features, nucleotide content, and genomic distribution of massive numbers of plasma cfDNA molecules, the study of fragmentomics aims at identifying new tools that augment existing ctDNA-based analyses and discover new ways to profile cancer from blood tests. Indeed, the characterization of aberrant lymphoma ctDNA fragment patterns and harnessing them with powerful machine-learning techniques are expected to unleash the potential of nonmutant molecules for liquid biopsy purposes. In this article, we review cfDNA fragmentomics as an emerging approach in the ctDNA research of B-cell lymphomas. We summarize the biology behind the formation of cfDNA fragment patterns and discuss the preanalytical and technical limitations faced with current methodologies. Then we go through the advances in the field of lymphomas and envision what other noninvasive tools based on fragment characteristics could be explored. Last, we place fragmentomics as one of the facets of ctDNA analyses in emerging multiview and multiomics liquid biopsies. We pay attention to the unknowns in the field of cfDNA fragmentation biology that warrant further mechanistic investigation to provide rational background for the development of these precision oncology tools and understanding of their limitations.
Collapse
Affiliation(s)
- Leo Meriranta
- Applied Tumor Genomics, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Esa Pitkänen
- Applied Tumor Genomics, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland; Institute for Molecular Medicine Finland (FIMM), HILIFE, Helsinki, Finland
| | - Sirpa Leppä
- Applied Tumor Genomics, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
42
|
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
|
43
|
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: 2] [Impact Index Per Article: 2.0] [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
|
44
|
Bravetti C, Degaud M, Armand M, Sourdeau E, Mokhtari K, Maloum K, Osman J, Verrier P, Houillier C, Roos-Weil D, Soussain C, Choquet S, Hoang-Xuan K, Le Garff-Tavernier M, Denis JA, Davi F. Combining MYD88 L265P mutation detection and clonality determination on CSF cellular and cell-free DNA improves diagnosis of primary CNS lymphoma. Br J Haematol 2023. [PMID: 36941788 DOI: 10.1111/bjh.18758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/22/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023]
Abstract
Diagnosis of primary central nervous system lymphoma (PCNSL) is challenging, and although brain biopsy remains the gold standard, cerebrospinal fluid (CSF) constitutes a less invasive source of lymphomatous biomarkers. In a retrospective cohort of 54 PCNSL cases tested at diagnosis or relapse, we evaluated the contribution of immunoglobulin heavy chain (IGH) gene clonality and MYD88 L265P detection on both CSF cell pellets and supernatants, in comparison with cytology, flow cytometry, interleukin (IL)-10 and IL-6 quantification. Clonality assessment included a new assay to detect partial IGH-DJ rearrangements. Clonal IGH rearrangements and/or MYD88 L265P mutation were detected in 27 (50%) cell pellets and 24 (44%) supernatant cell-free (cf) DNA. Combining analyses on both compartments, 36 (66%) cases had at least one detectable molecular marker, present only in cfDNA for 9 (16%) of them. While cytology and flow cytometry were positive in only 7 (13.0%) and 9 (17.3%) cases respectively, high IL-10 levels were observed in 36 (66.7%) cases. Overall, taking into account molecular and cytokine results, 46/54 (85%) cases had at least one lymphomatous biomarker detectable in the CSF. These results show that this combination of biomarkers evaluated on both cell pellet and supernatant CSF fractions improves significantly the biological diagnosis of PCNSL.
Collapse
Affiliation(s)
- Clotilde Bravetti
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Michaël Degaud
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Marine Armand
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Elise Sourdeau
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Karima Mokhtari
- Department of Neuropathology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Karim Maloum
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Jennifer Osman
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Patricia Verrier
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Caroline Houillier
- Department of Neurology-2, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), IHU, ICM, Sorbonne Université, Paris, France
| | - Damien Roos-Weil
- Department of Clinical Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Carole Soussain
- Division of Hematology, Institut Curie, Site Saint-Cloud, and INSERM U932, PSL Research University, Paris, France
| | - Sylvain Choquet
- Department of Clinical Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Khe Hoang-Xuan
- Department of Neurology-2, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP), IHU, ICM, Sorbonne Université, Paris, France
| | - Magali Le Garff-Tavernier
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| | - Jérôme Alexandre Denis
- Department of Endocrine and Oncological Biochemistry, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Centre de recherche Saint-Antoine (UMR_S 938), Biologie et thérapeutiques du cancer, Paris, France
| | - Frédéric Davi
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) and Sorbonne Université, Paris, France
| |
Collapse
|