1
|
Gajzer DC, Fromm JR. Flow Cytometry for B-Cell Non-Hodgkin and Hodgkin Lymphomas. Cancers (Basel) 2025; 17:814. [PMID: 40075660 PMCID: PMC11898643 DOI: 10.3390/cancers17050814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
Multi-parametric flow cytometry is a powerful diagnostic tool that permits rapid assessment of cellular antigen expression to quickly provide immunophenotypic information suitable for disease classification. This chapter describes the classification of B-cell non-Hodgkin lymphoma (B-NHL) by flow cytometry suitable for the clinical and research environment. In addition to describing the immunophenotypic patterns of the most common B-NHL (including examples of common B-NHL), the effect of anti-CD19, -CD20, and -CD38 therapies on the evaluation of flow cytometric data is also discussed. Over the last 15 years, our laboratory has developed flow cytometry combinations that can immunophenotype classic Hodgkin lymphoma (CHL), nodular lymphocyte predominant Hodgkin lymphoma (NLPHL), and T-cell/histocyte-rich large B-cell lymphoma (THRLBCL) and the use of these assays will be presented. The CHL assay combination is also particularly well suited to immunophenotype primary mediastinal large B-cell lymphoma (PMLBCL) and our experience immunophenotyping PMLBCL by flow cytometry will be discussed. Finally, an approach to the evaluation of the reactive infiltrate of CHL, NLPHL, and THRLBCL that can provide diagnostic information will also be provided.
Collapse
Affiliation(s)
| | - Jonathan R. Fromm
- Department of Laboratory Medicine and Pathology, University of Washington, 825 Eastlake Ave E, Seattle, WA 98109, USA;
| |
Collapse
|
2
|
Kutsch N, Robrecht S, Fink A, Lange E, Weide R, Kiehl MG, Sökler M, Schlag R, Vehling‐Kaiser U, Köchling G, Plöger C, Gregor M, Plesner T, Clausen MR, Oschlies I, Ritgen M, Herling M, Fischer K, Döhner H, Wendtner C, Kreuzer K, Stilgenbauer S, Hallek M, Böttcher S, Klapper W, Eichhorst B. The role of trephine bone marrow biopsies in the era of measurable residual disease -Results from the CLL10 trial of the German CLL Study Group (GCLLSG). Hemasphere 2024; 8:e126. [PMID: 39050548 PMCID: PMC11267170 DOI: 10.1002/hem3.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Affiliation(s)
- Nadine Kutsch
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Sandra Robrecht
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Anna Fink
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Elisabeth Lange
- Department of Hematology and OncologyEvangelical Hospital HammHammGermany
| | - Rudolf Weide
- Practice for Hematology and OncologyKoblenzGermany
| | - Michael G. Kiehl
- Department of Internal MedicineFrankfurt (Oder) General HospitalFrankfurt/OderGermany
| | - Martin Sökler
- Department II of Internal MedicineUniversity Hospital TübingenTübingenGermany
- Department of Hematology and OncologyHospital STS AG ThunThunSwitzerland
| | | | | | | | | | - Michael Gregor
- Division of HematologyCantonal Hospital of LucerneLucerneSwitzerland
| | - Torben Plesner
- Department of Hematology and University of Southern DenmarkVejle HospitalVejleDenmark
| | - Michael R. Clausen
- Department of Hematology and University of Southern DenmarkVejle HospitalVejleDenmark
| | - Ilske Oschlies
- Hematopathology SectionChristian‐Albrechts‐University KielKielGermany
| | - Matthias Ritgen
- Department II of Internal MedicineUniversity of Schleswig‐HolsteinKielGermany
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
- Department of Hematology, Cellular Therapy, Hemostaseology and InfectiologyUniversity Hospital LeipzigLeipzigGermany
| | - Kirsten Fischer
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Hartmut Döhner
- Department III of Internal MedicineUniversity Hospital of UlmUlmGermany
| | | | - Karl‐Anton Kreuzer
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Stephan Stilgenbauer
- Department of Hematology and OncologyHospital STS AG ThunThunSwitzerland
- Department of Hematology, Cellular Therapy, Hemostaseology and InfectiologyUniversity Hospital LeipzigLeipzigGermany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| | - Sebastian Böttcher
- Department III of Internal Medicine Medicine—Hematology/Oncology/Palliative CareRostock University Medical CenterRostockGermany
| | - Wolfram Klapper
- Hematopathology SectionChristian‐Albrechts‐University KielKielGermany
| | - Barbara Eichhorst
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf and German CLL Study GroupUniversity of CologneCologneGermany
| |
Collapse
|
3
|
Anderson MA, Bennett R, Badoux X, Best G, Chia N, Cochrane T, Cull G, Crassini K, Harrup R, Jackson S, Kuss B, Lasica M, Lew TE, Marlton P, Opat S, Palfreyman E, Polizzotto MN, Ratnasingam S, Seymour JF, Soosapilla A, Talaulikar D, Tam CS, Weinkove R, Wight J, Mulligan SP. Chronic lymphocytic leukaemia Australasian consensus practice statement. Intern Med J 2023; 53:1678-1691. [PMID: 37743239 DOI: 10.1111/imj.16207] [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: 11/30/2022] [Accepted: 07/30/2023] [Indexed: 09/26/2023]
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common haematological malignancy in Australia and New Zealand (ANZ). Considerable changes to diagnostic and management algorithms have occurred within the last decade. The availability of next-generation sequencing and measurable residual disease assessment by flow cytometry allow for advanced prognostication and response assessments. Novel therapies, including inhibitors of Bruton's tyrosine kinase (BTKi) and B-cell lymphoma 2 (BCL2) inhibitors, have transformed the treatment landscape for both treatment-naïve and relapsed/refractory disease, particularly for patients with high-risk genetic aberrations. Recommendations regarding appropriate supportive management continue to evolve, and special considerations are required for patients with CLL with respect to the global SARS-CoV-2 pandemic. The unique funding and treatment environments in Australasia highlight the need for specific local guidance with respect to the investigation and management of CLL. This consensus practice statement was developed by a broadly representative group of ANZ experts in CLL with endorsement by peak haematology bodies, with a view to providing this standardised guidance.
Collapse
Affiliation(s)
- Mary A Anderson
- Department of Clinical Haematology, The Royal Melbourne Hospital and The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Blood Cells and Blood Cancer, The Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rory Bennett
- Department of Clinical Haematology, The Royal Melbourne Hospital and The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Xavier Badoux
- St George Hospital, Sydney, New South Wales, Australia
| | - Giles Best
- Flinders University and Medical Centre, Adelaide, South Australia, Australia
| | - Nicole Chia
- Genomic Diagnostics, Healius Pathology, Brisbane, Queensland, Australia
| | - Tara Cochrane
- Gold Coast University Hospital, Griffith University, Gold Coast, Queensland, Australia
| | - Gavin Cull
- Sir Charles Gairdner Hospital, PathWest Laboratory Medicine and University of Western Australia, Perth, Western Australia, Australia
| | - Kyle Crassini
- Mid North Coast Cancer Institute, Coffs Harbour Health Campus, Coffs Harbour, New South Wales, Australia
| | - Rosemary Harrup
- Cancer and Blood Services Royal Hobart Hospital, Hobart, Tasmania, Australia
- Menzies Research Institute, University of Tasmania, Hobart, Tasmania, Australia
| | - Sharon Jackson
- Te Whatu Ora health New Zealand Counties Manukau, Auckland, New Zealand
| | - Bryone Kuss
- Flinders University and Medical Centre, Adelaide, South Australia, Australia
| | - Masa Lasica
- St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Thomas E Lew
- Department of Clinical Haematology, The Royal Melbourne Hospital and The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Blood Cells and Blood Cancer, The Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paula Marlton
- Department of Haematology, Princess Alexandra Hospital and University of Queensland, Brisbane, Queensland, Australia
| | - Stephen Opat
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Victoria, Australia
| | - Emma Palfreyman
- Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Mark N Polizzotto
- Department of Clinical Haematology, The Canberra Hospital, Canberra, Australian Capital Territory, Australia
- Clinical Hub for Interventional Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Sumita Ratnasingam
- St John of God Hospital Geelong, Geelong, Victoria, Australia
- University Hospital Geelong, Geelong, Victoria, Australia
- School of Medicine, Geelong Clinical School, Deakin University, Geelong, Victoria, Australia
| | - John F Seymour
- Department of Clinical Haematology, The Royal Melbourne Hospital and The Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Asha Soosapilla
- Flow Cytometry, Healius Pathology, Sydney, New South Wales, Australia
| | - Dipti Talaulikar
- Department of Diagnostic Genomics, ACT Pathology, Canberra Health Services, Canberra, Australian Capital Territory, Australia
- Department of Haematology, ACT Pathology, Canberra Health Services, Canberra, Australian Capital Territory, Australia
- Australian National University, Canberra, Australian Capital Territory, Australia
| | - Constantine S Tam
- Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Robert Weinkove
- Te Rerenga Ora Blood & Cancer Centre, Te Whatu Ora Health New Zealand Capital Coast & Hutt Valley, Wellington, New Zealand
- Cancer Immunotherapy Programme, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Joel Wight
- Department of Haematology and Bone Marrow Transplantation, Townsville University Hospital, Townsville, Queensland, Australia
- James Cook University, School of Medicine, Townsville, Queensland, Australia
| | - Stephen P Mulligan
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Healius Pathology, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Benintende G, Pozzo F, Innocenti I, Autore F, Fresa A, D’Arena G, Gattei V, Lurenti L. Measurable residual disease in chronic lymphocytic leukemia. Front Oncol 2023; 13:1112616. [PMID: 36865804 PMCID: PMC9971803 DOI: 10.3389/fonc.2023.1112616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
Measurable residual disease (MRD) is defined as the presence of residual cancer cells after treatment in patients with clinically undetectable disease, who would otherwise be considered in complete remission. It is a highly sensitive parameter which indicates the disease burden and predicts survival in this setting of patients. In recent years, MRD has gained a role in many hematological malignancies as a surrogate endpoint for clinical trials: undetectable MRD has been correlated to longer progression free survival (PFS) and overall survival (OS). New drugs and combinations have been developed with the aim to achieve MRD negativity, which would indicate favorable prognosis. Different methods to measure MRD have also been devised, which include flow cytometry, polymerase chain reaction (PCR) and next generation sequencing (NGS), with different sensitivity and accuracy in evaluating deep remission after treatment. In this review, we will analyze the current recommendations for the detection of MRD, with particular focus on its role in Chronic Lymphocytic Leukemia (CLL), as well as the different detection methods. Moreover, we will discuss the results of clinical trials and the role of MRD in new therapeutic schemes with inhibitors and monoclonal antibodies. MRD is not currently used in the clinical practice to evaluate response to treatment, due to technical and economical limitations, but it's gaining more and more interest in trials settings, especially since the introduction of venetoclax. The use of MRD in trials will likely be followed by a broader practical application in the future. The aim of this work is to provide a reader-friendly summary of the state of art in the field, as MRD will soon become an accessible tool to evaluate our patients, predict their survival and guide physician's therapeutic choices and preferences.
Collapse
Affiliation(s)
- Giulia Benintende
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy,*Correspondence: Giulia Benintende,
| | - Federico Pozzo
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Idanna Innocenti
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesco Autore
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alberto Fresa
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giovanni D’Arena
- “San Luca” Hospital, Azienda Sanitaria Locale (ASL) Salerno, Salerno, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Luca Lurenti
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
5
|
Detection and Characterization of Circulating Tumor Cells Using Imaging Flow Cytometry—A Perspective Study. Cancers (Basel) 2022; 14:cancers14174178. [PMID: 36077716 PMCID: PMC9454939 DOI: 10.3390/cancers14174178] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Liquid biopsy is non-invasive approach used to prognose and monitor tumor progression based on the detection and examination of metastasis-related events found in the patients’ blood (such as circulating tumor cells (CTCs), extracellular vesicles, and circulating nucleic acids). Different ultrasensitive techniques are applied to study those events and the biology of tumor dissemination, which in the future might complement standard diagnostics. Here, we suggest that CTCs analysis could be improved by the usage of imaging flow cytometry, combining advantages of both standard flow cytometry (high-scale analysis) and microscopy (high resolution) to investigate detailed features of those cells. From this perspective, we discuss the potential of this technology in the CTC field and present representative images of CTCs from breast and prostate cancer patients analyzed with this method. Abstract Tumor dissemination is one of the most-investigated steps of tumor progression, which in recent decades led to the rapid development of liquid biopsy aiming to analyze circulating tumor cells (CTCs), extracellular vesicles (EVs), and circulating nucleic acids in order to precisely diagnose and monitor cancer patients. Flow cytometry was considered as a method to detect CTCs; however, due to the lack of verification of the investigated cells’ identity, this method failed to reach clinical utility. Meanwhile, imaging flow cytometry combining the sensitivity and high throughput of flow cytometry and image-based detailed analysis through a high-resolution microscope might open a new avenue in CTC technologies and provide an open-platform system alternative to CellSearch®, which is still the only gold standard in this field. Hereby, we shortly review the studies on the usage of flow cytometry in CTC identification and present our own representative images of CTCs envisioned by imaging flow cytometry providing rationale that this novel technology might be a good tool for studying tumor dissemination, and, if combined with a high CTC yield enrichment method, could upgrade CTC-based diagnostics.
Collapse
|
6
|
Polygenic risk score and risk of monoclonal B-cell lymphocytosis in caucasians and risk of chronic lymphocytic leukemia (CLL) in African Americans. Leukemia 2022; 36:119-125. [PMID: 34285341 PMCID: PMC8727288 DOI: 10.1038/s41375-021-01344-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022]
Abstract
Monoclonal B-cell lymphocytosis (MBL) is a precursor to CLL. Other than age, sex, and CLL family-history, little is known about factors associated with MBL risk. A polygenic-risk-score (PRS) of 41 CLL-susceptibility variants has been found to be associated with CLL risk among individuals of European-ancestry(EA). Here, we evaluate these variants, the PRS, and environmental factors for MBL risk. We also evaluate these variants and the CLL-PRS among African-American (AA) and EA-CLL cases and controls. Our study included 560 EA MBLs, 869 CLLs (696 EA/173 AA), and 2866 controls (2631 EA/235 AA). We used logistic regression, adjusting for age and sex, to estimate odds ratios (OR) and 95% confidence intervals within each race. We found significant associations with MBL risk among 21 of 41 variants and with the CLL-PRS (OR = 1.86, P = 1.9 × 10-29, c-statistic = 0.72). Little evidence of any association between MBL risk and environmental factors was observed. We observed significant associations of the CLL-PRS with EA-CLL risk (OR = 2.53, P = 4.0 × 10-63, c-statistic = 0.77) and AA-CLL risk (OR = 1.76, P = 5.1 × 10-5, c-statistic = 0.62). Inherited genetic factors and not environmental are associated with MBL risk. In particular, the CLL-PRS is a strong predictor for both risk of MBL and EA-CLL, but less so for AA-CLL supporting the need for further work in this population.
Collapse
|
7
|
Multiparametric Flow Cytometry for MRD Monitoring in Hematologic Malignancies: Clinical Applications and New Challenges. Cancers (Basel) 2021; 13:cancers13184582. [PMID: 34572809 PMCID: PMC8470441 DOI: 10.3390/cancers13184582] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In hematologic cancers, Minimal Residual Disease (MRD) monitoring, using either molecular (PCR) or immunophenotypic (MFC) diagnostics, allows the identification of rare cancer cells, readily detectable either in the bone marrow or in the peripheral blood at very low levels, far below the limit of classic microscopy. In this paper, we outlined the state-of-the-art of MFC-based MRD detection in different hematologic settings, highlighting main recommendations and new challenges for using such method in patients with acute leukemias or chronic hematologic neoplasms. The combination of new molecular technologies with advanced flow cytometry is progressively allowing clinicians to design a personalized therapeutic path, proportionate to the biological aggressiveness of the disease, in particular by using novel immunotherapies, in view of a modern decision-making process, based on precision medicine. Abstract Along with the evolution of immunophenotypic and molecular diagnostics, the assessment of Minimal Residual Disease (MRD) has progressively become a keystone in the clinical management of hematologic malignancies, enabling valuable post-therapy risk stratifications and guiding risk-adapted therapeutic approaches. However, specific prognostic values of MRD in different hematological settings, as well as its appropriate clinical uses (basically, when to measure it and how to deal with different MRD levels), still need further investigations, aiming to improve standardization and harmonization of MRD monitoring protocols and MRD-driven therapeutic strategies. Currently, MRD measurement in hematological neoplasms with bone marrow involvement is based on advanced highly sensitive methods, able to detect either specific genetic abnormalities (by PCR-based techniques and next-generation sequencing) or tumor-associated immunophenotypic profiles (by multiparametric flow cytometry, MFC). In this review, we focus on the growing clinical role for MFC-MRD diagnostics in hematological malignancies—from acute myeloid and lymphoblastic leukemias (AML, B-ALL and T-ALL) to chronic lymphocytic leukemia (CLL) and multiple myeloma (MM)—providing a comparative overview on technical aspects, clinical implications, advantages and pitfalls of MFC-MRD monitoring in different clinical settings.
Collapse
|
8
|
Immune Gene Rearrangements: Unique Signatures for Tracing Physiological Lymphocytes and Leukemic Cells. Genes (Basel) 2021; 12:genes12070979. [PMID: 34198966 PMCID: PMC8329920 DOI: 10.3390/genes12070979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023] Open
Abstract
The tremendous diversity of the human immune repertoire, fundamental for the defense against highly heterogeneous pathogens, is based on the ingenious mechanism of immune gene rearrangements. Rearranged immune genes encoding the immunoglobulins and T-cell receptors and thus determining each lymphocyte's antigen specificity are very valuable molecular markers for tracing malignant or physiological lymphocytes. One of their most significant applications is tracking residual leukemic cells in patients with lymphoid malignancies. This so called 'minimal residual disease' (MRD) has been shown to be the most important prognostic factor across various leukemia subtypes and has therefore been given enormous attention. Despite the current rapid development of the molecular methods, the classical real-time PCR based approach is still being regarded as the standard method for molecular MRD detection due to the cumbersome standardization of the novel approaches currently in progress within the EuroMRD and EuroClonality NGS Consortia. Each of the molecular methods, however, poses certain benefits and it is therefore expectable that none of the methods for MRD detection will clearly prevail over the others in the near future.
Collapse
|
9
|
Wu X, Lu H, Pang T, Li X, Luo H, Tan H, Liu S. Association of minimal residual disease levels with clinical outcomes in patients with mantle cell lymphoma: A meta-analysis. Leuk Res 2021; 108:106605. [PMID: 34090063 DOI: 10.1016/j.leukres.2021.106605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023]
Abstract
Some studies have elucidated that Minimal residual disease (MRD) in patient with Mantle Cell Lymphoma (MCL) was a significant prognostic factor, with potential value in assessing overall survival (OS) and progression-free survival (PFS). However, most studies were widely varied in included population, sample sources and MRD detection time points. Some studies even have conflicting results. In view of this, a meta-analysis was performed to evaluate association of MRD levels with clinical outcomes in patients with MCL. We identified 7 included articles, which were published in recent 20 years. Then, we extracted or calculated hazard ratios (HRs) and their 95 % confidence intervals (CIs). Our results reveal that patients with MRD negativity have improved OS (HR = 0.63; 95 % CI: 0.50-0.79) and PFS (HR = 0.40, 95 % CI: 0.21-0.76), comparing with patients with MRD positivity. There are also consistent results in subgroups based on sample sources and MRD detection time points. Our study also demonstrates that MRD level is a strong prognostic factor of clinical outcomes. Thus, MRD is expected to be an effective clinical indicator for assessing prognosis and guide treatment decisions in MCL patients.
Collapse
Affiliation(s)
- Xue Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Hongyu Lu
- School of Medical Technology, Chengdu University of TCM, Chengdu, 611137, Sichuan Province, China; Key Laboratory of Transplant Engineering and Immunology, Regenerative Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Pang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan Province, China
| | - Xue Li
- Department of Laboratory Medicine, Hanyuan People's Hospital, Yuan, 25000, Sichuan, China
| | - Hongzhi Luo
- School of Laboratory Medicine, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hong Tan
- Department of General Surgery, Chengdu Integrated TCM&Western Medicine Hospital (Chengdu First People's Hospital), No.18 Vientiane North Road, Hi-tech Zone, Chengdu, 610041, China.
| | - Shan Liu
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
10
|
Slager SL, Lanasa MC, Marti GE, Achenbach SJ, Camp NJ, Abbasi F, Kay NE, Vachon CM, Cerhan JR, Johnston JB, Call TG, Rabe KG, Kleinstern G, Boddicker NJ, Norman AD, Parikh SA, Leis JF, Banerji V, Brander DM, Glenn M, Ferrajoli A, Curtin K, Braggio E, Shanafelt TD, McMaster ML, Weinberg JB, Hanson CA, Caporaso NE. Natural history of monoclonal B-cell lymphocytosis among relatives in CLL families. Blood 2021; 137:2046-2056. [PMID: 33512457 PMCID: PMC8057266 DOI: 10.1182/blood.2020006322] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/14/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic lymphocytic lymphoma (CLL) has one of the highest familial risks among cancers. Monoclonal B-cell lymphocytosis (MBL), the precursor to CLL, has a higher prevalence (13%-18%) in families with 2 or more members with CLL compared with the general population (5%-12%). Although, the rate of progression to CLL for high-count MBLs (clonal B-cell count ≥500/µL) is ∼1% to 5%/y, no low-count MBLs have been reported to progress to date. We report the incidence and natural history of MBL in relatives from CLL families. In 310 CLL families, we screened 1045 relatives for MBL using highly sensitive flow cytometry and prospectively followed 449 of them. MBL incidence was directly age- and sex-adjusted to the 2010 US population. CLL cumulative incidence was estimated using Kaplan-Meier survival curves. At baseline, the prevalence of MBL was 22% (235/1045 relatives). After a median follow-up of 8.1 years among 449 relatives, 12 individuals progressed to CLL with a 5-year cumulative incidence of 1.8%. When considering just the 139 relatives with low-count MBL, the 5-year cumulative incidence increased to 5.7%. Finally, 264 had no MBL at baseline, of whom 60 individuals subsequently developed MBL (2 high-count and 58 low-count MBLs) with an age- and sex-adjusted incidence of 3.5% after a median of 6 years of follow-up. In a screening cohort of relatives from CLL families, we reported progression from normal-count to low-count MBL to high-count MBL to CLL, demonstrating that low-count MBL precedes progression to CLL. We estimated a 1.1% annual rate of progression from low-count MBL, which is in excess of that in the general population.
Collapse
Affiliation(s)
- Susan L Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Mark C Lanasa
- Department of Medicine, Duke University, Duke Cancer Institute, Durham, NC
| | - Gerald E Marti
- Lymphoid Malignancies Section, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Sara J Achenbach
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Nicola J Camp
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Fatima Abbasi
- Center for Biologics Research and Evaluation, Food and Drug Administration, Silver Springs, MD
| | - Neil E Kay
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Celine M Vachon
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - James B Johnston
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Timothy G Call
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Kari G Rabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | - Aaron D Norman
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Sameer A Parikh
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Jose F Leis
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Versha Banerji
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Research Institute in Oncology and Hematology, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Danielle M Brander
- Department of Medicine, Duke University, Duke Cancer Institute, Durham, NC
| | - Martha Glenn
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Alessandra Ferrajoli
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Karen Curtin
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT
| | - Esteban Braggio
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Tait D Shanafelt
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA
| | - Mary L McMaster
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - J Brice Weinberg
- Department of Medicine, Duke University, Duke Cancer Institute, Durham, NC
- Department of Immunology, Duke University Medical Center, Durham, NC
- Durham Veterans Affairs Medical Center, Durham, NC; and
| | - Curtis A Hanson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
11
|
von Heydebrand F, Fuchs M, Kunz M, Voelkl S, Kremer AN, Oostendorp RAJ, Wilke J, Leitges M, Egle A, Mackensen A, Lutzny-Geier G. Protein kinase C-β-dependent changes in the glucose metabolism of bone marrow stromal cells of chronic lymphocytic leukemia. STEM CELLS (DAYTON, OHIO) 2021; 39:819-830. [PMID: 33539629 DOI: 10.1002/stem.3352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/15/2021] [Indexed: 11/10/2022]
Abstract
Survival of chronic lymphocytic leukemia (CLL) cells critically depends on the support of an adapted and therefore appropriate tumor microenvironment. Increasing evidence suggests that B-cell receptor-associated kinases such as protein kinase C-β (PKCβ) or Lyn kinase are essential for the formation of a microenvironment supporting leukemic growth. Here, we describe the impact of PKCβ on the glucose metabolism in bone marrow stromal cells (BMSC) upon CLL contact. BMSC get activated by CLL contact expressing stromal PKCβ that diminishes mitochondrial stress and apoptosis in CLL cells by stimulating glucose uptake. In BMSC, the upregulation of PKCβ results in increased mitochondrial depolarization and leads to a metabolic switch toward oxidative phosphorylation. In addition, PKCβ-deficient BMSC regulates the expression of Hnf1 promoting stromal insulin signaling after CLL contact. Our data suggest that targeting PKCβ and the glucose metabolism of the leukemic niche could be a potential therapeutic strategy to overcome stroma-mediated drug resistance.
Collapse
Affiliation(s)
- Franziska von Heydebrand
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maximilian Fuchs
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Meik Kunz
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Simon Voelkl
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anita N Kremer
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert A J Oostendorp
- Clinic and Polyclinic for Internal Medicine III: Hematology and Oncology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Jochen Wilke
- Practice for Oncology and Hematology, Fürth, Germany
| | - Michael Leitges
- Faculty of Medicine, Division of BioMedical Sciences, Craig L. Dobbin Genetics Research Centre, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Alexander Egle
- IIIrd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria.,Salzburg Cancer Research Institute (SCRI) with Laboratory of Immunological and Molecular Cancer Research (LIMCR), Salzburg, Austria.,Cancer Cluster Salzburg, Salzburg, Austria
| | - Andreas Mackensen
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gloria Lutzny-Geier
- Department of Medicine 5-Hematology and Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
12
|
Wake LM, Ahn IE, Farooqui MZ, Tian X, Stetler-Stevenson M, Marti GE, Wiestner A, Maric I. Dual antibody immunohistochemistry: an efficient and sensitive tool for the detection of residual disease in chronic lymphocytic leukemia. J Hematop 2019. [DOI: 10.1007/s12308-019-00372-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
13
|
Del Giudice I, Raponi S, Della Starza I, De Propris MS, Cavalli M, De Novi LA, Cappelli LV, Ilari C, Cafforio L, Guarini A, Foà R. Minimal Residual Disease in Chronic Lymphocytic Leukemia: A New Goal? Front Oncol 2019; 9:689. [PMID: 31555576 PMCID: PMC6727319 DOI: 10.3389/fonc.2019.00689] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), there is a growing interest for minimal residual disease (MRD) monitoring, due to the availability of drug combinations capable of unprecedented complete clinical responses. The standardized and most commonly applied methods to assess MRD in CLL are based on flow cytometry (FCM) and, to a lesser extent, real-time quantitative PCR (RQ-PCR) with allele-specific oligonucleotide (ASO) primers of immunoglobulin heavy chain genes (IgH). Promising results are being obtained using droplet digital PCR (ddPCR) and next generation sequencing (NGS)-based approaches, with some advantages and a potential higher sensitivity compared to the standardized methodologies. Plasma cell-free DNA can also be explored as a more precise measure of residual disease from all different compartments, including the lymph nodes. From a clinical point of view, CLL MRD quantification has proven an independent prognostic marker of progression-free survival (PFS) and overall survival (OS) after chemoimmunotherapy as well as after allogeneic transplantation. In the era of mechanism-driven drugs, the paradigms of CLL treatment are being revolutionized, challenging the use of chemoimmunotherapy even in first-line. The continuous administration of ibrutinib single agent has led to prolonged PFS and OS in relapsed/refractory and treatment naïve CLL, including those with TP53 deletion/mutation or unmutated IGHV genes, though the clinical responses are rarely complete. More recently, chemo-free combinations of venetoclax+rituximab, venetoclax+obinutuzumab or ibrutinib+venetoclax have been shown capable of inducing undetectable MRD in the bone marrow, opening the way to protocols exploring a MRD-based duration of treatment, aiming at disease eradication. Thus, beside a durable disease control desirable particularly for older patients and/or for those with comorbidities, a MRD-negative complete remission is becoming a realistic prospect for CLL patients in an attempt to obtain a long-lasting eradication and possibly cure of the disease. Here we discuss the standardized and innovative technical approaches for MRD detection in CLL, the clinical impact of MRD monitoring in chemoimmunotherapy and chemo-free trials and the future clinical implications of MRD monitoring in CLL patients outside of clinical trials.
Collapse
Affiliation(s)
- Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Sara Raponi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Maria Stefania De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Anna De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Vincenzo Cappelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Caterina Ilari
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Luciana Cafforio
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
14
|
Abstract
Flow-cytometric demonstration of the typical chronic lymphocytic leukemia (CLL) immunophenotype is vital for diagnosis. CLL has a characteristic immunophenotype, expressing CD5, CD19, dim CD20, dim CD22, CD23, bright CD43, dim CD45, dim to negative CD79b, dim CD81, CD200, and dim monoclonal surface immunoglobulin. This characteristic immunophenotype allows a definitive diagnosis and the ruling out of another leukemia or lymphoma. Flow cytometry also provides important prognostic information and accurate assessment of response to therapy. Here we describe optimal specimen collection, red cell lysis, appropriate panel, cell staining, acquisition on a flow cytometer, and analysis for CLL specimens.
Collapse
|
15
|
Gauthier M, Comont T, Vergez F, Ysebaert L. [Minimal residual disease in chronic lymphocytic leukemia: A still current issue in 2018]. Bull Cancer 2018; 105:1042-1051. [PMID: 30243477 DOI: 10.1016/j.bulcan.2018.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/19/2018] [Accepted: 07/01/2018] [Indexed: 12/21/2022]
Abstract
Minimal residual disease (MRD) is widely used in oncohematology. In chronic lymphocytic leukemia (CLL), it can be measured by flow cytometry or polymerase chain reaction and is getting a greater place, owing to the dramatic therapeutic advances in the management this disease. As MRD decrease after chemoimmunotherapy is associated with improved progression free and overall survivals, its measure is now recommended as a surrogate marker for cytotoxic drugs licensures. This association is independent from treatment received and raises a few questions, such as sequential MRD measures to stop treatment in case of an early deep response and on the opposite, treatment continuation until reaching undetectable MRD (with the possible use of maintenance therapy). Furthermore, following MRD after a cytotoxic treatment could lead clinical trials investigators to propose pre-emptive treatments in case of MRD re-growth, to avoid overt relaspe. MRD re-growth kinetics and CD4 count after treatment completion can improve MRD-based survival predictions. On the other hand, BCR inhibitors do not lead to undetectable MRD, but their association with chemoimmunotherapy increases the proportion of patients reaching that goal. Moreover, BCL2 inhibitors do lead to deep response including in the relapse/refractory setting, giving to MRD a central place in currently investigated treatments evaluation.
Collapse
Affiliation(s)
- Martin Gauthier
- IUCT-Oncopôle, service d'hématologie clinique, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France.
| | - Thibault Comont
- IUCT-Oncopôle, service de médecine interne, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France
| | - François Vergez
- IUCT-Oncopôle, laboratoire d'hématologie-immunophenotypage et hematologie cellulaire, 1 avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France
| | - Loïc Ysebaert
- IUCT-Oncopôle, service d'hématologie clinique, 1, avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France; IUCT-Oncopôle, laboratoire d'hématologie-immunophenotypage et hematologie cellulaire, 1 avenue Irène-Joliot-Curie, 31059 Toulouse cedex 9, France; CRCT, Inserm UMR 1037, 2, avenue Hubert-Curien, 31037 Toulouse cedex 1, France
| |
Collapse
|
16
|
Stilgenbauer S, Leblond V, Foà R, Böttcher S, Ilhan O, Knauf W, Mikuskova E, Renner C, Tausch E, Woszczyk D, Gresko E, Lundberg L, Moore T, Morris T, Robson S, Bosch F. Obinutuzumab plus bendamustine in previously untreated patients with CLL: a subgroup analysis of the GREEN study. Leukemia 2018; 32:1778-1786. [PMID: 29749403 PMCID: PMC6087718 DOI: 10.1038/s41375-018-0146-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/13/2018] [Accepted: 03/28/2018] [Indexed: 01/24/2023]
Abstract
GREEN (NCT01905943) is a non-randomized, open-label phase IIIb study investigating obinutuzumab alone or plus chemotherapy in chronic lymphocytic leukemia (CLL). We report a preplanned subgroup analysis of 158 previously untreated CLL patients receiving obinutuzumab-bendamustine (G-B). Patients received six 28-day cycles (C) of G-B: obinutuzumab day (D)1/D2 of C1 (25 mg D1/975 mg D2), 1000 mg D8 and D15 of C1, and D1 of C2-6; and bendamustine 70/90 mg/m2 D1 and D2 of C1-6. The primary endpoint was safety/tolerability. Grade ≥3 adverse events (AEs) occurred in 82.3% of patients, including neutropenia (49.4%), thrombocytopenia (12.0%) and febrile neutropenia (10.8%). Serious AEs included neutropenia (12.7%), febrile neutropenia (9.5%) and pneumonia (7.6%). Rates of grade ≥3 infections and infusion-related reactions were 20.3% and 17.1%, respectively. Due to tumor lysis syndrome (TLS; 8.2%), including two associated fatalities (one in another study cohort), additional risk-minimization measures were implemented. Overall response rate was 81.0%. After 32.8 months' median observation time, 2-year progression-free survival was 81.8%. Minimal residual disease was undetectable in 59.5% (94/158) and 27.8% (44/158) of patients for blood and bone marrow, respectively. Frontline G-B appears to have manageable toxicity with clinical activity in CLL. Careful TLS risk assessment, pretreatment and monitoring is required.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Bendamustine Hydrochloride/administration & dosage
- Drug Resistance, Neoplasm/drug effects
- Female
- Follow-Up Studies
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Neoplasm, Residual/drug therapy
- Neoplasm, Residual/pathology
- Prognosis
- Remission Induction
- Rituximab/administration & dosage
- Salvage Therapy
- Survival Rate
Collapse
Affiliation(s)
- Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Ulm, Germany.
- Klinik für Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany.
| | | | - Robin Foà
- Department of Cellular Biotechnologies and Hematology, 'Sapienza' University, Rome, Italy
| | - Sebastian Böttcher
- Second Department of Medicine, University of Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Clinic III, Hematology, Oncology and Palliative Medicine, University of Rostock, Rostock, Germany
| | | | - Wolfgang Knauf
- Onkologische Gemeinschaftspraxis, Agaplesion Bethanien Krankenhaus, Frankfurt, Germany
| | | | | | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Dariusz Woszczyk
- State Hospital, Opole, Poland
- Haematology Department, University of Opole, Provincial Hospital, Opole, Poland
| | | | | | - Tom Moore
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Thea Morris
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | | |
Collapse
|
17
|
Fludarabine and rituximab with escalating doses of lenalidomide followed by lenalidomide/rituximab maintenance in previously untreated chronic lymphocytic leukaemia (CLL): the REVLIRIT CLL-5 AGMT phase I/II study. Ann Hematol 2018; 97:1825-1839. [PMID: 29862437 PMCID: PMC6097797 DOI: 10.1007/s00277-018-3380-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/21/2018] [Indexed: 12/11/2022]
Abstract
Despite recent advances, chemoimmunotherapy remains a standard for fit previously untreated chronic lymphocytic leukaemia patients. Lenalidomide had activity in early monotherapy trials, but tumour lysis and flare proved major obstacles in its development. We combined lenalidomide in increasing doses with six cycles of fludarabine and rituximab (FR), followed by lenalidomide/rituximab maintenance. In 45 chemo-naive patients, included in this trial, individual tolerability of the combination was highly divergent and no systematic toxicity determining a maximum tolerated dose was found. Grade 3/4 neutropenia (71%) was high, but only 7% experienced grade 3 infections. No tumour lysis or flare > grade 2 was observed, but skin toxicity proved dose-limiting in nine patients (20%). Overall and complete response rates after induction were 89 and 44% by intention-to-treat, respectively. At a median follow-up of 78.7 months, median progression-free survival (PFS) was 60.3 months. Minimal residual disease and immunoglobulin variable region heavy chain mutation state predicted PFS and TP53 mutation most strongly predicted OS. Baseline clinical factors did not predict tolerance to the immunomodulatory drug lenalidomide, but pretreatment immunophenotypes of T cells showed exhausted memory CD4 cells to predict early dose-limiting non-haematologic events. Overall, combining lenalidomide with FR was feasible and effective, but individual changes in the immune system seemed associated with limiting side effects. clinicaltrials.gov (NCT00738829) and EU Clinical Trials Register (www.clinicaltrialsregister.eu, 2008-001430-27)
Collapse
|
18
|
Tomuleasa C, Selicean C, Cismas S, Jurj A, Marian M, Dima D, Pasca S, Petrushev B, Moisoiu V, Micu WT, Vischer A, Arifeen K, Selicean S, Zdrenghea M, Bumbea H, Tanase A, Grewal R, Pop L, Aanei C, Berindan-Neagoe I. Minimal residual disease in chronic lymphocytic leukemia: A consensus paper that presents the clinical impact of the presently available laboratory approaches. Crit Rev Clin Lab Sci 2018; 55:329-345. [PMID: 29801428 DOI: 10.1080/10408363.2018.1463508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy defined by the accumulation of mature lymphocytes in the lymphoid tissues, bone marrow, and blood. Therapy for CLL is guided according to the Rai and Binet staging systems. Nevertheless, state-of-the-art protocols in disease monitoring, diagnostics, and prognostics for CLL are based on the assessment of minimal residual disease (MRD). MRD is internationally considered to be the level of disease that can be detected by sensitive techniques and represents incomplete treatment and a probability of disease relapse. MRD detection has been continuously improved by the quick development of both flow cytometry and molecular biology technology, as well as by next-generation sequencing. Considering that MRD detection is moving more and more from research to clinical practice, where it can be an independent prognostic marker, in this paper, we present the methodologies by which MRD is evaluated, from translational research to clinical practice.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- a Department of Hematology , Ion Chiricuta Clinical Cancer Center , Cluj Napoca , Romania.,b Research Center for Functional Genomics and Translational Medicine/Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Cristina Selicean
- a Department of Hematology , Ion Chiricuta Clinical Cancer Center , Cluj Napoca , Romania
| | - Sonia Cismas
- c Department of Genetics , Victor Babes University of Medicine and Pharmacy , Timisoara , Romania.,d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Anca Jurj
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Mirela Marian
- a Department of Hematology , Ion Chiricuta Clinical Cancer Center , Cluj Napoca , Romania
| | - Delia Dima
- a Department of Hematology , Ion Chiricuta Clinical Cancer Center , Cluj Napoca , Romania
| | - Sergiu Pasca
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Bobe Petrushev
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Vlad Moisoiu
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Wilhelm-Thomas Micu
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Anna Vischer
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Kanza Arifeen
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Sonia Selicean
- d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Mihnea Zdrenghea
- a Department of Hematology , Ion Chiricuta Clinical Cancer Center , Cluj Napoca , Romania.,d Department of Hematology , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Horia Bumbea
- f Department of Hematology , Carol Davila University of Medicine and Pharmacy , Bucharest , Romania.,g Department of Hematology , University Clinical Hospital , Bucharest , Romania
| | - Alina Tanase
- h Department of Stem Cell Transplantation , Fundeni Clinical Institute , Bucharest , Romania
| | - Ravnit Grewal
- i South African Medical Research Council Bioinformatics Unit , The South African National Bioinformatics Institute (SANBI), University of the Western Cape , Bellville , South Africa
| | - Laura Pop
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| | - Carmen Aanei
- j Hematology Laboratory, Pole de Biologie-Pathologie , University Hospital of St. Etienne , St. Etienne , France
| | - Ioana Berindan-Neagoe
- e Research Center for Functional Genomics and Translational Medicine , Iuliu Hatieganu University of Medicine and Pharmacy , Cluj Napoca , Romania
| |
Collapse
|
19
|
Kovacs G, Robrecht S, Fink AM, Bahlo J, Cramer P, von Tresckow J, Maurer C, Langerbeins P, Fingerle-Rowson G, Ritgen M, Kneba M, Döhner H, Stilgenbauer S, Klapper W, Wendtner CM, Fischer K, Hallek M, Eichhorst B, Böttcher S. Minimal Residual Disease Assessment Improves Prediction of Outcome in Patients With Chronic Lymphocytic Leukemia (CLL) Who Achieve Partial Response: Comprehensive Analysis of Two Phase III Studies of the German CLL Study Group. J Clin Oncol 2017; 34:3758-3765. [PMID: 27573660 DOI: 10.1200/jco.2016.67.1305] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose To determine the value of minimal residual disease (MRD) assessments, together with the evaluation of clinical response in chronic lymphocytic leukemia according to the 2008 International Workshop on Chronic Lymphocytic Leukemia criteria. Patients and Methods Progression-free survival (PFS) and overall survival of 554 patients from two randomized trials of the German CLL Study Group (CLL8: fludarabine and cyclophosphamide [FC] v FC plus rituximab; CLL10: FC plus rituximab v bendamustine plus rituximab) were analyzed according to MRD assessed in peripheral blood at a threshold of 10-4 and clinical response. The prognostic value of different parameters defining a partial response (PR) was further investigated. Results Patients with MRD-negative complete remission (CR), MRD-negative PR, MRD-positive CR, and MRD-positive PR experienced a median PFS from a landmark at end of treatment of 61 months, 54 months, 35 months, and 21 months, respectively. PFS did not differ significantly between MRD-negative CR and MRD-negative PR; however, PFS was longer for MRD-negative PR than for MRD-positive CR ( P = .048) and for MRD-positive CR compared with MRD-positive PR ( P = .002). Compared with MRD-negative CR, only patients with MRD-positive PR had a significantly shorter overall survival (not reached v 72 months; P = .001), whereas there was no detectable difference for patients with MRD-negative PR or MRD-positive CR ( P = 0.612 and P = 0.853, respectively). Patients with MRD-negative PR who presented with residual splenomegaly had only a similar PFS (63 months) compared with patients with MRD-negative CR (61 months; P = .354), whereas patients with MRD-negative PR with lymphadenopathy showed a shorter PFS (31 months; P < .001). Conclusion MRD quantification allows for improved PFS prediction in both patients who achive PR and CR, which thus supports its application in all responders. In contrast to residual lymphadenopathy, persisting splenomegaly does not impact outcome in patients with MRD-negative PR.
Collapse
Affiliation(s)
- Gabor Kovacs
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Sandra Robrecht
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Anna Maria Fink
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Jasmin Bahlo
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Paula Cramer
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Julia von Tresckow
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Christian Maurer
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Petra Langerbeins
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Günter Fingerle-Rowson
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Matthias Ritgen
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Michael Kneba
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Hartmut Döhner
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Stephan Stilgenbauer
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Wolfram Klapper
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Clemens-Martin Wendtner
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Kirsten Fischer
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Michael Hallek
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Barbara Eichhorst
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| | - Sebastian Böttcher
- Gabor Kovacs, Sandra Robrecht, Anna Maria Fink, Jasmin Bahlo, Paula Cramer, Julia von Tresckow, Christian Maurer, Petra Langerbeins, Kirsten Fischer, Michael Hallek, and Barbara Eichhorst, University of Cologne and Center of Integrated Oncology Cologne/Bonn, Cologne; Matthias Ritgen, Michael Kneba, and Sebastian Böttcher, University of Schleswig-Holstein, Lubeck; Hartmut Döhner and Stephan Stilgenbauer, University of Ulm, Ulm; Wolfram Klapper, University of Kiel, Kiel; Clemens-Martin Wendtner, Klinikum Schwabing, Munich, Germany; and Günter Fingerle-Rowson, F. Hoffmann La-Roche, Basel, Switzerland
| |
Collapse
|
20
|
Fink AM, Bahlo J, Robrecht S, Al-Sawaf O, Aldaoud A, Hebart H, Jentsch-Ullrich K, Dörfel S, Fischer K, Wendtner CM, Nösslinger T, Ghia P, Bosch F, Kater AP, Döhner H, Kneba M, Kreuzer KA, Tausch E, Stilgenbauer S, Ritgen M, Böttcher S, Eichhorst B, Hallek M. Lenalidomide maintenance after first-line therapy for high-risk chronic lymphocytic leukaemia (CLLM1): final results from a randomised, double-blind, phase 3 study. LANCET HAEMATOLOGY 2017; 4:e475-e486. [PMID: 28916311 DOI: 10.1016/s2352-3026(17)30171-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 01/27/2023]
Abstract
BACKGROUND The combined use of genetic markers and detectable minimal residual disease identifies patients with chronic lymphocytic leukaemia with poor outcome after first-line chemoimmunotherapy. We aimed to assess lenalidomide maintenance therapy in these high-risk patients. METHODS In this randomised, double-blind, phase 3 study (CLLM1; CLL Maintenance 1 of the German CLL Study Group), patients older than 18 years and diagnosed with immunophenotypically confirmed chronic lymphocytic leukaemia with active disease, who responded to chemoimmunotherapy 2-5 months after completion of first-line therapy and who were assessed as having a high risk for an early progression with at least a partial response after four or more cycles of first-line chemoimmunotherapy, were eligible if they had high minimal residual disease levels or intermediate levels combined with an unmutated IGHV gene status or TP53 alterations. Patients were randomly assigned (2:1) to receive either lenalidomide (5 mg) or placebo. Randomisation was done with a fixed block size of three, and was stratified according to the minimal residual disease level achieved after first-line therapy. Maintenance was started with 5 mg daily, and was escalated to the target dose of 15 mg. If tolerated, medication was administered until disease progression. The primary endpoint was progression-free survival according to an independent review. The pre-planned interim analysis done by intention to treat was done after 20% of the calculated progression-free survival events. This study is registered with ClinicalTrials.gov, number NCT01556776; treatment in the lenalidomide group is still ongoing. FINDINGS Between July 5, 2012, and March 15, 2016, 468 previously untreated patients with chronic lymphocytic leukaemia were screened for the study; 379 (81%) were not eligible. Recruitment was closed prematurely due to poor accrual after 89 of 200 planned patients were randomly assigned: 60 (67%) enrolled patients were assigned to the lenalidomide group and 29 (33%) to the placebo group, of whom 56 (63%) received lenalidomide and 29 (33%) placebo, with a median of 11·0 (IQR 4·5-20·5) treatment cycles at data cutoff. After a median observation time of 17·9 months (IQR 9·1-28·1), the hazard ratio for progression-free survival assessed by an independent review was 0·168 (95% CI 0·074-0·379). Median progression-free survival was 13·3 months (95% CI 9·9-19·7) in the placebo group and not reached (95% CI 32·3-not evaluable) in the lenalidomide group. The most frequent adverse events were skin disorders (35 patients [63%] in the lenalidomide group vs eight patients [28%] in the placebo group), gastrointestinal disorders (34 [61%] vs eight [28%]), infections (30 [54%] vs 19 [66%]), haematological toxicity (28 [50%] vs five [17%]), and general disorders (28 [50%] vs nine [31%]). One fatal adverse event was reported in each of the treatment groups (one [2%] patient with fatal acute lymphocytic leukaemia in the lenalidomide group and one patient (3%) with fatal multifocal leukoencephalopathy in the placebo group). INTERPRETATION Lenalidomide is an efficacious maintenance therapy reducing the relative risk of progression in first-line patients with chronic lymphocytic leukaemia who do not achieve minimal residual disease negative disease state following chemoimmunotherapy approaches. The toxicity seems to be acceptable considering the poor prognosis of the eligible patients. The trial independently confirms the clinical significance of a novel, minimal residual disease-based algorithm to predict short progression-free survival, which might be incorporated in future clinical trials to identify candidates for additional maintenance treatment. FUNDING Celgene Corporation.
Collapse
Affiliation(s)
- Anna Maria Fink
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany.
| | - Jasmin Bahlo
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Sandra Robrecht
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Othman Al-Sawaf
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Ali Aldaoud
- Gemeinschaftspraxis für Hämatologie und Onkologie, Leipzig, Germany
| | - Holger Hebart
- Zentrum für Innere Medizin, Hämatologie/Onkologie, Stauferklinikum Schwäbisch-Gmünd, Germany
| | | | | | - Kirsten Fischer
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Clemens-Martin Wendtner
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany; Department of Hematology, Oncology, Immunology, Palliative Care, Infectious Diseases and Tropical Medicine, Klinikum Schwabing, Munich, Germany
| | - Thomas Nösslinger
- 3rd Department for Hematology and Oncology, Hanusch Hospital, Vienna, Austria
| | - Paolo Ghia
- Strategic Research Program on CLL, Università Vita-Salute San Raffaele and IRCCS Istituto Scientifico San Raffaele, Milan, Italy
| | - Francesc Bosch
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain; on behalf of the Spanish Group of CLL (GELLC)
| | - Arnon P Kater
- Department of Hematology, Academic Medical Center Amsterdam, University of Amsterdam, Netherlands; on behalf of the Dutch-Belgium HOVON CLL study group
| | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Michael Kneba
- Campus Kiel, 2nd Deptartment of Internal Medicine, University of Schleswig-Holstein, Kiel, Germany
| | - Karl-Anton Kreuzer
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Eugen Tausch
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | | | - Matthias Ritgen
- Campus Kiel, 2nd Deptartment of Internal Medicine, University of Schleswig-Holstein, Kiel, Germany
| | - Sebastian Böttcher
- Campus Kiel, 2nd Deptartment of Internal Medicine, University of Schleswig-Holstein, Kiel, Germany; Department of Medicine III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Barbara Eichhorst
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine and Center of Integrated Oncology Cologne-Bonn, German CLL Study Group, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Results of the randomized phase IIB ADMIRE trial of FCR with or without mitoxantrone in previously untreated CLL. Leukemia 2017; 31:2085-2093. [PMID: 28216660 DOI: 10.1038/leu.2017.65] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/24/2016] [Accepted: 11/29/2016] [Indexed: 11/09/2022]
Abstract
ADMIRE was a multicenter, randomized-controlled, open, phase IIB superiority trial in previously untreated chronic lymphocytic leukemia. Conventional front-line therapy in fit patients is fludarabine, cyclophosphamide and rituximab (FCR). Initial evidence from non-randomized phase II trials suggested that the addition of mitoxantrone to FCR (FCM-R) improved remission rates. Two hundred and fifteen patients were recruited to assess the primary end point of complete remission (CR) rates according to International Workshop on Chronic Lymphocytic Leukemia criteria. Secondary end points were progression-free survival (PFS), overall survival (OS), overall response rate, minimal residual disease (MRD) negativity and safety. At final analysis, CR rates were 69.8 FCR vs 69.3% FCM-R (adjusted odds ratio (OR): 0.97; 95% confidence interval (CI): (0.53-1.79), P=0.932). MRD-negativity rates were 59.3 FCR vs 50.5% FCM-R (adjusted OR: 0.70; 95% CI: (0.39-1.26), P=0.231). During treatment, 60.0% (n=129) of participants received granulocyte colony-stimulating factor as secondary prophylaxis for neutropenia, a lower proportion on FCR compared with FCM-R (56.1 vs 63.9%). The toxicity of both regimens was acceptable. There are no significant differences between the treatment groups for PFS and OS. The trial demonstrated that the addition of mitoxantrone to FCR did not increase the depth of response. Oral FCR was well tolerated and resulted in impressive responses in terms of CR rates and MRD negativity compared with historical series with intravenous chemotherapy.
Collapse
|
22
|
Robak T, Hellmann A, Kloczko J, Loscertales J, Lech-Maranda E, Pagel JM, Mato A, Byrd JC, Awan FT, Hebart H, Garcia-Marco JA, Hill BT, Hallek M, Eisenfeld AJ, Stromatt SC, Jaeger U. Randomized phase 2 study of otlertuzumab and bendamustine versus bendamustine in patients with relapsed chronic lymphocytic leukaemia. Br J Haematol 2016; 176:618-628. [PMID: 27977057 PMCID: PMC5324531 DOI: 10.1111/bjh.14464] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/20/2016] [Indexed: 11/27/2022]
Abstract
Otlertuzumab (TRU‐016) is a humanized anti‐CD37 protein therapeutic that triggers direct caspase‐independent apoptosis of malignant B cells and induces antibody‐dependent cell‐mediated cytotoxicity. Patients with relapsed chronic lymphocytic leukaemia (CLL) received either otlertuzumab (20 mg/kg) weekly by IV infusion for two 28‐day cycles then every 14 days for four 28‐day cycles and IV bendamustine (70 mg/m2) on Days 1 and 2 of each cycle for up to six 28‐day cycles or bendamustine alone. Thirty‐two patients were treated with otlertuzumab and bendamustine and 33 with bendamustine alone. Overall response rate according to the International Workshop on Chronic Lymphocytic Leukaemia criteria was 69% in the otlertuzumab and bendamustine arm and 39% in the bendamustine alone arm (P = 0·025). Median progression‐free survival (PFS) was 15·9 months in the otlertuzumab and bendamustine arm and 10·2 months in the bendamustine alone arm (P = 0·0192). There was a higher incidence of pyrexia (34% vs. 12%) and neutropenia (59% vs. 39%) with the combination but this did not result in a higher incidence of severe (grade 3/4) infections (13% vs. 27%). This combination significantly increased the response rate and prolonged the PFS over single agent bendamustine in patients with relapsed or refractory CLL.
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Haematology, Medical University of Lodz and Copernicus Memorial Hospital, Lodz, Poland
| | - Andrzej Hellmann
- Department of Hematology, Medical University of Gdansk, Gdansk, Poland
| | - Janusz Kloczko
- Department of Haematology, Medical University of Bialystok, Bialystok, Poland
| | | | - Ewa Lech-Maranda
- Department of Haematology, Institute of Haematology and Transfusion Medicine, Warsaw, Poland.,Department of Haematology and Transfusion Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - John M Pagel
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Anthony Mato
- Center for CLL, University of Pennsylvania and Hackensack University, Philadelphia, PA, USA
| | - John C Byrd
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Farrukh T Awan
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Holger Hebart
- Department of Haematology/Oncology, Stauferklinikum Schwaebisch Gmuend, Mutlangen, Germany
| | | | - Brian T Hill
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Michael Hallek
- Department I of Internal Medicine, Centre of Integrated Oncology Koln and Cluster of Excellence on Cellular Stress Responses in Aging (CECAD) University of Cologne, Cologne, Germany.,German CLL Study Group
| | | | | | - Ulrich Jaeger
- German CLL Study Group.,Department of Medicine I, Division of Haematology and Haemostaseology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Liu T, Li X, You S, Bhuyan SS, Dong L. Effectiveness of AMD3100 in treatment of leukemia and solid tumors: from original discovery to use in current clinical practice. Exp Hematol Oncol 2016; 5:19. [PMID: 27429863 PMCID: PMC4947283 DOI: 10.1186/s40164-016-0050-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022] Open
Abstract
AMD3100, also known as plerixafor, was originally developed as an anti-human immunodeficiency virus (HIV) drug, and later characterized as a C-X-C chemokine receptor type 4 (CXCR4) antagonist. Previous reviews have focused on the application of AMD3100 in the treatment of HIV, but a comprehensive evaluation of AMD3100 in the treatment of leukemia, solid tumor, and diagnosis is lacking. In this review, we broadly describe AMD3100, including the background, functional mechanism and clinical applications. Until the late 1990s, CXCR4 was known as a crucial factor for hematopoietic stem and progenitor cell (HSPC) retention in bone marrow. Subsequently, the action and synergy of plerixafor with Granulocyte-colony stimulating factor (G-CSF) led to the clinical approval of plerixafor as the first compound for mobilization of HSPCs. The amount of HSPC mobilization and the rapid kinetics promoted additional clinical uses. Recently, CXCR4/CXCL12 (C-X-C motif chemokine 12) axis was found to be involved in a variety of roles in tumors, including leukemic stem cell (LSC) homing and signaling transduction. Thus, CXCR4 targeting has been a treatment strategy against leukemia and solid tumors. Understanding this mechanism will help shed light on therapeutic potential for HIV infection, inflammatory diseases, stem-cell mobilization, leukemia, and solid tumors. Clarifying the CXCR4/CXCL12 axis and role of AMD3100 will help remove malignant cells from the bone marrow niche, rendering them more accessible to targeted therapeutic agents.
Collapse
Affiliation(s)
- Tao Liu
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Department of Oncology, The Affiliated Jiangyin Hospital of Southeast University Medical College, Wuxi, 214400 Jiangsu People's Republic of China
| | - Xiaobo Li
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA.,Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, 300193 China
| | - Shuo You
- Department of Neurosurgery, Winship Cancer Institute, Emory University, Atlanta, GA 30322 USA
| | - Soumitra S Bhuyan
- School of Public Health, Division of Health Systems, Management, and Policy, The University of Memphis, Memphis, TN 38152 USA
| | - Lei Dong
- Division of Hematology/Oncology, Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Emory University School of Medicine, 1760 Haygood Drive NE, HSRB E363, Atlanta, GA 30322 USA
| |
Collapse
|
24
|
A complementary role of multiparameter flow cytometry and high-throughput sequencing for minimal residual disease detection in chronic lymphocytic leukemia: an European Research Initiative on CLL study. Leukemia 2015; 30:929-36. [PMID: 26639181 PMCID: PMC4832072 DOI: 10.1038/leu.2015.313] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 08/14/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022]
Abstract
In chronic lymphocytic leukemia (CLL) the level of minimal residual disease (MRD) after therapy is an independent predictor of outcome. Given the increasing number of new agents being explored for CLL therapy, using MRD as a surrogate could greatly reduce the time necessary to assess their efficacy. In this European Research Initiative on CLL (ERIC) project we have identified and validated a flow-cytometric approach to reliably quantitate CLL cells to the level of 0.0010% (10−5). The assay comprises a core panel of six markers (i.e. CD19, CD20, CD5, CD43, CD79b and CD81) with a component specification independent of instrument and reagents, which can be locally re-validated using normal peripheral blood. This method is directly comparable to previous ERIC-designed assays and also provides a backbone for investigation of new markers. A parallel analysis of high-throughput sequencing using the ClonoSEQ assay showed good concordance with flow cytometry results at the 0.010% (10−4) level, the MRD threshold defined in the 2008 International Workshop on CLL guidelines, but it also provides good linearity to a detection limit of 1 in a million (10−6). The combination of both technologies would permit a highly sensitive approach to MRD detection while providing a reproducible and broadly accessible method to quantify residual disease and optimize treatment in CLL.
Collapse
|
25
|
Eliminating minimal residual disease as a therapeutic end point: working toward cure for patients with CLL. Blood 2015; 127:279-86. [PMID: 26576865 DOI: 10.1182/blood-2015-08-634816] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/04/2015] [Indexed: 12/16/2022] Open
Abstract
Deep remission and prolonged disease-free survival can be achieved with first-line chemoimmunotherapy (CIT), such as combined fludarabine, cyclophosphamide, and rituximab, in the majority of patients with chronic lymphocytic leukemia (CLL). More modest results are reported with less intense regimens like obinutuzumab plus chlorambucil. Clinical assessment has limited sensitivity in detecting residual disease responsible for subsequent relapse, even including morphologic bone marrow (BM) evaluation. Multicolor flow cytometry and polymerase chain reaction (PCR)-based methods can detect minimal residual disease (MRD) to a sensitivity of ≥1:10,000 (10(-4)). Achieving BM MRD-negative complete remission (CR) is associated with superior progression-free survival (PFS) and overall survival; MRD status is the single best posttreatment predictor of long-term outcomes after CIT. Newer oral B-cell receptor signaling pathway inhibitors are highly effective at controlling disease, but best monotherapy responses are typically partial remission, and patients must remain on treatment to maintain disease control. Therapeutic progress is still needed for CLL. We propose that targeting MRD provides opportunity to realize this progress. Achieving BM MRD-negative CR is a prerequisite for long-term unmaintained disease-free survival and potential for cure. We review available methodologies for detecting MRD and correlations with posttreatment outcomes. We discuss the potential utility of MRD to direct individualized therapy. Finally, we discuss the importance of MRD-negative status as a surrogate marker for longer PFS in clinical studies to allow more rapid determination of clinical benefit.
Collapse
|
26
|
Lamanna N. New oral small molecules in the treatment of chronic lymphocytic leukemia. Cancer 2015; 121:1917-26. [PMID: 25690403 DOI: 10.1002/cncr.29130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 09/15/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022]
Abstract
There has been a dramatic change in therapy for chronic lymphocytic leukemia (CLL) over the last 20 years. In 1990, available therapy produced complete responses in <5% of treated patients. This is in marked contrast to modern regimens, which are reported to reliably produce complete responses in approximately 40% to 50% of patients. This remarkable improvement has been attributable to combination chemoimmunotherapy agents that have contributed to the backbone of therapy for patients with CLL. However, the disease is still incurable and these modern treatment regimens have been somewhat limited to the treatment of younger, physically "fit" patients with CLL due to their increased toxicity, including enhanced myelosuppression and immunosuppression. In addition, because patients receive multiple therapies during the course of their lifetime, the mounting toxicities as well as decreased efficacy often limit the repeated use of these more aggressive combination therapies. Fortunately, over the past 5 years, there has been an explosion of new active agents that have demonstrated remarkable activity in patients with recurrent/refractory disease as well as those who harbor poor cytogenetic abnormalities. The current review focuses on some of the novel small molecules that have either been approved or are at the forefront of clinical development in the treatment of patients with CLL.
Collapse
Affiliation(s)
- Nicole Lamanna
- Leukemia Service, Department of Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
27
|
Tempescul A, Bagacean C, Riou C, Bendaoud B, Hillion S, Debant M, Buors C, Berthou C, Renaudineau Y. Ofatumumab capacity to deplete B cells from chronic lymphocytic leukaemia is affected by C4 complement exhaustion. Eur J Haematol 2015; 96:229-35. [DOI: 10.1111/ejh.12573] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Adrian Tempescul
- Department of Clinical Haematology; Institute of Oncology and Haematology; CHRU morvan; Brest France
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
| | - Cristina Bagacean
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
| | - Catherine Riou
- Department of Clinical Haematology; Institute of Oncology and Haematology; CHRU morvan; Brest France
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
| | - Boutahar Bendaoud
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
- Laboratory of Immunology and Immunotherapy; CHRU Morvan; Brest France
| | - Sophie Hillion
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
- Laboratory of Immunology and Immunotherapy; CHRU Morvan; Brest France
| | - Marjolaine Debant
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
| | - Caroline Buors
- Laboratory of Hematology; CHRU Cavale Blanche; Brest France
| | - Christian Berthou
- Department of Clinical Haematology; Institute of Oncology and Haematology; CHRU morvan; Brest France
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
| | - Yves Renaudineau
- INSERM ESPRI ERI29 Laboratory of Immunotherapy and B Cell Diseases; Reseau Epigenetique et Reseau Canaux Ioniques du Cancéropole Grand Ouest; Brest France
- Laboratory of Immunology and Immunotherapy; CHRU Morvan; Brest France
| |
Collapse
|
28
|
Zent CS, Call TG, Bowen DA, Conte MJ, LaPlant BR, Witzig TE, Ansell SM, Weiner GJ. Early treatment of high risk chronic lymphocytic leukemia with alemtuzumab, rituximab and poly-(1-6)-beta-glucotriosyl-(1-3)- beta-glucopyranose beta-glucan is well tolerated and achieves high complete remission rates. Leuk Lymphoma 2015; 56:2373-8. [PMID: 25676035 PMCID: PMC4573786 DOI: 10.3109/10428194.2015.1016932] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Poly-[1-6]-β-glucopyranosyl-[1-3]-β-glucopyranose (PGG) beta glucan is a Saccharomyces cerevisiae derived 1,3/1,6 glucose polymer with innate immune system activation potential. This phase I/II clinical trial enrolled 20 eligible patients with chronic lymphocytic leukemia with high-risk biological markers for early initial treatment with alemtuzumab, rituximab and PGG beta glucan (1-2-4 mg/kg/dose) over 31 days. PGG beta glucan at 4 mg/kg was well tolerated and used for the phase II study. There were three grade 3-4 toxicities at least possibly attributed to treatment. Nineteen (95%) patients responded to treatment with 13 (65%) complete responses. All patients were alive at a median follow-up of 24.4 months (range: 9.5-37). Eleven patients had progressive disease (median 17.6 months, 95% confidence interval [CI]: 9.7, 32.1) and eight patients were retreated (median 35.3 months, 95% CI: 17.9, not reached). We conclude that PGG beta glucan, alemtuzumab and rituximab treatment is tolerable and results in a high complete response rate.
Collapse
MESH Headings
- Aged
- Alemtuzumab
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Chromosome Aberrations
- Disease Progression
- Female
- Follow-Up Studies
- Humans
- Kaplan-Meier Estimate
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Male
- Middle Aged
- Remission Induction
- Rituximab/administration & dosage
- Treatment Outcome
- beta-Glucans/administration & dosage
Collapse
Affiliation(s)
| | | | | | | | - Betsy R. LaPlant
- Department of Health Sciences Research, Mayo Clinic, Rochester MN
| | | | | | - George J. Weiner
- Holden Comprehensive Cancer Center and Department of Internal Medicine, University of Iowa, Iowa City IA
| |
Collapse
|
29
|
Stromal cell-mediated glycolytic switch in CLL cells involves Notch-c-Myc signaling. Blood 2015; 125:3432-6. [PMID: 25778534 DOI: 10.1182/blood-2014-10-607036] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/09/2015] [Indexed: 11/20/2022] Open
Abstract
It is well established that the stromal niche exerts a protective effect on chronic lymphocytic leukemia (CLL) cells, thereby also affecting their drug sensitivity. One hallmark of malignant cells is metabolic reprogramming, which is mostly represented by a glycolytic shift known as the Warburg effect. Because treatment resistance can be linked to metabolic alterations, we investigated whether bone marrow stromal cells impact the bioenergetics of primary CLL cells. In fact, stromal contact led to an increase of aerobic glycolysis and the cells' overall glycolytic capacity accompanied by an increased glucose uptake, expression of glucose transporter, and glycolytic enzymes. Activation of Notch signaling and of its direct transcriptional target c-Myc contributed to this metabolic switch. Based on these observations, CLL cells' acquired increased glucose dependency as well as Notch-c-Myc signaling could be therapeutically exploited in an effort to overcome stroma-mediated drug resistance.
Collapse
|
30
|
Demirci T, Yeğin ZA, Kurşunoğlu N, Yılmaz Z, Suyanı E, Özkurt ZN, Yağcı M. Prevalence of monoclonal B lymphocytosis in first-degree relatives of chronic lymphocytic leukemia patients in Turkey. Turk J Haematol 2015; 32:29-34. [PMID: 25805672 PMCID: PMC4439904 DOI: 10.4274/tjh.2013.0288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: Monoclonal B lymphocytosis (MBL) is considered to be a precursor state for chronic lymphocytic leukemia (CLL). This study was planned to evaluate the MBL prevalence in first-degree relatives of CLL patients in Turkey, which is considered to be an ethnic and geographic bridge between the Eastern and Western worlds. Materials and Methods: A total of 136 volunteers [median age: 40 (17-77) years; male/female: 60/76] from 61 families were included. Flow cytometry analysis by 4-colour staining was used for MBL diagnosis. Results: MBL was demonstrated in 17 cases (12.5%). A total of 14 cases (10.3%) were classified as CLL-like MBL, while 3 (2.2%) exhibited a non-CLL-like phenotype. The prevalence of MBL was 12.72% in subjects aged less than 40 years, 12.28% in subjects between 40 and 60 years, and 40% in subjects over 60 years, without statistical significance (p>0.05). A total of 115 cases were evaluated for intermarriage, which was observed in 19 cases (16.5%). The prevalence of MBL did not differ based on intermarriage status (p>0.05). Conclusion: The current report is the first MBL prevalence study in a Eurasian population that demonstrates a similar distribution pattern of MBL in Anatolian CLL kindreds. Further efforts should be made to refine our understanding of the natural history and clinical outcomes of MBL.
Collapse
Affiliation(s)
- Taner Demirci
- Gazi University Faculty of Medicine, Department of Internal Medicine, Ankara, Turkey. E-mail:
| | | | | | | | | | | | | |
Collapse
|
31
|
Keeney M, Halley JG, Rhoads DD, Ansari MQ, Kussick SJ, Karlon WJ, Mehta KU, Dorfman DM, Linden MA. Marked Variability in Reported Minimal Residual Disease Lower Level of Detection of 4 Hematolymphoid Neoplasms: A Survey of Participants in the College of American Pathologists Flow Cytometry Proficiency Testing Program. Arch Pathol Lab Med 2015; 139:1276-80. [DOI: 10.5858/arpa.2014-0543-cp] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context
Flow cytometry is often applied to minimal residual disease (MRD) testing in hematolymphoid neoplasia. Because flow-based MRD tests are developed in the laboratory, testing methodologies and lower levels of detection (LODs) are laboratory dependent.
Objectives
To broadly survey flow cytometry laboratories about MRD testing in laboratories, if performed, including indications and reported LODs.
Design
Voluntary supplemental questions were sent to the 549 laboratories participating in the College of American Pathologists (CAP) FL3-A Survey (Flow Cytometry—Immunophenotypic Characterization of Leukemia/Lymphoma) in the spring of 2014.
Results
A total of 500 laboratories (91%) responded to the supplemental questions as part of the FL3-A Survey by April 2014; of those 500 laboratories, 167 (33%) currently perform MRD for lymphoblastic leukemia, 118 (24%) for myeloid leukemia, 99 (20%) for chronic lymphocytic leukemia, and 91 (18%) for plasma cell myeloma. Other indications include non-Hodgkin lymphoma, hairy cell leukemia, neuroblastoma, and myelodysplastic syndrome. Most responding laboratories that perform MRD for lymphoblastic leukemia reported an LOD of 0.01%. For myeloid leukemia, chronic lymphocytic leukemia, and plasma cell myeloma, most laboratories indicated an LOD of 0.1%. Less than 3% (15 of 500) of laboratories reported LODs of 0.001% for one or more MRD assays performed.
Conclusions
There is major heterogeneity in the reported LODs of MRD testing performed by laboratories subscribing to the CAP FL3-A Survey. To address that heterogeneity, changes to the Flow Cytometry Checklist for the CAP Laboratory Accreditation Program are suggested that will include new requirements that each laboratory (1) document how an MRD assay's LOD is measured, and (2) include the LOD or lower limit of enumeration for flow-based MRD assays in the final diagnostic report.
Collapse
Affiliation(s)
- Michael Keeney
- From the Department of Hematology, London Health Sciences Centre, London, Ontario, Canada (Mr Keeney); the Department of Surveys, College of American Pathologists, Northfield, Illinois (Ms Halley); the Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (Dr Rhoads); the Department of Clinical Pathology, Cleveland Clinic, Cleveland, Ohio (Dr Ansari); PhenoPat
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Prognosis and therapy of chronic lymphocytic leukemia and small lymphocytic lymphoma. Cancer Treat Res 2015; 165:147-75. [PMID: 25655609 DOI: 10.1007/978-3-319-13150-4_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by a highly variable clinical course that has guided treatment principles in as much as anti-leukemic therapy is reserved for patients with active disease. This heterogeneity is somewhat dissected by prognostic markers, many of which represent pathogenic mechanisms. Recently, the introduction of highly active targeted agents and maturing data on predictive markers may lead to more individualized therapeutic approaches. In this chapter, we review key prognostic markers, current and emerging therapy, and will attempt to outline a future "where the two may connect".
Collapse
|
33
|
Janel A, Dubois-Galopin F, Bourgne C, Berger J, Tarte K, Boiret-Dupré N, Boisgard S, Verrelle P, Déchelotte P, Tournilhac O, Berger MG. The Chronic Lymphocytic Leukemia Clone Disrupts the Bone Marrow Microenvironment. Stem Cells Dev 2014; 23:2972-82. [DOI: 10.1089/scd.2014.0229] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Alexandre Janel
- Hématologie Biologique, CHU (University Hospital Center) Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
- EA7283 CREaT, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | | | - Céline Bourgne
- Hématologie Biologique, CHU (University Hospital Center) Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
| | - Juliette Berger
- Hématologie Biologique, CHU (University Hospital Center) Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
| | - Karin Tarte
- INSERM U917–MICA, University of Medicine, Rennes, France
| | - Nathalie Boiret-Dupré
- Hématologie Biologique, CHU (University Hospital Center) Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
- EA7283 CREaT, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | - Stéphane Boisgard
- Orthopédie Traumatologie, CHU (University Hospital Center) Clermont-Fd, Hospital Montpied, Clermont-Ferrand, France
| | - Pierre Verrelle
- EA7283 CREaT, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
| | - Pierre Déchelotte
- Anatomie Pathologique, CHU Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
| | - Olivier Tournilhac
- EA7283 CREaT, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- Hématologie Clinique Adulte, CHU Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
| | - Marc G. Berger
- Hématologie Biologique, CHU (University Hospital Center) Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
- EA7283 CREaT, Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- Hématologie Clinique Adulte, CHU Clermont-Fd, Hopital Estaing, Clermont-Ferrand, France
| |
Collapse
|
34
|
Overexpression of EZH2 associates with a poor prognosis in chronic lymphocytic leukemia. Blood Cells Mol Dis 2014; 54:97-102. [PMID: 25131810 DOI: 10.1016/j.bcmd.2014.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 07/24/2014] [Indexed: 11/20/2022]
Abstract
EZH2, a histone methyltransferase, is overexpressed in several human tumors, but whether it exerts any impact in chronic lymphocytic leukemia (CLL) remains unknown. We used real time PCR to investigate the expression profile of EZH1 and EZH2 in 59 CLL patients, 10 samples of purified B-cells from healthy donors and 12 normal adult tissues. EZH2 was overexpressed in CLL patients and correlates with high white blood cell count, ZAP-70 expression and chromosomal abnormalities. EHZ1 expression does not correlate with CLL progression. EZH2 overexpression is related to a poor prognosis of CLL and could be a useful tool to assess its aggressiveness.
Collapse
|
35
|
Johansson U, Bloxham D, Couzens S, Jesson J, Morilla R, Erber W, Macey M. Guidelines on the use of multicolour flow cytometry in the diagnosis of haematological neoplasms. British Committee for Standards in Haematology. Br J Haematol 2014; 165:455-88. [PMID: 24620735 DOI: 10.1111/bjh.12789] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
36
|
Raponi S, Della Starza I, De Propris MS, Del Giudice I, Mauro FR, Marinelli M, Di Maio V, Piciocchi A, Foà R, Guarini A. Minimal residual disease monitoring in chronic lymphocytic leukaemia patients. A comparative analysis of flow cytometry and ASO IgH RQ-PCR. Br J Haematol 2014; 166:360-8. [PMID: 24735016 DOI: 10.1111/bjh.12887] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022]
Abstract
Minimal residual disease (MRD) is becoming increasingly important in chronic lymphocytic leukaemia (CLL) as treatment strategies are progressively improving. The primary objective of this study was to compare the applicability of three different flow cytometric approaches: basic 4-colour analysis, European Research Initiative in CLL (ERIC) consensus method and 8-colour analysis. Secondly, we investigated the sensitivity and specificity of flow cytometry (FC) compared to molecular analyses for MRD detection. A total of 462 CLL samples were evaluated by basic FC; in 143, ERIC consensus method was also performed and all three FC methodologies were applied in a subgroup of 10 cases. No discordance in defining MRD-positive/negative samples was observed between the FC methods; within positive samples, the ERIC consensus method and 8-colour analysis showed the most accurate results. MRD was analysed by FC and polymerase chain reaction (PCR) in 243 cases: concordant results were obtained in 199/243 samples (81·9%); 42/243 were FC-/PCR+. Overall, the sensitivity and specificity of FC compared to PCR was 96·5% and 77·2%, respectively. Both FC and PCR proved suitable for the detection of MRD and prediction of progression-free survival, which was significantly reduced in MRD-positive patients, regardless of the methodology. These results offer the rationale for a strategy to monitor MRD in CLL patients.
Collapse
Affiliation(s)
- Sara Raponi
- Haematology, Department of Cellular Biotechnologies and Haematology, Sapienza University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Targeting the proliferative and chemoresistant compartment in chronic lymphocytic leukemia by inhibiting survivin protein. Leukemia 2014; 28:1993-2004. [PMID: 24618734 DOI: 10.1038/leu.2014.96] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/20/2014] [Accepted: 03/04/2014] [Indexed: 12/19/2022]
Abstract
Chronic lymphocytic leukemia (CLL) cells located in proliferation centers are constantly stimulated by accessory cells, which provide them with survival and proliferative signals and mediate chemotherapy resistance. Herein, we designed an experimental strategy with the aim of mimicking the microenvironment found in the proliferative centers to specifically target actively proliferating CLL cells. For this, we co-cultured CLL cells and bone marrow stromal cells with concomitant CD40 and Toll-like receptor 9 stimulation. This co-culture system induced proliferation, cell-cycle entry and marked resistance to treatment with fludarabine and bendamustine. Proliferating CLL cells clustered together showed a typical morphology of activated B cells and expressed survivin protein, a member of the inhibitor of apoptosis family that is mainly expressed by CLL cells in the proliferation centers. With the aim of specifically targeting actively proliferating and chemoresistant CLL cells, we investigated the effects of treatment with YM155, a small-molecule survivin inhibitor. YM155 treatment suppressed the co-culture-induced survivin expression and that was sufficient to inhibit proliferation and effectively induce apoptosis particularly in the proliferative subset of CLL cells. Interestingly, sensitivity to YM155 was independent from common prognostic markers, including 17p13.1 deletion. Altogether, these findings provide a rationale for clinical development of YM155 in CLL.
Collapse
|
38
|
Abstract
Circulating monoclonal B cells may be detected in healthy adults, a condition called monoclonal B-cell lymphocytosis (MBL). MBL has also been identified in donated blood, but no systematic study of blood donors has been reported. Using sensitive and specific laboratory methods, we detected MBL in 149 (7.1%; 95% confidence interval, 6.0% to 8.3%) of 2098 unique donors ages 45 years or older in a Midwestern US regional blood center between 2010 and 2011. Most of the 149 donors had low-count MBL, including 99 chronic lymphocytic leukemia-like (66.4%), 22 atypical (14.8%), and 19 CD5(-) (12.8%) immunophenotypes. However, 5 donors (3.4%) had B-cell clonal counts above 500 cells per µL, including 3 with 1693 to 2887 cells per µL; the clone accounted for nearly all their circulating B cells. Four donors (2.7%) had 2 distinct MBL clones. Of 51 MBL samples in which immunoglobulin heavy chain (IGH)V-D-J genotypes could be determined, 71% and 29% used IGHV3- and IGHV4-family genes, respectively. Sequencing revealed 82% with somatic hypermutation, whereas 18% had >98% germ-line identity, including 5 with entirely germ-line sequences. In conclusion, MBL prevalence is much higher in blood donors than previously reported, and although uncommon, the presence of high-count MBL warrants further investigations to define the biological fate of the transfused cells in recipients.
Collapse
|
39
|
Hedley BD, Keeney M. Technical issues: flow cytometry and rare event analysis. Int J Lab Hematol 2013; 35:344-50. [PMID: 23590661 DOI: 10.1111/ijlh.12068] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 01/14/2013] [Indexed: 12/21/2022]
Abstract
Flow cytometry has become an essential tool for identification and characterization of hematological cancers and now, due to technological improvements, allows the identification and rapid enumeration of small tumor populations that may be present after induction therapy (minimal residual disease, MRD). The quantitation of MRD has been shown to correlate with relapse and survival rates in numerous diseases and in certain cases, and evidence of MRD is used to alter treatment protocols. Recent improvements in hardware allow for high data rate collection. Improved fluorochromes take advantage of violet laser excitation and maximize signal-to-noise ratio allowing the population of interest to be isolated in multiparameter space. This isolation, together with a low background rate, permits for detection of residual tumor populations in a background of normal cells. When counting such rare events, the distribution is governed by Poisson statistics, with precision increasing with higher numbers of cells collected. In several hematological malignancies, identification of populations at frequencies of 0.01% and lower has been attained. The choice of antibodies used in MRD detection facilitates the definition of a fingerprint to identify abnormal populations throughout treatment. Tumor populations can change phenotype, and an approach that relies on 'different from normal' has proven useful, particularly in the acute leukemias. Flow cytometry can and is used for detection of MRD in many hematological diseases; however, standardized approaches for specific diseases must be developed to ensure precise identification and enumeration that may alter the course of patient treatment.
Collapse
Affiliation(s)
- B D Hedley
- Special Hematology, London Health Sciences Centre, London, ON, Canada
| | | |
Collapse
|
40
|
The Role of Minimal Residual Disease Measurements in the Therapy for CLL. Hematol Oncol Clin North Am 2013; 27:267-88. [DOI: 10.1016/j.hoc.2013.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Kempin S. Update on Chronic Lymphocytic Leukemia: Overview of New Agents and Comparative Analysis. Curr Treat Options Oncol 2013; 14:144-55. [DOI: 10.1007/s11864-013-0229-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
42
|
Böttcher S, Ritgen M, Kneba M. Flow cytometric MRD detection in selected mature B-cell malignancies. Methods Mol Biol 2013; 971:149-174. [PMID: 23296963 DOI: 10.1007/978-1-62703-269-8_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The quantification of submicroscopic minimal residual disease (MRD) after therapy proved to have independent prognostic significance in many mature B-cell malignancies. With the advent of routine bench-top cytometers capable of simultaneously analyzing ≥ 4 colors and with improved standardization, flow cytometry has become the method of choice for MRD assessments in some lymphoma entities. Herein we describe general aspects of flow cytometric standardization. Using chronic lymphocytic leukemia (CLL) and mantle cell lymphoma (MCL) as examples we explain in detail the application of flow cytometry for MRD detection.
Collapse
MESH Headings
- Bone Marrow Cells/pathology
- Flow Cytometry/instrumentation
- Flow Cytometry/methods
- Flow Cytometry/standards
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukocytes, Mononuclear/pathology
- Light
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/pathology
- Neoplasm, Residual/diagnosis
- Polymerase Chain Reaction
- Reference Standards
- Scattering, Radiation
- Staining and Labeling
Collapse
Affiliation(s)
- Sebastian Böttcher
- Second Department of Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany.
| | | | | |
Collapse
|
43
|
A look into the future: can minimal residual disease guide therapy and predict prognosis in chronic lymphocytic leukemia? Hematology 2012. [DOI: 10.1182/asheducation.v2012.1.97.3798195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Over the past 2 decades, dramatic improvements in the efficacy of treatments for chronic lymphocytic leukemia have led to progressively higher percentages of clinical complete remissions. A molecular eradication of the leukemia has become not only a desirable, but also an achievable, end point that needs to be evaluated within clinical trials. The assessment of complete remission only at the clinical and morphological level is insufficient, at least for physically fit patients. The detection of minimal residual disease (MRD) in chronic lymphocytic leukemia has become feasible using PCR-based or flow cytometric techniques that reproducibly allow reaching the detection level of less than 1 leukemic cell per 10 000 leukocytes (10−4), the level currently defined as MRD− status. Emerging data indicate that the MRD status during and at the end of treatment is one of the most powerful predictors of progression-free and overall survival. This predictor appears to be independent of clinical response, type or line of therapy, and known biological markers. For these reasons, the time is ripe to test the use of MRD as a surrogate marker of clinical end points and as a real-time marker of efficacy and/or resistance to the administered therapies. In the near future, clinical trials will determine whether MRD assessment can be used for guiding therapy, either to improve quality of responses through consolidation or to prevent relapses through preemptive therapies based on the reappearance of MRD.
Collapse
|
44
|
Sivina M, Hartmann E, Vasyutina E, Boucas JM, Breuer A, Keating MJ, Wierda WG, Rosenwald A, Herling M, Burger JA. Stromal cells modulate TCL1 expression, interacting AP-1 components and TCL1-targeting micro-RNAs in chronic lymphocytic leukemia. Leukemia 2012; 26:1812-20. [PMID: 22460735 DOI: 10.1038/leu.2012.63] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 01/31/2012] [Accepted: 02/21/2012] [Indexed: 02/03/2023]
Abstract
The tissue microenvironment in chronic lymphocytic leukemia (CLL) has an increasingly recognized role in disease progression, but the molecular mechanisms of cross talk between CLL cells and their microenvironment remain incompletely defined. Bone marrow stromal cells (BMSC) protect CLL cells from apoptosis in a contact-dependent fashion, and have been used for the identification of key pathways such as the CXCR4-CXCL12 axis. To further dissect the molecular impact of BMSC on survival and the molecular activation signature of CLL cells, we co-cultured CLL cells with different BMSC. Gene expression profiling of CLL cells revealed that the lymphoid proto-oncogene TCL1 was among the top genes upregulated in CLL cells by BMSC. TCL1 mRNA and protein upregulation by BMSC was paralleled by decreases of TCL1-interacting FOS/JUN, and confirmed by qRT-PCR, immunoblotting, immunoprecipitations, and flow cytometry. Stroma mediated increases in TCL1 were also associated with decreased levels of TCL1-regulatory micro-RNAs (miR-29b, miR-181b, miR-34b). These findings demonstrate that the microenvironment has a proactive role in the regulation of the known signaling enhancer and pro-survival molecule TCL1 in CLL. This provides a further rationale for therapeutically targeting the cross talk between CLL and BMSC.
Collapse
Affiliation(s)
- M Sivina
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Improving efficiency and sensitivity: European Research Initiative in CLL (ERIC) update on the international harmonised approach for flow cytometric residual disease monitoring in CLL. Leukemia 2012; 27:142-9. [PMID: 23041722 DOI: 10.1038/leu.2012.216] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Detection of minimal residual disease (MRD) in chronic lymphocytic leukaemia (CLL) is becoming increasingly important as treatments improve. An internationally harmonised four-colour (CLR) flow cytometry MRD assay is widely used but has limitations. The aim of this study was to improve MRD analysis by identifying situations where a less time-consuming CD19/CD5/κ/λ analysis would be sufficient for detecting residual CLL, and develop a six-CLR antibody panel that is more efficient for cases requiring full MRD analysis. In 784 samples from CLL patients after treatment, it was possible to determine CD19/CD5/κ/λ thresholds that identified cases with detectable MRD with 100% positive predictive value (PPV). However, CD19/CD5/κ/λ analysis was unsuitable for predicting iwCLL/NCI response status or identifying cases with no detectable MRD. For the latter cases requiring a full MRD assessment, a six-CLR assay was designed comprising CD19/CD5/CD20 with (1) CD3/CD38/CD79b and (2) CD81/CD22/CD43. There was good correlation between four-CLR and six-CLR panels in dilution studies and clinical samples, with 100% concordance for detection of residual disease at the 0.01% (10(-4)) level (n=59) and good linearity even at the 0.001-0.01% (10(-5)-10(-4)) level. A six-CLR panel therefore provides equivalent results to the four-CLR panel but it requires fewer reagents, fewer cells and a much simpler analysis approach.
Collapse
|
46
|
Human peripheral blood mononuclear cells exhibit heterogeneous CD52 expression levels and show differential sensitivity to alemtuzumab mediated cytolysis. PLoS One 2012; 7:e39416. [PMID: 22761788 PMCID: PMC3382607 DOI: 10.1371/journal.pone.0039416] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 05/24/2012] [Indexed: 11/25/2022] Open
Abstract
Alemtuzumab is a monoclonal antibody that targets cell surface CD52 and is effective in depleting lymphocytes by cytolytic effects in vivo. Although the cytolytic effects of alemtuzumab are dependent on the density of CD52 antigen on cells, there is scant information regarding the expression levels of CD52 on different cell types. In this study, CD52 expression was assessed on phenotypically distinct subsets of lymphoid and myeloid cells in peripheral blood mononuclear cells (PBMCs) from normal donors. Results demonstrate that subsets of PBMCs express differing levels of CD52. Quantitative analysis showed that memory B cells and myeloid dendritic cells (mDCs) display the highest number while natural killer (NK) cells, plasmacytoid dendritic cells (pDCs) and basophils have the lowest number of CD52 molecules per cell amongst lymphoid and myeloid cell populations respectively. Results of complement dependent cytolysis (CDC) studies indicated that alemtuzumab mediated profound cytolytic effects on B and T cells with minimal effect on NK cells, basophils and pDCs, correlating with the density of CD52 on these cells. Interestingly, despite high CD52 levels, mDCs and monocytes were less susceptible to alemtuzumab-mediated CDC indicating that antigen density alone does not define susceptibility. Additional studies indicated that higher expression levels of complement inhibitory proteins (CIPs) on these cells partially contributes to their resistance to alemtuzumab mediated CDC. These results indicate that alemtuzumab is most effective in depleting cells of the adaptive immune system while leaving innate immune cells relatively intact.
Collapse
|
47
|
Cecic IK, Li G, MacAulay C. Technologies supporting analytical cytology: clinical, research and drug discovery applications. JOURNAL OF BIOPHOTONICS 2012; 5:313-326. [PMID: 22271675 DOI: 10.1002/jbio.201100093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 12/08/2011] [Accepted: 12/30/2011] [Indexed: 05/31/2023]
Abstract
The tools and techniques developed for analytical cytology have become invaluable in expanding the development of cancer screening programs and biomarker discovery for personalized medicine. Detecting cellular, molecular, and functional changes of diseased tissue as defined by quantitative analytical methodologies has enhanced the field of medical diagnostics and prognostics. The focus of this review is to outline applications and recent technical advances in flow cytometry, laser scanning cytometry, image cytometry, and quantitative image analysis, as they pertain to clinical, research, and drug discovery applications.
Collapse
Affiliation(s)
- Ivana K Cecic
- Integrative Oncology Department, BC Cancer Research Centre, Vancouver, BC, Canada
| | | | | |
Collapse
|
48
|
Böttcher S, Ritgen M, Fischer K, Stilgenbauer S, Busch RM, Fingerle-Rowson G, Fink AM, Bühler A, Zenz T, Wenger MK, Mendila M, Wendtner CM, Eichhorst BF, Döhner H, Hallek MJ, Kneba M. Minimal Residual Disease Quantification Is an Independent Predictor of Progression-Free and Overall Survival in Chronic Lymphocytic Leukemia: A Multivariate Analysis From the Randomized GCLLSG CLL8 Trial. J Clin Oncol 2012; 30:980-8. [DOI: 10.1200/jco.2011.36.9348] [Citation(s) in RCA: 351] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose To determine the clinical significance of flow cytometric minimal residual disease (MRD) quantification in chronic lymphocytic leukemia (CLL) in addition to pretherapeutic risk factors and to compare the prognostic impact of MRD between the arms of the German CLL Study Group CLL8 trial. Patients and Methods MRD levels were prospectively quantified in 1,775 blood and bone marrow samples from 493 patients randomly assigned to receive fludarabine and cyclophosphamide (FC) or FC plus rituximab (FCR). Patients were categorized by MRD into low- (< 10−4), intermediate- (≥ 10−4 to <10−2), and high-level (≥ 10−2) groups. Results Low MRD levels during and after therapy were associated with longer progression-free survival (PFS) and overall survival (OS; P < .0001). Median PFS is estimated at 68.7, 40.5, and 15.4 months for low, intermediate, and high MRD levels, respectively, when assessed 2 months after therapy. Compared with patients with low MRD, greater risks of disease progression were associated with intermediate and high MRD levels (hazard ratios, 2.49 and 14.7, respectively; both P < .0001). Median OS was 48.4 months in patients with high MRD and was not reached for lower MRD levels. MRD remained predictive for OS and PFS in multivariate analyses that included the most important pretherapeutic risk markers in CLL. PFS and OS did not differ between treatment arms within each MRD category. However, FCR induced low MRD levels more frequently than FC. Conclusion MRD levels independently predict OS and PFS in CLL. Therefore, MRD quantification might serve as a surrogate marker to assess treatment efficacy in randomized trials before clinical end points can be evaluated.
Collapse
Affiliation(s)
- Sebastian Böttcher
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Matthias Ritgen
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Kirsten Fischer
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Stephan Stilgenbauer
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Raymonde M. Busch
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Günter Fingerle-Rowson
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Anna Maria Fink
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Andreas Bühler
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Thorsten Zenz
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Michael Karl Wenger
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Myriam Mendila
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Clemens-Martin Wendtner
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Barbara F. Eichhorst
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Hartmut Döhner
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Michael J. Hallek
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| | - Michael Kneba
- Sebastian Böttcher, Matthias Ritgen, and Michael Kneba, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel; Kirsten Fischer, Günter Fingerle-Rowson, Anna Maria Fink, Clemens-Martin Wendtner, Barbara F. Eichhorst, and Michael J. Hallek, University of Cologne, Cologne; Stephan Stilgenbauer, Andreas Bühler, Thorsten Zenz, and Hartmut Döhner, University of Ulm, Ulm; Raymonde M. Busch, Technical University, Munich, Germany; and Michael Karl Wenger and Myriam Mendila, Hoffmann-La Roche, Basel,
| |
Collapse
|
49
|
Bazargan A, Tam CS, Keating MJ. Predicting survival in chronic lymphocytic leukemia. Expert Rev Anticancer Ther 2012; 12:393-403. [PMID: 22369330 DOI: 10.1586/era.12.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
There is increasing interest in the use of prognostic markers that may predict survival and guide management in patients diagnosed with the early stages of chronic lymphocytic leukemia (CLL). Currently, the most important traditional prognostic factors include clinical staging, lymphocyte doubling time and β2-microglobulin/thymidine kinase; and the most important novel markers include karyotypic aberrations (typically assessed by FISH probes or CpG oligonucleotide karyotyping) and IgVH mutation status. Although each of these factors have individually shown significant correlations with survival, there is increasing appreciation that the most complete information may be obtained by using a combination of several factors in prognostic normograms or models. In this article, we review the current state-of-the-art with regards to CLL prognostic factors and discuss how they can be applied in the clinic.
Collapse
Affiliation(s)
- Ali Bazargan
- Hematology Department, St Vincent's Hospital, Melbourne, VIC, Australia
| | | | | |
Collapse
|
50
|
Pott C. Minimal residual disease detection in mantle cell lymphoma: technical aspects and clinical relevance. Semin Hematol 2012; 48:172-84. [PMID: 21782059 DOI: 10.1053/j.seminhematol.2011.05.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The prognostic impact of minimal residual disease (MRD) has been demonstrated for several hematologic malignancies. While in acute lymphoblastic leukemias MRD assessment by polymerase chain reaction (PCR)-based methods has been established as an important tool for clinical risk assessment and is part of clinical management, data demonstrating a prognostic value of MRD in mantle cell lymphoma (MCL) were sparse and results from randomized trials have been published only recently. In the present review technical aspects of different MRD detection methods are discussed, as well as the prognostic relevance of MRD in the context of clinical trials in patients with MCL. Furthermore, recommendations are given for workflow and useful implication of MRD in future clinical trials design.
Collapse
Affiliation(s)
- Christiane Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| |
Collapse
|