1
|
Braish J, Cerchione C, Ferrajoli A. An overview of prognostic markers in patients with CLL. Front Oncol 2024; 14:1371057. [PMID: 38817892 PMCID: PMC11137234 DOI: 10.3389/fonc.2024.1371057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 06/01/2024] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a low-grade B-cell lymphoproliferative disorder. It is the most prevalent type of leukemia in the western countries, with a median age at diagnosis of 70 years. In 2023, it is estimated that there will be 18,740 new cases of CLL, and an estimated 4,490 people will die of this disease. It represents 1.0% of all new cancer cases in the U.S. The rate of new cases was 4.6 per 100,000 men and women per year based on 2016-2020 cases, age-adjusted. Death rates from CLL are higher among older adults, or those 75 and older. The death rate was 1.1 per 100,000 men and women per year based on 2016-2020 deaths, age-adjusted. A common question that patients with CLL ask during their first clinic visit is: "How long will it be before I would need treatment?" Although this might seem like a simple question, the answer is not straight forward. CLL is a heterogenous disease, with a variable clinical course. Some patients may present with an aggressive disease requiring early initiation of treatment, while others have an indolent course and some, having so called smoldering CLL, may never need treatment. The variability in disease course can make predicting disease prognosis a complicated process. This brings forth the importance of establishing prognostic models that can predict disease course, time to treatment, and survival outcomes in such a heterogenous disease. The Rai and Binet staging systems were developed in the late 1970s to early 1980s. They separated patients into different stages based on clinical characteristics and laboratory findings. These simple staging systems are still in use; however, several prognostic markers need to be added for an individualized assessment and, with the recent development of genomic techniques leading to better understanding of CLL at the molecular level, newer prognostic markers have emerged.
Collapse
Affiliation(s)
- Julie Braish
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Claudio Cerchione
- Hematology Unit, Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori” (IRST), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Meldola, Italy
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
2
|
Moia R, Gaidano G. Prognostication in chronic lymphocytic leukemia. Semin Hematol 2024; 61:83-90. [PMID: 38523019 DOI: 10.1053/j.seminhematol.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/26/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is the most common type of leukemia in Western countries. CLL is a highly heterogeneous disease: some patients may never require therapy and others relapse several times after different therapeutic strategies. Therefore, in CLL, prognostic markers are essential to capture high-risk patients for different clinical endpoints including early treatment requirement, early progression after BTK or BCL2 inhibitors and Richter transformation. In early stage CLL, different biological and clinical biomarkers have been identified to predict time to treatment requirement that could be used to identify the most appropriate population for early intervention clinical trial. However, at the moment, the standard of care for early stage CLL remains watch & wait since no survival benefit has been identified in clinical trials with chemoimmunotherapy and with BTK inhibitors. In patients requiring treatment TP53 disruptions identify high-risk patients who benefit the most from long-term continuous therapy with BTKi. On the opposite side of the spectrum, IGHV mutated patients devoid of TP53 disruption benefit the most from fixed-duration therapy with venetoclax-obinutuzumab. In between, the highly heterogenous subgroup of patients with IGHV unmutated genes represents the group in which further efforts are needed to identify additional prognostic biomarkers aimed at selecting patients who can benefit from fixed-duration and patients who can benefit from long term BTKi therapy. In the context of the aggressive transformation of CLL, namely Richter syndrome, the clonal relationship to the CLL counterpart represents the strongest prognostic biomarker. Clonally related Richter syndrome still represents an unmet clinical need which requires further efforts to identify new therapeutic strategies.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Humans
- Prognosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Mutation
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
3
|
Del Giudice I, Foà R. Measurable residual disease in chronic lymphocytic leukemia. Where do we stand? Leukemia 2023; 37:2339-2342. [PMID: 37838758 DOI: 10.1038/s41375-023-02066-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/16/2023]
Affiliation(s)
- Ilaria Del Giudice
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| |
Collapse
|
4
|
Thompson PA, Bazinet A, Wierda WG, Tam CS, O'Brien SM, Saha S, Peterson CB, Plunkett W, Keating MJ. Sustained remissions in CLL after frontline FCR treatment with very-long-term follow-up. Blood 2023; 142:1784-1788. [PMID: 37595283 PMCID: PMC11383921 DOI: 10.1182/blood.2023020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/20/2023] Open
Abstract
Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab (FCR) achieves durable remissions, with flattening of the progression-free survival (PFS) curve in patients with mutated immunoglobulin heavy chain variable gene (IGHV-M). We updated long-term follow-up results from the original 300-patient FCR study initiated at MD Anderson in 1999. The current median follow-up is 19.0 years. With this extended follow-up, the median PFS for patients with IGHV-M was 14.6 years vs 4.2 years for patients with unmutated IGHV (IGHV-UM). Disease progression beyond 10 years was uncommon. In total, 16 of 94 (17%) patients in remission at 10 years subsequently progressed with the additional follow-up compared with the patients in our prior report in 2015. Only 4 of 45 patients (9%) with IGHV-M progressed beyond 10 years. Excluding Richter transformation, 96 of 300 patients (32%) developed 106 other malignancies, with 19 of 300 (6.3%) developing therapy-related myeloid neoplasms (tMNs), which were fatal in 16 of 19 (84%). No pretreatment patient characteristics predicted the risk of tMNs. In summary, FCR remains an option for patients with IGHV-M chronic lymphocytic leukemia (CLL), with a significant fraction achieving functional cure of CLL. A risk-benefit assessment is warranted when counseling patients, balancing potential functional cure with the risk of late relapses and serious secondary malignancies.
Collapse
Affiliation(s)
- Philip A Thompson
- Clinical Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
- Clinical Haematology, Royal Melbourne Hospital, Parkville, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Constantine S Tam
- Department of Haematology, The Alfred Hospital, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Susan M O'Brien
- Department of Medicine, UCI Health Chao Family Comprehensive Cancer Center, Orange, CA
| | - Satabdi Saha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Christine B Peterson
- Department of Developmental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William Plunkett
- Department of Developmental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
5
|
Couillez G, Morel P, Clichet V, Fourdrain L, Delette C, Harrivel V, Gubler B, Rottier C, Derreumaux S, Margat E, Garcon L, Marolleau JP, Boyer T. Flow cytometry as a fast, cost-effective tool to assess IGHV mutational status in CLL. Blood Adv 2023; 7:4701-4704. [PMID: 36287221 PMCID: PMC10468354 DOI: 10.1182/bloodadvances.2022008033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Guillaume Couillez
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Pierre Morel
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Valentin Clichet
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Ludivine Fourdrain
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Caroline Delette
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Véronique Harrivel
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Brigitte Gubler
- Laboratoire d’Oncobiologie Moléculaire, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR 4666, Université Picardie Jules Verne, Amiens, France
| | - Camille Rottier
- Laboratoire d’Oncobiologie Moléculaire, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Sophie Derreumaux
- Laboratoire d’Hématologie, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Emilie Margat
- Laboratoire d’Hématologie, Centre Hospitalier de Lens, Lens, France
| | - Loic Garcon
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR 4666, Université Picardie Jules Verne, Amiens, France
| | - Jean-Pierre Marolleau
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR 4666, Université Picardie Jules Verne, Amiens, France
| | - Thomas Boyer
- Service d’Hématologie Biologique, Centre de Biologie Humaine, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
- UR 4666, Université Picardie Jules Verne, Amiens, France
| |
Collapse
|
6
|
Michallet AS, Letestu R, Le Garff-Tavernier M, Campos L, Ticchioni M, Dilhuydy MS, Morisset S, Rouille V, Mahé B, Laribi K, Villemagne B, Ferrant E, Tournilhac O, Delmer A, Molina L, Leblond V, Tomowiak C, de Guibert S, Orsini-Piocelle F, Banos A, Carassou P, Cartron G, Fornecker LM, Ysebaert L, Dartigeas C, Truchan-Graczyk M, Vilque JP, Schleinitz TA, Cymbalista F, Leprêtre S, Lévy V, Nguyen-Khac F, Feugier P. A fixed-duration immunochemotherapy approach in CLL: 5.5-year results from the phase 2 ICLL-07 FILO trial. Blood Adv 2023; 7:3936-3945. [PMID: 37026799 PMCID: PMC10410135 DOI: 10.1182/bloodadvances.2022009594] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/08/2023] Open
Abstract
In previously untreated, medically fit patients with chronic lymphocytic leukemia (CLL), research is focused on developing fixed-duration strategies to improve long-term outcomes while sparing patients from serious toxicities. The ICLL-07 trial evaluated a fixed-duration (15-month) immunochemotherapy approach in which after obinutuzumab-ibrutinib induction for 9 months, patients (n = 10) in complete remission (CR) with bone marrow (BM) measurable residual disease (MRD) <0.01% continued only ibrutinib 420 mg/day for 6 additional months (I arm), whereas the majority (n = 115) received up to 4 cycles of fludarabine/cyclophosphamide-obinutuzumab 1000 mg alongside the ibrutinib (I-FCG arm). Primary analysis at month 16 showed that 84 of 135 (62.2%) patients enrolled achieved CR with a BM MRD <0.01%. Here, we report follow-up at median 63 months. Peripheral blood (PB) MRD was assessed 6 monthly beyond the end of treatment using a highly sensitive (10-6) flow cytometry technique. In the I-FCG arm, the PB MRD <0.01% rate (low-level positive <0.01% or undetectable with limit of detection ≤10-4) in evaluable patients was still 92.5% (74/80) at month 40 and 80.6% (50/62) at month 64. No differences in the PB MRD status were apparent per to the IGHV mutational status. In the overall population, 4-year progression-free and overall survival rates were 95.5% and 96.2%, respectively. Twelve deaths occurred overall. Fourteen serious adverse events occurred beyond the end of treatment. Thus, our fixed-duration immunochemotherapy approach produced deep and sustained PB MRD responses, high survival rates, and low long-term toxicity. A randomized trial is needed to compare our immunochemotherapy approach with a chemotherapy-free strategy. This trial was registered at www.clinicaltrials.gov as #NCT02666898.
Collapse
Affiliation(s)
| | - Rémi Letestu
- Hematology Laboratory, Avicenne Hospital, Hôpitaux universitaires Paris Seine Saint-Denis (HUPSSD), Assistance Publique–Hôpitaux de Paris (AP-HP), Bobigny, France
| | | | - Lydia Campos
- Department of Hematology Biology, Le Centre Hospitalier Universitaire (CHU) Saint Etienne, Saint Etienne, France
| | | | | | | | - Valérie Rouille
- Department of Hematology, CHU Montpellier, Montpellier, France
| | - Béatrice Mahé
- Department of Hematology, Nantes University Hospital, Nantes, France
| | - Kamel Laribi
- Department of Hematology, CH Le Mans, Le Mans, France
| | | | - Emmanuelle Ferrant
- Department of Hematology and Oncology, CH Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard, Pierre-Benite, Lyon, France
| | - Olivier Tournilhac
- Department of Hematology and Cellular Therapy, CHU Hotel Dieu Hématologie, Clermont-Ferrand, France
| | - Alain Delmer
- Department of Hematology, CHU Reims, Reims, France
| | - Lysiane Molina
- Department of Hematology, CHU Grenoble, La Tronche, France
| | - Véronique Leblond
- Department of Hematology, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | | | | | | | - Anne Banos
- Department of Hematology, CH Côte Basque, Bayonne, France
| | | | - Guillaume Cartron
- Department of Hematology, CHU Montpellier, Montpellier, France
- Department of Hematology, Montpellier University Hospital, Montpelllier, France
| | | | - Loic Ysebaert
- Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | | | | | - Jean-Pierre Vilque
- Institut d'Hématologie de Basse-Normandie, Centre François Baclesse, Caen, France
| | | | | | - Stéphane Leprêtre
- Inserm U1245 and Department of Hematology, Centre Centre Henri-Becquerel and Normandie University UNIROUEN, Rouen, France
| | - Vincent Lévy
- Department of Clinical Research, Avicenne Hospital, HUPSSD, AP-HP, Bobigny, France
| | | | - Pierre Feugier
- Department of Hematology, Centre Hospitalier Régional Universitaire de Nancy, Hôpitaux de Brabois, Vandoeuvre-lès-Nancy, France
| | - French Innovative Leukemia Organization (FILO) CLL group
- Department of Hematology, Centre Léon Bérard, Lyon, France
- Hematology Laboratory, Avicenne Hospital, Hôpitaux universitaires Paris Seine Saint-Denis (HUPSSD), Assistance Publique–Hôpitaux de Paris (AP-HP), Bobigny, France
- Department of Hematology Biology, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
- Department of Hematology Biology, Le Centre Hospitalier Universitaire (CHU) Saint Etienne, Saint Etienne, France
- Department of Immunology, CHU Nice, Nice, France
- Department of Hematology, CHU Bordeaux, Pessac, France
- Department of Hematology, CHU Montpellier, Montpellier, France
- Department of Hematology, Nantes University Hospital, Nantes, France
- Department of Hematology, CH Le Mans, Le Mans, France
- Department of Hematology, CHD Vendée, La Roche-sur-Yon, France
- Department of Hematology and Oncology, CH Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard, Pierre-Benite, Lyon, France
- Department of Hematology and Cellular Therapy, CHU Hotel Dieu Hématologie, Clermont-Ferrand, France
- Department of Hematology, CHU Reims, Reims, France
- Department of Hematology, CHU Grenoble, La Tronche, France
- Department of Hematology, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
- Department of Hematology, CHU Poitiers, Poitiers, France
- Department of Hematology, CHU Rennes, Rennes, France
- Hematology Service, CH Annecy, Annecy, France
- Department of Hematology, CH Côte Basque, Bayonne, France
- Department of Hematology, CH Metz, Metz, France
- Department of Hematology, Montpellier University Hospital, Montpelllier, France
- Department of Hematology, Strasbourg University Hospital, Strasbourg, France
- Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
- Department of Hematology, CHU Tours, Hôpital Bretonneau, Tours, France
- Department of Hematology, CHU Angers, Angers, France
- Institut d'Hématologie de Basse-Normandie, Centre François Baclesse, Caen, France
- Department of Hematology, Institut Paoli Calmettes, Marseille, Marseille France
- Department of Hematology, Avicenne Hospital, HUPSSD, AP-HP, Bobigny, France
- Inserm U1245 and Department of Hematology, Centre Centre Henri-Becquerel and Normandie University UNIROUEN, Rouen, France
- Department of Clinical Research, Avicenne Hospital, HUPSSD, AP-HP, Bobigny, France
- Department of Hematology, Centre Hospitalier Régional Universitaire de Nancy, Hôpitaux de Brabois, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
7
|
Urso A, Cavazzini F, Ballardini MP, Gambara S, Consolo S, Rigolin GM, Cuneo A. First-Line Treatment of Older Patients with CLL: A New Approach in the Chemo-Free Era. Cancers (Basel) 2023; 15:3859. [PMID: 37568676 PMCID: PMC10417156 DOI: 10.3390/cancers15153859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Bruton tyrosine kinase inhibitors (BTKi) and the BCL2 inhibitor venetoclax, with or without the anti-CD20 monoclonal antibody Obinutuzumab, represent the preferred options for the first-line therapy of CLL because they are more effective and may improve quality of life. However, patient inclusion criteria are heterogeneous across trials designed for older patients, and the identification of CLL-specific parameters identifying unfit patients at risk of developing drug-specific adverse events is required to guide treatment choice. Due to inclusion/exclusion criteria in trials, higher discontinuation rates with BTKi were reported in real-world studies, and registry analyses provided useful information on factors predicting earlier discontinuation in a real-world setting. Though targeted agents were shown to be cost-effective treatments in high-income countries, the out-of-pocket expenses may limit accessibility to these drugs, and the overall expenditure for new drugs in CLL is projected to increase substantially, posing an issue for sustainability. This being said, the choice of a finite-duration treatment based on venetoclax-containing regimens or treatment until progression with BTKi is today possible in high-income countries, and the therapy choice drivers are represented by coexisting medical conditions rather than age, patient expectations, logistics, and sustainability.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Antonio Cuneo
- Hematology Unit, University of Ferrara, 44121 Ferrara, Italy (F.C.); (M.P.B.); (S.C.)
| |
Collapse
|
8
|
Maher N, Mouhssine S, Matti BF, Alwan AF, Gaidano G. Treatment Refractoriness in Chronic Lymphocytic Leukemia: Old and New Molecular Biomarkers. Int J Mol Sci 2023; 24:10374. [PMID: 37373521 PMCID: PMC10299596 DOI: 10.3390/ijms241210374] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common leukemia in adults. Despite its indolent clinical course, therapy refractoriness and disease progression still represent an unmet clinical need. Before the advent of pathway inhibitors, chemoimmunotherapy (CIT) was the commonest option for CLL treatment and is still widely used in areas with limited access to pathway inhibitors. Several biomarkers of refractoriness to CIT have been highlighted, including the unmutated status of immunoglobulin heavy chain variable genes and genetic lesions of TP53, BIRC3 and NOTCH1. In order to overcome resistance to CIT, targeted pathway inhibitors have become the standard of care for the treatment of CLL, with practice-changing results obtained through the inhibitors of Bruton tyrosine kinase (BTK) and BCL2. However, several acquired genetic lesions causing resistance to covalent and noncovalent BTK inhibitors have been reported, including point mutations of both BTK (e.g., C481S and L528W) and PLCG2 (e.g., R665W). Multiple mechanisms are involved in resistance to the BCL2 inhibitor venetoclax, including point mutations that impair drug binding, the upregulation of BCL2-related anti-apoptotic family members, and microenvironmental alterations. Recently, immune checkpoint inhibitors and CAR-T cells have been tested for CLL treatment, obtaining conflicting results. Potential refractoriness biomarkers to immunotherapy were identified, including abnormal levels of circulating IL-10 and IL-6 and the reduced presence of CD27+CD45RO- CD8+ T cells.
Collapse
Affiliation(s)
- Nawar Maher
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Samir Mouhssine
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| | - Bassam Francis Matti
- Department of Hematology and Bone Marrow Transplant, Hematology and Bone Marrow Transplant Center, Baghdad 00964, Iraq;
| | - Alaa Fadhil Alwan
- Department of Clinical Hematology, The National Center of Hematology, Mustansiriyah University, Baghdad 10015, Iraq;
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy; (N.M.); (S.M.)
| |
Collapse
|
9
|
Bourbon E, Chabane K, Mosnier I, Bouvard A, Thonier F, Ferrant E, Michallet AS, Poulain S, Hayette S, Sujobert P, Huet S. Next-CLL: A New Next-Generation Sequencing-Based Method for Assessment of IGHV Gene Mutational Status in Chronic Lymphoid Leukemia. J Mol Diagn 2023; 25:274-283. [PMID: 36773701 DOI: 10.1016/j.jmoldx.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/27/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Current guidelines for patients with chronic lymphocytic leukemia (CLL) recommend mutation status determination of the clonotypic IGHV gene before treatment initiation to guide the choice of first-line therapy. Currently, commercially available next-generation sequencing (NGS) solutions have technical constraints, as they necessitate at least a 2 × 300 bp sequencing, which restricts their use for routine practice. The cost of the commercial kits also represents an important drawback. We present a new method called Next-CLL, a ready-to-use strategy to evaluate IGHV gene mutation status using any NGS device (including 2 × 150 bp sequencers) in routine diagnostic laboratories. The performance of Next-CLL was validated on genomic DNA and cDNA obtained from 80 patients with CLL at diagnosis. Next-CLL identified a productive clone in 100% of cases, whereas PCR with Sanger sequencing led to a 12.5% failure rate. Next-CLL had 100% concordance with the reference technique for IGHV gene identification and allowed assessment of the IGHV mutation status from the leader sequence, following international guidelines. Comparing a large retrospective series of samples, analyzed by using Sanger sequencing (n = 773) or Next-CLL (n = 352), showed no bias in IGHV usage or mutational status, further validating our strategy in the real-life setting. Next-CLL represents a straightforward workflow for IGHV analysis in routine practice to assess clonal architecture and prognosis of patients with CLL.
Collapse
Affiliation(s)
- Estelle Bourbon
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Kaddour Chabane
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Isabelle Mosnier
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Anne Bouvard
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France
| | | | - Emmanuelle Ferrant
- Department of Clinical Hematology, Hospices Civils de Lyon, Pierre-Bénite, France
| | | | - Stéphanie Poulain
- Hematology Laboratory, Biology and Pathology Center, University Clinical Center of Lille, Lille, France; Team "Factors of persistence of leukemic cells," CANTHER Laboratory, UMR 9020 CNRS-U1277 INSERM, ONCOLILLE Cancer Institute, University of Lille, Lille, France
| | - Sandrine Hayette
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France; Team Lymphoma Immuno-Biology, Centre International de Recherche en Infectiologie U111 INSERM, Lyon, France
| | - Pierre Sujobert
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France; Team Lymphoma Immuno-Biology, Centre International de Recherche en Infectiologie U111 INSERM, Lyon, France; University Claude Bernard Lyon I, Lyon, France
| | - Sarah Huet
- Hematology Laboratory, Hospices Civils de Lyon, Pierre-Bénite, France; Team Lymphoma Immuno-Biology, Centre International de Recherche en Infectiologie U111 INSERM, Lyon, France; University Claude Bernard Lyon I, Lyon, France.
| |
Collapse
|
10
|
Markova EE, Nikitin EA, Dmitrieva EA, Mayorоva SY, Kochkareva YB, Naumova EV, Lugovskaya SA, Pochtar ME, Petrenko АА, Kislova МI, Biderman BV, Sudarikov АB, Obukhova ТN, Ptushkin VV. Obinutuzumab in combination with chlorambucil in first line treatment of elderly patients with chronic lymphocytic leukemia. RUSSIAN JOURNAL OF HEMATOLOGY AND TRANSFUSIOLOGY 2022. [DOI: 10.35754/0234-5730-2022-67-4-478-490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Introduction. Optimal therapy for elderly patients with chronic lymphocytic leukemia (CLL) is the subject of intensive research.Aim – to study the safety of obinutuzumab, as well as the selection of the optimal scheme of its use in patients with CLL, complicated by diabetes mellitus, renal insuffi ciency, cardiac comorbidity.Materials and methods. The study included primary patients with CLL having indications requiring therapy. The inclusion criteria were: Cumulative Illness Rating Scale, CIRS) (CIRS) > 6 and or glomerular fi ltration rate (GFR) < 70 mL/min, the lower limit of GFR was not restricted. The study focused on patients with diabetes mellitus, renal failure and signifi cant cardiac pathology. Patients with Richter’s syndrome, CNS involvement, HBs-antigen, and 17p deletion were not included. In the fi rst cycle obinutuzumab was administered at a dose of 100 or 25 mg on the fi rst day and 900 or 975 mg on the second day, then at a dose of 1000 mg on days 8 and 15. For all subsequent cycles, obinutuzumab was given at a dose of 1000 mg on day 1. The dosage of chlorambucil was 10 mg/m2 from days 1 to 7. Treatment cycles in totals of 6 were repeated every 28 days.Results. The study included 90 patients. Median age was 73.5 years, range – 60–89 years, there were 49 men (54 %) and 41 women (46 %). Twenty-four patients (27 %) had stage C, IGHV unmutated status was detected in 76 % of patients. The median creatinine clearance was 48.6 mL/min (25–110). The median CIRS score was 3 (range – 1–14). Thirty-one patients (34 %) had signifi cant cardiovascular comorbidity (previous myocardial infarction, coronary artery stenting or bypass, HF ≥ II NYHA, peripheral artery disease) as well as hemodynamically signifi cant valvular disease. Fifteen patients (17 %) had diabetes mellitus and 71 patients (79 %) had creatinine clearance < 70 ml/min. Infusion reactions to obinutuzumab grade ≥ II were reported in 29 patients (32 %). Hospitalization on the day of administration or the next day after the fi rst administration was required in 5 cases (5.5 %). Twenty-seven (30 %) patients could not complete 6 cycles. The largest number of patients (14 people, 15.5 %) stopped treatment after 1 course. The causes were the development of persistent cytopenia (n = 4), grade IV reaction to obinutuzumab (n = 3), patient’s refusal (n = 2), infectious complications (n = 2), severe tumor lysis syndrome (n = 1), acute pancreatitis (n = 1) and toxicodermia (n = 1). The leading cause of premature discontinuation on subsequent cycles was persistent neutropenia. Progression during treatment occurred in 3 patients only. Overall survival was signifi cantly predicted by CIRS (maximum discriminatory value of 3, p = 0.013) as well as GFR < 50 mL/min (p = 0.03). No other associations were identifi ed. At least 1 episode of grade III–IV neutropenia occurred in 41 % of patients. Grade IV neutropenia was associated with creatinine clearance < 60 mL/min (p = 0.05), baseline neutrophil level < 2 × 109/L (p = 0.0001), baseline monocyte level < 0.3 × 109/L (p = 0.007) and age > 70 years (p = 0.01). The effectiveness of treatment was evaluated in patients who completed at least 3 cycles of therapy. Complete remission was achieved in 26 patients (35 %), partial remission – in 41 (54 %), stabilization – in 4 (5 %), progression was noted in 3 (4 %). Sixteen patients (18 %) were not available to respond to the assessment. Minimal residual disease < 0.01 % in the bone marrow after completion of treatment was found in 17 patients (19 %), within 0.01–0.9 % – in 25 (28 %) patients. The median follow-up from the date of therapy initiation was 39.7 months (range – 0.6–72 months). The median relapse-free survival was not reached, and 2- and 3-year survival rates were 81 and 62 %, respectively. Poor relapse-free survival signifi cantly correlated with unmutated IGHV genes (HR = 2.4, 95 % CI: 1.12–5.0, p = 0.02) and partial response as opposed to complete response (HR = 3.35; 95 % CI: 1.45–7.7, p = 0.03).Conclusion. The results of our study have practical implications as obinutuzumab is actively integrated into modern treatment regimens. Infusion reactions pose a high risk of complications in elderly patients. The dose of obinutuzumab on day 1 of administration in elderly patients, should not exceed 25 mg. The G-Clb regimen may not be optimal in patients over 75 years of age due to the unpredictable risk of complications. In patients at high risk of neutropenia, it may be appropriate to consider primary prophylaxis. ClbG is an effective regimen that resulted in high rate of MRD-negative responses and prolonged relapse-free survival.
Collapse
Affiliation(s)
- E. E. Markova
- Botkin City Clinical Hospital of the Moscow Health Department
| | - E. A. Nikitin
- Botkin City Clinical Hospital of the Moscow Health Department; Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - E. A. Dmitrieva
- Botkin City Clinical Hospital of the Moscow Health Department; Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - S. Yu. Mayorоva
- Botkin City Clinical Hospital of the Moscow Health Department
| | | | - E. V. Naumova
- Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - S. A. Lugovskaya
- Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - M. E. Pochtar
- Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - А. А. Petrenko
- Botkin City Clinical Hospital of the Moscow Health Department; Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| | - М. I. Kislova
- Botkin City Clinical Hospital of the Moscow Health Department
| | | | | | | | - V. V. Ptushkin
- Botkin City Clinical Hospital of the Moscow Health Department; Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation
| |
Collapse
|
11
|
Ahn IE, Brown JR. Selecting initial therapy in CLL. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:323-328. [PMID: 36485152 PMCID: PMC9820765 DOI: 10.1182/hematology.2022000343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Targeted therapy is a powerful treatment option in chronic lymphocytic leukemia (CLL) that has outperformed conventional chemoimmunotherapy in most clinical settings. Except for selected young, fit patients with a mutated immunoglobulin heavy chain variable region gene, most patients benefit from targeted therapy with either a continuous BTK inhibitor or 1-year fixed-duration venetoclax-obinutuzumab as first-line treatment of CLL. Treatment selection is driven by patient-, treatment-, and disease-related factors, encompassing patient preference, concomitant medications, comorbidities, safety profile of the regimen, and TP53 aberration. Clinical trials are actively investigating the simultaneous inhibition of Bruton's tyrosine kinase (BTK) and B-cell lymphoma 2 (BCL-2) proteins with or without a CD20 monoclonal antibody, which can achieve deep response in most patients (52%-89% undetectable minimal residual disease in bone marrow).
Collapse
|
12
|
Leslie LA, Gangan N, Tan H, Huang Q. Clinical and economic burden of first-line chemoimmunotherapy by risk status in chronic lymphocytic leukemia. Curr Med Res Opin 2022; 38:2149-2161. [PMID: 36205521 DOI: 10.1080/03007995.2022.2133468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To evaluate the trend in cytogenetic/molecular testing rate in chronic lymphocytic leukemia (CLL) and assess the clinical and economic burden of first-line (1 L) treatment with chemoimmunotherapy (CIT) by risk status. METHODS This retrospective cohort study identified patients with CLL from a U.S. managed care population. Medical records were obtained for eligible patients who initiated 1 L CIT between 1/1/2007 and 7/31/2019 and underwent prognostic testing to classify them as high risk (del(17p), TP53 mutation, del(11q), unmutated IGHV or complex karyotype) or as non-high risk by FISH only (non-del(17p) and non-del(11q)). Study outcomes included testing rate, time to next treatment (TTNT) or death, time to treatment failure (defined as time to change of therapy, non-chemotherapy intervention, hospice care or death), and total plan paid costs (medical + pharmacy) per patient per month (PPPM) in the 1 L period. Cox proportional hazard models and generalized linear models were used to calculate adjusted hazard ratio or rate ratio. RESULTS Among the 1,808 patients with CLL, 612 were FISH or IGHV tested and the rate of testing increased from 30% to 44% from 2007-2019. High-risk patients (n = 119) had 65% higher risk of next treatment or death (median time: 2.4 vs 3.7 years), 65% higher risk of treatment failure (median time: 3.0 vs 4.9 years), and 33% higher costs ($12,194 vs $9,055, p = 0.027) during 1 L treatment than non-high risk patients (n = 134). CONCLUSIONS High-risk CLL patients treated with 1 L chemoimmunotherapy have poorer clinical and economic outcomes compared to non-high risk patients. Assessment of genetic risk remains suboptimal.
Collapse
Affiliation(s)
| | | | | | - Qing Huang
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
| |
Collapse
|
13
|
Visentin A, Mauro FR, Catania G, Fresa A, Vitale C, Sanna A, Mattiello V, Cibien F, Sportoletti P, Gentile M, Rigolin GM, Quaglia FM, Murru R, Gozzetti A, Molica S, Marchetti M, Pravato S, Angotzi F, Cellini A, Scarfò L, Reda G, Coscia M, Laurenti L, Ghia P, Foà R, Cuneo A, Trentin L. Obinutuzumab plus chlorambucil versus ibrutinib in previously untreated chronic lymphocytic leukemia patients without TP53 disruptions: A real-life CLL campus study. Front Oncol 2022; 12:1033413. [PMID: 36479077 PMCID: PMC9719965 DOI: 10.3389/fonc.2022.1033413] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
One of the main issues in the treatment of patients with chronic lymphocytic leukemia (CLL) deals with the choice between continuous or fixed-duration therapy. Continuous ibrutinib (IB), the first-in-class BTK inhibitor, and obinutuzumab-chlorambucil (G-CHL) are commonly used therapies for elderly and/or comorbid patients. No head-to-head comparison has been carried out. Within the Italian campus CLL network, we performed a retrospective study on CLL patients without TP53 disruption treated with IB or G-CHL as first-line therapy. Patients in the G-CHL arm had a higher CIRS score and the worst renal function. The overall response rates between the G-CHL and IB arms were similar, but more complete remissions (CRs) were achieved with G-CHL (p = 0.0029). After a median follow-up of 30 months, the progression-free survival (PFS, p = 0.0061) and time to next treatment (TTNT, p = 0.0043), but not overall survival (OS, p = 0.6642), were better with IB than with G-CHL. Similar results were found after propensity score matching and multivariate analysis. While PFS and TTNT were longer with IB than with G-CHL in IGHV unmutated patients (p = 0.0190 and 0.0137), they were superimposable for IGHV mutated patients (p = 0.1900 and 0.1380). In the G-CHL arm, the depth of response (79% vs. 68% vs. 38% for CR, PR and SD/PD; p < 0.0001) and measurable residual disease (MRD) influenced PFS (78% vs. 53% for undetectable MRD vs. detectable MRD, p = 0.0203). Hematological toxicities were common in the G-CHL arm, while IB was associated with higher costs. Although continuous IB provides better disease control in CLL, IGHV mutated patients and those achieving an undetectable MRD show a marked clinical and economic benefit from a fixed-duration obinutuzumab-based treatment.
Collapse
Affiliation(s)
- Andrea Visentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Francesca Romana Mauro
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Rome, Italy
| | - Gioachino Catania
- Division of Hematology, Hospital Saints (A. O. SS) Antonio e Biagio and Cesare Arrigo, Alessandria, Italy
| | - Alberto Fresa
- Hematology Institute, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Candida Vitale
- Department of Molecular Biotechnology and health Sciences, University of Torino and Division of Hematology, University Hospital (A.O.U.) Città della Salute e della Scienza di Torino, Torino, Italy
| | | | - Veronica Mattiello
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore, University of Milan, Milan, Italy
| | | | - Paolo Sportoletti
- Hematology and Clinical Immunology Unit, University of Perugia, Perugia, Italy
| | | | - Gian Matteo Rigolin
- Hematology Section, Department of Medical Sciences, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - Francesca Maria Quaglia
- Department of Medicine, Section of Hematology, University of Verona and Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Roberta Murru
- Hematology and Stem Cell Transplantation Unit, Ospedale A. Businco ARNAS “G. Brotzu”, Cagliari, Italy
| | | | - Stefano Molica
- Department Hematology-Oncology, Azienda Ospedaliera Pugliese-Ciaccio, Catanzaro, Italy
| | - Monia Marchetti
- Division of Hematology, Hospital Saints (A. O. SS) Antonio e Biagio and Cesare Arrigo, Alessandria, Italy
| | - Stefano Pravato
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Francesco Angotzi
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Alessandro Cellini
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
| | - Lydia Scarfò
- Strategic Program on CLL, University Health and Science “San Raffaele”, Milan, Italy
| | - Gianluigi Reda
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore, University of Milan, Milan, Italy
| | - Marta Coscia
- Department of Molecular Biotechnology and health Sciences, University of Torino and Division of Hematology, University Hospital (A.O.U.) Città della Salute e della Scienza di Torino, Torino, Italy
| | - Luca Laurenti
- Hematology Institute, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Paolo Ghia
- Strategic Program on CLL, University Health and Science “San Raffaele”, Milan, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, “Sapienza” University, Rome, Italy
| | - Antonio Cuneo
- Hematology Section, Department of Medical Sciences, Azienda Ospedaliera-Universitaria, Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - Livio Trentin
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padua, Padova, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| |
Collapse
|
14
|
Lovell AR, Jammal N, Bose P. Selecting the optimal BTK inhibitor therapy in CLL: rationale and practical considerations. Ther Adv Hematol 2022; 13:20406207221116577. [PMID: 35966045 PMCID: PMC9373150 DOI: 10.1177/20406207221116577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) inhibitors have dramatically changed the treatment of newly diagnosed and relapsed/refractory chronic lymphocytic leukemia (CLL). Ibrutinib, acalabrutinib, and zanubrutinib are Food and Drug Administration (FDA)-approved BTK inhibitors that have all demonstrated progression-free survival (PFS) benefit compared with chemoimmunotherapy. The efficacy of these agents compared to one another is under study; however, current data suggest they provide similar efficacy. Selectivity for BTK confers different adverse effect profiles, and longer follow-up and real-world use have characterized side effects over time. The choice of BTK inhibitor is largely patient-specific, and this review aims to highlight the differences among the agents and guide the choice of BTK inhibitor in clinical practice.
Collapse
Affiliation(s)
- Alexandra R. Lovell
- Division of Pharmacy, The University of Texas
MD Anderson Cancer Center, Houston, TX, USA
| | - Nadya Jammal
- Division of Pharmacy, The University of Texas
MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| |
Collapse
|
15
|
Genetic Testing at Diagnosis Has Prognostic Value in Patients with Chronic Lymphocytic Leukemia including at Early Stages. Diagnostics (Basel) 2022; 12:diagnostics12081802. [PMID: 35892513 PMCID: PMC9394282 DOI: 10.3390/diagnostics12081802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) has a variable clinical evolution, with some patients living treatment-free for decades while others require therapy shortly after diagnosis. In a consecutive series of 217 CLL patients, molecular biomarkers with prognostic value (IGHV status, TP53 mutations, and cytogenetics), whose analysis is recommended prior to treatment start, were studied at diagnosis. Multivariate analyses identified prognostic variables for overall survival (OS) and time to first treatment (TTFT) and validated the CLL-IPI and IPS-E variables for all or early-stage patients (Rai 0–2/Binet A), respectively. Unmutated IGHV was associated with shorter OS and TTFT, even for early-stage patients. Lymphocyte count was not statistically significant for TTFT of early-stage patients in multivariate analysis. Our results validate the prognostic value of IGHV mutational status at diagnosis for OS and TTFT, including for early stages. Our findings suggest a role for molecular and mutational analysis at diagnosis in future prospective studies.
Collapse
|
16
|
Shanafelt TD, Wang XV, Hanson CA, Paietta EM, O'Brien S, Barrientos J, Jelinek DF, Braggio E, Leis JF, Zhang CC, Coutre SE, Barr PM, Cashen AF, Mato AR, Singh AK, Mullane MP, Little RF, Erba H, Stone RM, Litzow M, Tallman M, Kay NE. Long-term outcomes for ibrutinib-rituximab and chemoimmunotherapy in CLL: updated results of the E1912 trial. Blood 2022; 140:112-120. [PMID: 35427411 PMCID: PMC9283968 DOI: 10.1182/blood.2021014960] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/25/2022] [Indexed: 01/14/2023] Open
Abstract
Herein, we present the long-term follow-up of the randomized E1912 trial comparing the long-term efficacy of ibrutinib-rituximab (IR) therapy to fludarabine, cyclophosphamide, and rituximab (FCR) and describe the tolerability of continuous ibrutinib. The E1912 trial enrolled 529 treatment-naïve patients aged ≤70 years with chronic lymphocytic leukemia (CLL). Patients were randomly assigned (2:1 ratio) to receive IR or 6 cycles of FCR. With a median follow-up of 5.8 years, median progression-free survival (PFS) is superior for IR (hazard ratio [HR], 0.37; P < .001). IR improved PFS relative to FCR in patients with both immunoglobulin heavy chain variable region (IGHV) gene mutated CLL (HR: 0.27; P < .001) and IGHV unmutated CLL (HR: 0.27; P < .001). Among the 354 patients randomized to IR, 214 (60.5%) currently remain on ibrutinib. Among the 138 IR-treated patients who discontinued treatment, 37 (10.5% of patients who started IR) discontinued therapy due to disease progression or death, 77 (21.9% of patients who started IR) discontinued therapy for adverse events (AEs)/complications, and 24 (6.8% of patients who started IR) withdrew for other reasons. Progression was uncommon among patients able to remain on ibrutinib. The median time from ibrutinib discontinuation to disease progression or death among those who discontinued treatment for a reason other than progression was 25 months. Sustained improvement in overall survival (OS) was observed for patients in the IR arm (HR, 0.47; P = .018). In conclusion, IR therapy offers superior PFS relative to FCR in patients with IGHV mutated or unmutated CLL, as well as superior OS. Continuous ibrutinib therapy is tolerated beyond 5 years in the majority of CLL patients. This trial was registered at www.clinicaltrials.gov as #NCT02048813.
Collapse
Affiliation(s)
| | | | | | | | - Susan O'Brien
- University of California Irvine Medical Center, Irvine, CA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kaufman M, Yan XJ, Li W, Ghia EM, Langerak AW, Rassenti LZ, Belessi C, Kay NE, Davi F, Byrd JC, Pospisilova S, Brown JR, Catherwood M, Davis Z, Oscier D, Montillo M, Trentin L, Rosenquist R, Ghia P, Barrientos JC, Kolitz JE, Allen SL, Rai KR, Stamatopoulos K, Kipps TJ, Neuberg D, Chiorazzi N. Impact of the Types and Relative Quantities of IGHV Gene Mutations in Predicting Prognosis of Patients With Chronic Lymphocytic Leukemia. Front Oncol 2022; 12:897280. [PMID: 35903706 PMCID: PMC9315922 DOI: 10.3389/fonc.2022.897280] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with CLL with mutated IGHV genes (M-CLL) have better outcomes than patients with unmutated IGHVs (U-CLL). Since U-CLL usually express immunoglobulins (IGs) that are more autoreactive and more effectively transduce signals to leukemic B cells, B-cell receptor (BCR) signaling is likely at the heart of the worse outcomes of CLL cases without/few IGHV mutations. A corollary of this conclusion is that M-CLL follow less aggressive clinical courses because somatic IGHV mutations have altered BCR structures and no longer bind stimulatory (auto)antigens and so cannot deliver trophic signals to leukemic B cells. However, the latter assumption has not been confirmed in a large patient cohort. We tried to address the latter by measuring the relative numbers of replacement (R) mutations that lead to non-conservative amino acid changes (Rnc) to the combined numbers of conservative (Rc) and silent (S) amino acid R mutations that likely do not or cannot change amino acids, "(S+Rc) to Rnc IGHV mutation ratio". When comparing time-to-first-treatment (TTFT) of patients with (S+Rc)/Rnc ≤ 1 and >1, TTFTs were similar, even after matching groups for equal numbers of samples and identical numbers of mutations per sample. Thus, BCR structural change might not be the main reason for better outcomes for M-CLL. Since the total number of IGHV mutations associated better with longer TTFT, better clinical courses appear due to the biologic state of a B cell having undergone many stimulatory events leading to IGHV mutations. Analyses of larger patient cohorts will be needed to definitively answer this question.
Collapse
Affiliation(s)
- Matthew Kaufman
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Xiao-Jie Yan
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Wentian Li
- The Robert S. Boas Center for Genomics & Human Genetics, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Emanuela M. Ghia
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Anton W. Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Laura Z. Rassenti
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | | | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Frederic Davi
- Department of Biological Hematology, Hôpital Pitié-Salpêtrière (AP-HP), Sorbonne Université, Paris, France
| | - John C. Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sarka Pospisilova
- Department of Internal Medicine - Hematology and Oncology and Department of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Jennifer R. Brown
- Chronic Lymphocytic Leukemia Center, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Mark Catherwood
- Clinical Hematology, Belfast City Hospital, Belfast, Ireland
| | - Zadie Davis
- Department of Molecular Pathology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - David Oscier
- Department of Hematology, Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Marco Montillo
- Department of Hematology & Oncology, Niguarda Cancer Center, Niguarda Hospital, Milan, Italy
| | - Livio Trentin
- Hematology Unit, Department of Medicine-(DIMED), University of Padua University Hospital, Padua, Italy
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Ghia
- Division of Experimental Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Jacqueline C. Barrientos
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Jonathan E. Kolitz
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Steven L. Allen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Kanti R. Rai
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| | - Kostas Stamatopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Thomas J. Kipps
- Center for Novel Therapeutics, Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Departments of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Uniondale, NY, United States
- Northwell Health Cancer Institute, Lake Success, NY, United States
| |
Collapse
|
18
|
Langlois de Septenville A, Boudjoghra M, Bravetti C, Armand M, Salson M, Giraud M, Davi F. Immunoglobulin Gene Mutational Status Assessment by Next Generation Sequencing in Chronic Lymphocytic Leukemia. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2453:153-167. [PMID: 35622326 DOI: 10.1007/978-1-0716-2115-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
B cell receptor (BcR) immunoglobulins (IG) display a tremendous diversity due to complex DNA rearrangements, the V(D)J recombination, further enhanced by the somatic hypermutation process. In chronic lymphocytic leukemia (CLL), the mutational load of the clonal BcR IG expressed by the leukemic cells constitutes an important prognostic and predictive biomarker. Here, we provide a reliable methodology capable of determining the mutational status of IG genes in CLL using high-throughput sequencing, starting from leukemic cell DNA or RNA.
Collapse
Affiliation(s)
| | - Myriam Boudjoghra
- AP-HP, Pitié-Salpêtrière Hospital, Laboratory of Hematology, Paris, France
| | - Clotilde Bravetti
- AP-HP, Pitié-Salpêtrière Hospital, Laboratory of Hematology, Paris, France
- Sorbonne Université, Paris, France
| | - Marine Armand
- AP-HP, Pitié-Salpêtrière Hospital, Laboratory of Hematology, Paris, France
- Sorbonne Université, Paris, France
| | - Mikaël Salson
- Université de Lille, CNRS, UMR 9189-CRIStAL, Inria, Lille, France
| | - Mathieu Giraud
- Université de Lille, CNRS, UMR 9189-CRIStAL, Inria, Lille, France
| | - Frederic Davi
- AP-HP, Pitié-Salpêtrière Hospital, Laboratory of Hematology, Paris, France.
- Sorbonne Université, Paris, France.
| |
Collapse
|
19
|
Immunoglobulin gene sequence analysis in chronic lymphocytic leukemia: the 2022 update of the recommendations by ERIC, the European Research Initiative on CLL. Leukemia 2022; 36:1961-1968. [PMID: 35614318 PMCID: PMC9343247 DOI: 10.1038/s41375-022-01604-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/04/2022] [Accepted: 05/12/2022] [Indexed: 11/18/2022]
Abstract
The somatic hypermutation (SHM) status of the clonotypic immunoglobulin heavy variable (IGHV) gene is a critical biomarker for assessing the prognosis of patients with chronic lymphocytic leukemia (CLL). Importantly, independent studies have documented that IGHV SHM status is also a predictor of responses to therapy, including both chemoimmunotherapy (CIT) and novel, targeted agents. Moreover, immunogenetic analysis in CLL has revealed that different patients may express (quasi)identical, stereotyped B cell receptor immunoglobulin (BcR IG) and are classified into subsets based on this common feature. Patients in certain stereotyped subsets display consistent biology, clinical presentation, and outcome that are distinct from other patients, even with concordant IGHV gene SHM status. All of the above highlights the relevance of immunogenetic analysis in CLL, which is considered a cornerstone for accurate risk stratification and clinical decision making. Recommendations for robust immunogenetic analysis exist thanks to dedicated efforts by ERIC, the European Research Initiative on CLL, covering all test phases, from the pre-analytical and analytical to the post-analytical, pertaining to the analysis, interpretation, and reporting of the findings. That said, these recommendations apply to Sanger sequencing, which is increasingly being superseded by next generation sequencing (NGS), further underscoring the need for an update. Here, we present an overview of the clinical utility of immunogenetics in CLL and update our analytical recommendations with the aim to assist in the refined management of patients with CLL.
Collapse
|
20
|
Liu L, Cheng X, Yang H, Lian S, Jiang Y, Liang J, Chen X, Mo S, Shi Y, Zhao S, Li J, Jiang R, Yang DH, Wu Y. BCL-2 expression promotes immunosuppression in chronic lymphocytic leukemia by enhancing regulatory T cell differentiation and cytotoxic T cell exhaustion. Mol Cancer 2022; 21:59. [PMID: 35193595 PMCID: PMC8862474 DOI: 10.1186/s12943-022-01516-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/21/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) results in increased susceptibility to infections. T cell dysfunction is not associated with CLL in all patients; therefore, it is important to identify CLL patients with T cell defects. The role of B-cell lymphoma-2 (BCL-2) in CLL has been explored; however, few studies have examined its role in T cells in CLL patients. Herein, we have investigated the regulatory role of BCL-2 in T cells in the CLL tumor microenvironment. METHODS The expression of BCL-2 in T cells was evaluated using flow cytometry. The regulatory roles of BCL-2 were investigated using single-cell RNA sequencing (scRNA-seq) and verified using multi-parameter flow cytometry on CD4 and CD8 T cells. The clinical features of BCL-2 expression in T cells in CLL were also explored. RESULTS We found a significant increase in BCL-2 expression in the T cells of CLL patients (n = 266). Single cell RNA sequencing (scRNA-seq) indicated that BCL-2+CD4+ T cells had the gene signature of increased regulatory T cells (Treg); BCL-2+CD8+ T cells showed the gene signature of exhausted cytotoxic T lymphocytes (CTL); and increased expression of BCL-2 was associated with T cell activation and cellular adhesion. The results from scRNA-seq were verified in peripheral T cells from 70 patients with CLL, wherein BCL-2+CD4+ T cells were enriched with Tregs and had higher expression of interleukin-10 and transforming growth factor-β than BCL-2-CD4+ T cells. BCL-2 expression in CD8+T cells was associated with exhausted cells (PD-1+Tim-3+) and weak expression of granzyme B and perforin. T cell-associated cytokine profiling revealed a negative association between BCL-2+ T cells and T cell activation. Decreased frequencies and recovery functions of BCL-2+T cells were observed in CLL patients in complete remission after treatment with venetoclax. CONCLUSION BCL-2 expression in the T cells of CLL patients is associated with immunosuppression via promotion of Treg abundance and CTL exhaustion.
Collapse
Affiliation(s)
- Lu Liu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Xianfeng Cheng
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Hui Yang
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Senlin Lian
- Jiangsu Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China.,State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuegen Jiang
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Jinhua Liang
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Xiao Chen
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Suo Mo
- Department of Clinical laboratory, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yu Shi
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Sishu Zhao
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Jianyong Li
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China
| | - Runqiu Jiang
- Jiangsu Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, China. .,State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China. .,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Dong-Hua Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Yujie Wu
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China. .,Key Laboratory of Hematology of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
21
|
Forconi F, Lanham SA, Chiodin G. Biological and Clinical Insight from Analysis of the Tumor B-Cell Receptor Structure and Function in Chronic Lymphocytic Leukemia. Cancers (Basel) 2022; 14:663. [PMID: 35158929 PMCID: PMC8833472 DOI: 10.3390/cancers14030663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
The B-cell receptor (BCR) is essential to the behavior of the majority of normal and neoplastic mature B cells. The identification in 1999 of the two major CLL subsets expressing unmutated immunoglobulin (Ig) variable region genes (U-IGHV, U-CLL) of pre-germinal center origin and poor prognosis, and mutated IGHV (M-CLL) of post-germinal center origin and good prognosis, ignited intensive investigations on structure and function of the tumor BCR. These investigations have provided fundamental insight into CLL biology and eventually the mechanistic rationale for the development of successful therapies targeting BCR signaling. U-CLL and M-CLL are characterized by variable low surface IgM (sIgM) expression and signaling capacity. Variability of sIgM can in part be explained by chronic engagement with (auto)antigen at tissue sites. However, other environmental elements, genetic changes, and epigenetic signatures also contribute to the sIgM variability. The variable levels have consequences on the behavior of CLL, which is in a state of anergy with an indolent clinical course when sIgM expression is low, or pushed towards proliferation and a more aggressive clinical course when sIgM expression is high. Efficacy of therapies that target BTK may also be affected by the variable sIgM levels and signaling and, in part, explain the development of resistance.
Collapse
Affiliation(s)
- Francesco Forconi
- School of Cancer Sciences, Cancer Research UK and NIHR Experimental Cancer Medicine Centres, University of Southampton, Southampton SO16 6YD, UK; (S.A.L.); (G.C.)
- Department of Haematology, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
| | - Stuart A. Lanham
- School of Cancer Sciences, Cancer Research UK and NIHR Experimental Cancer Medicine Centres, University of Southampton, Southampton SO16 6YD, UK; (S.A.L.); (G.C.)
| | - Giorgia Chiodin
- School of Cancer Sciences, Cancer Research UK and NIHR Experimental Cancer Medicine Centres, University of Southampton, Southampton SO16 6YD, UK; (S.A.L.); (G.C.)
| |
Collapse
|
22
|
Pepper AGS, Zucchetto A, Norris K, Tissino E, Polesel J, Soe Z, Allsup D, Hockaday A, Ow PL, Hillmen P, Rawstron A, Catovsky D, Bulian P, Bomben R, Baird DM, Fegan CD, Gattei V, Pepper C. Combined analysis of IGHV mutations, telomere length and CD49d identifies long-term progression-free survivors in TP53 wild-type CLL treated with FCR-based therapies. Leukemia 2022; 36:271-274. [PMID: 34148055 PMCID: PMC8727296 DOI: 10.1038/s41375-021-01322-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/24/2022]
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/analysis
- Cyclophosphamide/administration & dosage
- Follow-Up Studies
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/genetics
- Integrin alpha4/genetics
- Integrin alpha4/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mutation
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Prognosis
- Rituximab/administration & dosage
- Survival Rate
- Telomere Homeostasis
- Tumor Suppressor Protein p53/genetics
- Vidarabine/administration & dosage
- Vidarabine/analogs & derivatives
Collapse
Affiliation(s)
- Andrea G S Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Kevin Norris
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Erika Tissino
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Zarni Soe
- Leeds Teaching Hospital Trust, Leeds, United Kingdom
| | - David Allsup
- Hull York Medical School, University of Hull, Hull, United Kingdom
| | - Anna Hockaday
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Pei Loo Ow
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Peter Hillmen
- Section of Experimental Haematology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Andrew Rawstron
- Section of Experimental Haematology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | | | - Pietro Bulian
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Duncan M Baird
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Christopher D Fegan
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom.
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom.
| |
Collapse
|
23
|
Are we finally getting personal? Moving towards a personalized approach in chronic lymphocytic leukemia. Semin Cancer Biol 2022; 84:329-338. [DOI: 10.1016/j.semcancer.2022.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 12/21/2022]
|
24
|
Gamaleldin M, Moussa M, Eldin Imbaby S. Role of interleukin-10 (1082G/A) and splicing factor 3B subunit 1 (2098A/G) gene polymorphisms in chronic lymphocytic leukemia. JOURNAL OF APPLIED HEMATOLOGY 2022. [DOI: 10.4103/joah.joah_93_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
van der Straten L, Hengeveld PJ, Kater AP, Langerak AW, Levin MD. Treatment Approaches to Chronic Lymphocytic Leukemia With High-Risk Molecular Features. Front Oncol 2021; 11:780085. [PMID: 34956898 PMCID: PMC8695615 DOI: 10.3389/fonc.2021.780085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/23/2021] [Indexed: 12/30/2022] Open
Abstract
The clinical course of chronic lymphocytic leukemia (CLL) is highly variable. Over the past decades, several cytogenetic, immunogenetic and molecular features have emerged that identify patients suffering from CLL with high-risk molecular features. These biomarkers can clearly aid prognostication, but may also be capable of predicting the efficacy of various treatment strategies in subgroups of patients. In this narrative review, we discuss treatment approaches to CLL with high-risk molecular features. Specifically, we review and provide a comprehensive overview of clinical trials evaluating the efficacy of chemotherapy, chemoimmunotherapy and novel agent-based treatments in CLL patients with TP53 aberrations, deletion of the long arm of chromosome 11, complex karyotype, unmutated IGHV, B cell receptor stereotypy, and mutations in NOTCH1 or BIRC3. Furthermore, we discuss future pharmaceutical and immunotherapeutic perspectives for CLL with high-risk molecular features, focusing on agents currently under investigation in clinical trials.
Collapse
Affiliation(s)
- Lina van der Straten
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, Netherlands.,Laboratory Medical Immunology, Department of Immunology, Erasmus MC, Rotterdam, Netherlands.,Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, Netherlands
| | - Paul J Hengeveld
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, Netherlands.,Laboratory Medical Immunology, Department of Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Arnon P Kater
- Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| | - Anton W Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC, Rotterdam, Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, Netherlands
| |
Collapse
|
26
|
Jain N, Thompson P, Burger J, Ferrajoli A, Takahashi K, Estrov Z, Borthakur G, Bose P, Kadia T, Pemmaraju N, Sasaki K, Konopleva M, Jabbour E, Garg N, Wang X, Kanagal-Shamanna R, Patel K, Wang W, Jorgensen J, Wang S, Lopez W, Ayala A, Plunkett W, Gandhi V, Kantarjian H, O'Brien S, Keating M, Wierda WG. Ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) regimen for chronic lymphocytic leukemia (CLL) with mutated IGHV and without TP53 aberrations. Leukemia 2021; 35:3421-3429. [PMID: 34007049 DOI: 10.1038/s41375-021-01280-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 04/20/2021] [Accepted: 04/30/2021] [Indexed: 12/16/2022]
Abstract
Chemoimmunotherapy with combined fludarabine, cyclophosphamide and rituximab (FCR) has been an effective treatment for patients with chronic lymphocytic leukemia (CLL). We initiated a phase II trial for previously untreated patients with CLL with mutated IGHV and absence of del(17p)/TP53 mutation. Patients received ibrutinib, fludarabine, cyclophosphamide, and obinutuzumab (iFCG) for three cycles. Patients who achieved complete remission (CR)/CR with incomplete count recvoery (CRi) with marrow undetectable measurable residual disease (U-MRD) received additional nine cycles of ibrutinib with three cycles of obinutuzumab; all others received nine additional cycles of ibrutinib and obinutuzumab. Patients in marrow U-MRD remission after cycle 12 discontinued all treatment, including ibrutinib. Forty-five patients were treated. The median follow-up is 41.3 months. Among the total 45 treated patients, after three cycles, 38% achieved CR/CRi and 87% achieved marrow U-MRD. After cycle 12, the corresponding numbers were 67% and 91%, respectively. Overall, 44/45 (98%) patients achieved marrow U-MRD as best response. No patient had CLL progression. The 3-year progression-free survival (PFS) and overall survival (OS) were 98% and 98%, respectively. Per trial design, all patients who completed cycle 12 discontinued ibrutinib, providing for a time-limited therapy. Grade 3-4 neutropenia and thrombocytopenia occurred in 58% and 40% patients, respectively. The iFCG regimen with only 3 cycles of chemotherapy is an effective, time-limited regimen for patients with CLL with mutated IGHV and without del(17p)/TP53 mutation.
Collapse
MESH Headings
- Adenine/administration & dosage
- Adenine/analogs & derivatives
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Cyclophosphamide/administration & dosage
- Female
- Follow-Up Studies
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Mutation
- Piperidines/administration & dosage
- Prognosis
- Survival Rate
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Vidarabine/administration & dosage
- Vidarabine/analogs & derivatives
Collapse
Affiliation(s)
- Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip Thompson
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jan Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zeev Estrov
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naveen Garg
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sa Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wanda Lopez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Ayala
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Varsha Gandhi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Susan O'Brien
- Chao Family Comprehensive Cancer Center, University of California Irvine Medical Center, Orange, CA, USA
| | - Michael Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
27
|
González-Rincón J, Garcia-Vela JA, Gómez S, Fernández-Cuevas B, Nova-Gurumeta S, Pérez-Sanz N, Alcoceba M, González M, Anguita E, López-Jiménez J, González-Barca E, Yáñez L, Pérez-Persona E, de la Serna J, Fernández-Zarzoso M, Deben G, Peñalver FJ, Fernández MC, de Oteyza JP, Andreu MÁ, Ruíz-Guinaldo MÁ, Paz-Arias R, García-Malo MD, Recasens V, Collado R, Córdoba R, Navarro-Matilla B, Sánchez-Beato M, García-Marco JA. Genomic mutation profile in progressive chronic lymphocytic leukemia patients prior to first-line chemoimmunotherapy with FCR and rituximab maintenance (REM). PLoS One 2021; 16:e0257353. [PMID: 34506616 PMCID: PMC8432772 DOI: 10.1371/journal.pone.0257353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic Lymphocytic Leukemia (CLL) is the most prevalent leukemia in Western countries and is notable for its variable clinical course. This variability is partly reflected by the mutational status of IGHV genes. Many CLL samples have been studied in recent years by next-generation sequencing. These studies have identified recurrent somatic mutations in NOTCH1, SF3B1, ATM, TP53, BIRC3 and others genes that play roles in cell cycle, DNA repair, RNA metabolism and splicing. In this study, we have taken a deep-targeted massive sequencing approach to analyze the impact of mutations in the most frequently mutated genes in patients with CLL enrolled in the REM (rituximab en mantenimiento) clinical trial. The mutational status of our patients with CLL, except for the TP53 gene, does not seem to affect the good results obtained with maintenance therapy with rituximab after front-line FCR treatment.
Collapse
Affiliation(s)
- Julia González-Rincón
- Lymphoma Research Group, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDPHISA), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Sagrario Gómez
- Lymphoma Research Group, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDPHISA), Madrid, Spain
| | - Belén Fernández-Cuevas
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain
| | - Sara Nova-Gurumeta
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain
| | - Nuria Pérez-Sanz
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain
| | - Miguel Alcoceba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematology, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Marcos González
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hematology, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Eduardo Anguita
- Hematology, Hospital Clínico San Carlos, IdISSC, UCM, Madrid, Spain
| | | | - Eva González-Barca
- Hematology, Institut Català d’Oncologia, IDIBELL, L’Hospitalet de LLobregat, Spain
| | - Lucrecia Yáñez
- Hematology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | - Rosa Collado
- Citogenetics and molecular biology laboratory, Consorcio Hospital General Universitario, Valencia, Spain
| | - Raúl Córdoba
- Hematology, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Belén Navarro-Matilla
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain
| | - Margarita Sánchez-Beato
- Lymphoma Research Group, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDPHISA), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- * E-mail: (MSB); (JAGM)
| | - José A. García-Marco
- Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), Madrid, Spain
- * E-mail: (MSB); (JAGM)
| |
Collapse
|
28
|
Crombie JL, Brown JR. The future of antibody therapy in chronic lymphocytic leukemia. Expert Opin Emerg Drugs 2021; 26:323-336. [PMID: 34375544 DOI: 10.1080/14728214.2021.1966414] [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] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Outcomes in chronic lymphocytic leukemia (CLL) have been dramatically improved with the addition of anti-CD20 antibodies to chemotherapy, defining a new standard of care for many years. More recently, therapies targeting fundamental signaling and anti-apoptotic pathways within the CLL cell have demonstrated dramatic clinical responses, including in patients with high-risk prognostic markers, thus emerging as preferred therapy for many patients. While the addition of anti-CD20 antibodies to traditional chemotherapy resulted in significant improvements in outcomes, the role of monoclonal antibodies in the era of targeted agents remains an active area of investigation. Furthermore, since the advent of next-generation anti-CD20 antibodies, the role of specific anti-CD20 antibodies remains an open question. AREAS COVERED In this review, we highlight the important role that anti-CD20 antibody therapy has had in the field of CLL, both when used with chemotherapy and in combination with targeted therapy, as well as the current studies that are further exploring this treatment paradigm in the modern era. EXPERT OPINION While anti-CD20 antibodies have played a pivotal role in the treatment of CLL, additional studies will be required to determine the optimal application of these therapies in combination with targeted therapy.
Collapse
Affiliation(s)
- Jennifer L Crombie
- Department of Medical Oncology , Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jennifer R Brown
- Department of Medical Oncology , Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
29
|
Cherng HJJ, Jain N. First-Line Therapy for Chronic Lymphocytic Leukemia: Bruton Tyrosine Kinase or BCL2 or Both? Hematol Oncol Clin North Am 2021; 35:725-738. [PMID: 34174983 PMCID: PMC9185694 DOI: 10.1016/j.hoc.2021.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Novel therapies largely have replaced chemoimmunotherapy as optimal first-line treatment of chronic lymphocytic leukemia (CLL). Approved novel therapies for CLL in the first-line setting include Bruton tyrosine kinase inhibitors, ibrutinib and acalabrutinib, and the BCL2 inhibitor venetoclax. Each of these novel agents has its own unique attributes and they have not been compared head to head in randomized trials. This review summarizes the pivotal trials that led to the approval of novel agents and compares the features of each agent to guide treatment decisions in treatment-naïve CLL. Ongoing studies investigating combinations of novel agents in the first-line setting also are discussed.
Collapse
Affiliation(s)
- Hua-Jay J Cherng
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0463, Houston, TX 77030, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0428, Houston, TX 77030, USA.
| |
Collapse
|
30
|
A fixed-duration, measurable residual disease-guided approach in CLL: follow-up data from the phase 2 ICLL-07 FILO trial. Blood 2021; 137:1019-1023. [PMID: 33167024 DOI: 10.1182/blood.2020008164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/07/2020] [Indexed: 11/20/2022] Open
Abstract
Trials assessing first-line, fixed-duration approaches in chronic lymphocytic leukemia (CLL) are yielding promising activity, but few long-term data are available. We report follow-up data from a phase 2 trial (ICLL07 FILO) in previously untreated, medically fit patients (N = 135). Patients underwent obinutuzumab-ibrutinib induction for 9 months; then, following evaluation (N = 130 evaluable), those in complete remission and with bone marrow measurable residual disease (BM MRD) <0.01% (n = 10) received ibrutinib for 6 additional months; those in partial remission and/or with BM MRD ≥0.01%, the majority (n = 120), also received 4 cycles of immunochemotherapy (fludarabine/cyclophosphamide-obinutuzumab). Beyond end of treatment, responses were assessed every 3 month and peripheral blood MRD every 6 months. At median follow-up 36.7 months from treatment start, progression-free and overall survival rates (95% confidence interval) at 3 years were 95.7% (92.0% to 99.5%) and 98% (95.1% to 100%), respectively. Peripheral blood MRD <0.01% rates were 97%, 96%, 90%, 84%, and 89% at months 16, 22, 28, 34, and 40, respectively. No new treatment-related or serious adverse event occurred beyond end of treatment. Thus, in previously untreated, medically fit patients with CLL, a fixed-duration (15 months), MRD-guided approach achieved high survival rates, a persistent MRD benefit beyond the end of treatment, and low long-term toxicity. This trial was registered at www.clinicaltrials.gov as #NCT02666898.
Collapse
|
31
|
The Biology of Chronic Lymphocytic Leukemia: Diagnostic and Prognostic Implications. ACTA ACUST UNITED AC 2021; 27:266-274. [PMID: 34398553 DOI: 10.1097/ppo.0000000000000534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
ABSTRACT The high degree of clinical heterogeneity of chronic lymphocytic leukemia (CLL) is influenced by the disease molecular complexity. Genetic studies have allowed to better understand CLL biology and to identify molecular biomarkers of clinical relevance. TP53 disruption represents the strongest prognosticator of chemorefractoriness and indicates the use of Bruton tyrosine kinase inhibitors (BTKis) and BCL2 inhibitors. Unmutated IGHV (immunoglobulin heavy variable) genes also predict refractoriness to chemoimmunotherapy; importantly, when treated with B-cell receptor inhibitors or BCL2 inhibitors, IGHV unmutated patients display an outcome similar to that of IGHV mutated CLL. Before choosing treatment, a comprehensive assessment of TP53 and IGHV status is recommended by all guidelines for CLL clinical management. In case of fixed-duration therapeutic strategies, monitoring of minimal residual disease may provide a tool to decide treatment duration. The current precision medicine management of CLL patients might be further improved by the adoption of novel biomarkers that are emerging as clinically meaningful for this disease.
Collapse
|
32
|
Abstract
ABSTRACT In this article, we carry out an overview on the management options available for chronic lymphocytic leukemia (CLL) patients and discuss possible treatment decisions, taking into account the issue of sustainability and availability. Targeted agents have shown to be superior compared with chemoimmunotherapy (CIT) in terms of progression-free survival in high-risk CLL. In the majority of studies, however, continuous treatment was compared with fixed-duration CIT and no overall survival or progression-free survival-2 (time from randomization to second progression or death) advantage could be documented. Meanwhile, a substantial financial burden on both patients and payers has raised issues about affordability and adherence to treatment. Therefore, value-based pricing of new drugs has been used to set up price negotiation policies in several countries, and fixed-duration therapy has shown to be less costly than continuous treatment. Thus, CIT continues to have a role in the treatment of CLL patients with a favorable genetic profile, that is, with a mutated IGHV gene profile and a wild-type TP53. Targeted treatment represents the preferred choice in patients with an unmutated IGHV gene configuration and/or a TP53 disruption, provided that adherence to treatment is guaranteed and bearing in mind that should costly drugs not be available for frontline treatment, new agents can be very effective as first salvage treatment.
Collapse
|
33
|
Time-limited, Combined Regimen in Chronic Lymphocytic Leukemia: A Promising Strategy to Achieve a Drug Holiday. Curr Med Sci 2021; 41:431-442. [PMID: 34181208 DOI: 10.1007/s11596-021-2385-3] [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: 04/22/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
Chemoimmunotherapy (CIT) is defined as standard first line treatment for chronic lymphocytic leukemia (CLL) patients while patients with unfavorable biological characteristics such as unmutated immunoglobulin heavy chain (UM-IGHV) and TP53 aberration failed to benefit from it. The emergency of the small molecular targeted agents including Bruton's tyrosine kinase (BTK) inhibitor (BTKi) leads to a brand-new era, from a CIT to a chemo-free era in CLL. However, the treatment of target agents is not enough to attain a deep remission and high rate of complete remission (CR), especially in patients with high risks. The long duration brought about problems, such as cost, drug resistance and toxicity. To benefit CLL in progression free survival (PFS) and long-term remission, exploration of time-limited therapies, mainly with BTKi plus CIT and BCL2i based combination therapy has become a mainstream in clinical trials. The time-limited combination therapy shed light on the promising potentiality to attain sustainable deep remission and partly overcame the risk factors, although long term follow-up is required to consolidate the conclusion. In this review, we intend to introduce key results of clinical trials with combination therapy, discuss the achievements and limitations and put forward future direction for clinical trial design in this field.
Collapse
|
34
|
Affiliation(s)
- M Hallek
- University Hospital of Cologne Department I of Internal Medicine Cologne Germany
| |
Collapse
|
35
|
From Biomarkers to Models in the Changing Landscape of Chronic Lymphocytic Leukemia: Evolve or Become Extinct. Cancers (Basel) 2021; 13:cancers13081782. [PMID: 33917885 PMCID: PMC8068228 DOI: 10.3390/cancers13081782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/27/2021] [Accepted: 04/05/2021] [Indexed: 12/23/2022] Open
Abstract
Simple Summary Chronic lymphocytic leukemia (CLL) is characterized by a highly variable clinical course. Thus, predicting the outcome of patients with this disease is a topic of special interest. The rapidly changing treatment landscape of CLL has questioned the value of classical biomarkers and prognostic models. Herein we examine the current state-of-the-art of prognostic and predictive biomarkers in the setting of new oral targeted agents with special focus on the most controversial findings over the last years. We also discuss the available information on the role of “old” and “new” prognostic models in the era of oral small molecules. Abstract Chronic lymphocytic leukemia (CLL) is an extremely heterogeneous disease. With the advent of oral targeted agents (Tas) the treatment of CLL has undergone a revolution, which has been accompanied by an improvement in patient’s survival and quality of life. This paradigm shift also affects the value of prognostic and predictive biomarkers and prognostic models, most of them inherited from the chemoimmunotherapy era but with a different behavior with Tas. This review discusses: (i) the role of the most relevant prognostic and predictive biomarkers in the setting of Tas; and (ii) the validity of classic and new scoring systems in the context of Tas. In addition, a critical point of view about predictive biomarkers with special emphasis on 11q deletion, novel resistance mutations, TP53 abnormalities, IGHV mutational status, complex karyotype and NOTCH1 mutations is stated. We also go over prognostic models in early stage CLL such as IPS-E. Finally, we provide an overview of the applicability of the CLL-IPI for patients treated with Tas, as well as the emergence of new models, generated with data from patients treated with Tas.
Collapse
|
36
|
Sharman JP, Brander DM, Mato AR, Ghosh N, Schuster SJ, Kambhampati S, Burke JM, Lansigan F, Schreeder MT, Lunin SD, Zweibach A, Shtivelband M, Travis PM, Chandler JC, Kolibaba KS, Sportelli P, Miskin HP, Weiss MS, Flinn IW. Ublituximab plus ibrutinib versus ibrutinib alone for patients with relapsed or refractory high-risk chronic lymphocytic leukaemia (GENUINE): a phase 3, multicentre, open-label, randomised trial. LANCET HAEMATOLOGY 2021; 8:e254-e266. [PMID: 33631112 DOI: 10.1016/s2352-3026(20)30433-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Patients with chronic lymphocytic leukaemia and high-risk features have poorer outcomes on ibrutinib than those without high-risk features. The aim of this study was to assess the benefit of adding ublituximab, an anti-CD20 monoclonal antibody, to ibrutinib therapy in this population. METHODS We did a randomised, phase 3, multicentre study (GENUINE) of patients aged 18 years or older with relapsed or refractory chronic lymphocytic leukaemia with at least one of 17p deletion, 11q deletion, or TP53 mutation, at 119 clinics in the USA and Israel. Eligible patients had received at least one previous chronic lymphocytic leukaemia therapy and had an Eastern Cooperative Oncology Group performance status of 2 or lower. We randomised patients (1:1) using permuted block randomisation with a block size of four and stratified by previous lines of therapy (one vs two or more) to receive ibrutinib alone or ibrutinib in combination with ublituximab. Treatment allocation was not masked to patients or investigators. Ibrutinib was given orally daily at 420 mg for all cycles. Ublituximab was given intravenously in 28-day cycles, with increasing doses during cycle 1 (≤150 mg on day 1, 750 mg on day 2, and 900 mg on days 8 and 15) and continuing at 900 mg on day 1 of cycles 2-6. After cycle 6, ublituximab was given at 900 mg every three cycles. The study was initially designed with co-primary endpoints of progression-free survival and overall response rate but due to protracted patient accrual, the protocol was amended to have a single primary endpoint of independent review committee-assessed overall response rate (defined as the proportion of patients who had a partial response, complete response, or complete response with incomplete marrow recovery according to the 2008 International Workshop on CLL criteria) in the intention-to-treat population. Safety was evaluated in the population of patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02301156, and the final analysis is presented. FINDINGS 224 patients were assessed for eligibility, of whom 126 patients were enrolled and randomly assigned to receive ublituximab plus ibrutinib (n=64) or ibrutinib alone (n=62) between Feb 6, 2015, and Dec 19, 2016. After a median follow-up of 41·6 months (IQR 36·7-47·3), the overall response rate was 53 (83%) of 64 patients in the ublituximab plus ibrutinib group and 40 (65%) of 62 patients in the ibrutinib group (p=0·020). 117 patients, including 59 in the ublituximab plus ibrutinib group and 58 in the ibrutinib group, received at least one dose of treatment and were included in safety analyses. Most adverse events were grade 1 or 2. The most common grade 3 and 4 adverse events were neutropenia (11 [19%] patients in the ublituximab plus ibrutinib group and seven [12%] in the ibrutinib group), anaemia (five [8%] and five [9%]), and diarrhoea (six [10%] and three [5%]). The most common serious adverse events were pneumonia (six [10%] in the ublituximab plus ibrutinib group and four [7%] in the ibrutinib group), atrial fibrillation (four [7%] and one [2%]), sepsis (four [7%] and one [2%]), and febrile neutropenia (three [5%] and one [2%]). Two patients in the ublituximab plus ibrutinib group died due to adverse events (one cardiac arrest and one failure to thrive), neither of which were treatment-related. Five patients in the ibrutinib group died due to adverse events, including one cardiac arrest, one cerebral infarction, one intracranial haemorrhage, one Pneumocystis jirovecii pneumonia infection, and one unexplained death; the death due to cardiac arrest was considered to be treatment-related. INTERPRETATION The addition of ublituximab to ibrutinib resulted in a statistically higher overall response rate without affecting the safety profile of ibrutinib monotherapy in patients with relapsed or refractory high-risk chronic lymphocytic leukaemia. These findings provide support for the addition of ublituximab to Bruton tyrosine kinase inhibitors for the treatment of these patients. FUNDING TG Therapeutics.
Collapse
Affiliation(s)
- Jeff P Sharman
- Willamette Valley Cancer Institute, US Oncology Research, Eugene, OR, USA.
| | - Danielle M Brander
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Health System, Durham, NC, USA
| | - Anthony R Mato
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Nilanjan Ghosh
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Stephen J Schuster
- Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Suman Kambhampati
- Sarah Cannon Cancer Center at Research Medical Center, Kansas City, MO, USA
| | - John M Burke
- Rocky Mountain Cancer Centers, US Oncology Research, Aurora, CO, USA
| | - Frederick Lansigan
- Hematology/Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | | | | | - Alexander Zweibach
- Cancer Care Centers of South Texas, US Oncology Research, New Braunfels, TX, USA
| | | | | | | | | | | | | | | | - Ian W Flinn
- Sarah Cannon Research Institute, Nashville, TN, USA
| |
Collapse
|
37
|
Stilgenbauer S, Bosch F, Ilhan O, Kisro J, Mahé B, Mikuskova E, Osmanov D, Reda G, Robinson S, Tausch E, Turgut M, Wójtowicz M, Böttcher S, Perretti T, Trask P, Van Hoef M, Leblond V, Foà R. Safety and efficacy of obinutuzumab alone or with chemotherapy in previously untreated or relapsed/refractory chronic lymphocytic leukaemia patients: Final analysis of the Phase IIIb GREEN study. Br J Haematol 2021; 193:325-338. [PMID: 33605445 DOI: 10.1111/bjh.17326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/21/2020] [Indexed: 02/04/2023]
Abstract
The manageable toxicity profile of obinutuzumab (GA101; G) alone or with chemotherapy in first-line (1L; fit and non-fit) and relapsed/refractory (R/R) patients with chronic lymphocytic leukaemia (CLL) was established in the primary analysis of the Phase IIIb GREEN trial (Clinicaltrials.gov: NCT01905943). The final analysis (cut-off, 31 January 2019) is reported here. Patients received G (1000 mg) alone (G-mono; fit and non-fit patients) or with chemotherapy [fludarabine and cyclophosphamide (FC; fit patients); chlorambucil (non-fit patients); bendamustine (any patient)]. Study endpoints were safety (primary) and efficacy (secondary). Subgroup analyses were performed on prognostic biomarkers in 1L CLL. Overall, 630 patients received 1L and 341 received R/R CLL treatment. At the final analysis, no new safety signals were observed [Grade ≥ 3 adverse events (AEs): 1L 82·7%, R/R 84·5%; serious AEs: 1L 58·1%, R/R 62·5%]. Neutropenia (1L 50·5%, R/R 53·4%) and thrombocytopenia (1L 14·6%, R/R 19·1%) were the most common Grade 3-5 AEs. G-mono-, G-bendamustine and G-FC-treated patients with unmutated immunoglobulin heavy chain trended towards shorter progression-free survival. Achievement of minimal residual disease negativity was greatest in 1L patients treated with G-FC. In this final analysis of the GREEN trial, the safety profile of G was consistent with current risk management strategies. Biomarker analyses supported efficacy in the specific subgroups.
Collapse
Affiliation(s)
- Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Ulm and Innere Medizin I, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Francesc Bosch
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Osman Ilhan
- Internal Medical Sciences Departments, Ankara University School of Medicine, Ankara, Turkey
| | - Jens Kisro
- Onkologische Schwerpunktpraxis Lübeck, Lübeck, Germany
| | - Béatrice Mahé
- Clinical Hematology, CHU Nantes Hôtel-Dieu, Nantes, France
| | - Eva Mikuskova
- Department of Hemato-oncology II, National Cancer Institute, Bratislava, Slovakia Blokhin
| | - Dzhelil Osmanov
- Cancer Research Center, Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Gianluigi Reda
- UOC Ematologia - Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Sue Robinson
- QEII Health Sciences Centre, Halifax, NS, Canada
| | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Mehmet Turgut
- Department of Internal Medical Sciences, Ondokuz Mayis University, Samsun, Turkey
| | - Marcin Wójtowicz
- Clinical Department of Hematology, Hematological Oncology and Internal Diseases, Szpital Wojewodski, Opole, Poland
| | - Sebastian Böttcher
- Department III of Internal Medicine, Rostock University Medical Center, Rostock (current affiliation) and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Perretti
- PDB Biostatistics -Medical Affairs, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Peter Trask
- Patient Centered Outcomes Research, Genentech Inc, South San Francisco, CA, USA
| | - Marlies Van Hoef
- Global Product Development - Medical Affairs, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Véronique Leblond
- Clinical Hematology, Sorbonne Université, AP-HP Hôpital Pitié Salpêtrière, Paris, France
| | - Robin Foà
- Division of Hematology, Sapienza University, Rome, Italy
| |
Collapse
|
38
|
Abstract
Patients with chronic lymphocytic leukemia can be divided into three categories: those who are minimally affected by the problem, often never requiring therapy; those that initially follow an indolent course but subsequently progress and require therapy; and those that from the point of diagnosis exhibit an aggressive disease necessitating treatment. Likewise, such patients pass through three phases: development of the disease, diagnosis, and need for therapy. Finally, the leukemic clones of all patients appear to require continuous input from the exterior, most often through membrane receptors, to allow them to survive and grow. This review is presented according to the temporal course that the disease follows, focusing on those external influences from the tissue microenvironment (TME) that support the time lines as well as those internal influences that are inherited or develop as genetic and epigenetic changes occurring over the time line. Regarding the former, special emphasis is placed on the input provided via the B-cell receptor for antigen and the C-X-C-motif chemokine receptor-4 and the therapeutic agents that block these inputs. Regarding the latter, prominence is laid upon inherited susceptibility genes and the genetic and epigenetic abnormalities that lead to the developmental and progression of the disease.
Collapse
MESH Headings
- Disease Progression
- Humans
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Mutation
- PAX5 Transcription Factor/metabolism
- Receptors, Antigen, B-Cell
- Signal Transduction
- Tumor Microenvironment
Collapse
Affiliation(s)
- Nicholas Chiorazzi
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Shih-Shih Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York 11030, USA
| | - Kanti R Rai
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York 11549, USA
| |
Collapse
|
39
|
Morabito F, Tripepi G, Del Poeta G, Mauro FR, Reda G, Sportoletti P, Laurenti L, Coscia M, Herishanu Y, Bossio S, Varettoni M, Murru R, Chiarenza A, Visentin A, Condoluci A, Moia R, Pietrasanta D, Loseto G, Consoli U, Scortechini I, Rossi FM, Zucchetto A, Al-Janazreh H, Vigna E, Martino EA, Mendicino F, Cassin R, D'Arrigo G, Galimberti S, Rago A, Angeletti I, Biagi A, Del Giudice I, Bomben R, Neri A, Fronza G, Monti P, Menichini P, Cutrona G, Jaksic O, Rossi D, Di Raimondo F, Cuneo A, Gaidano G, Polliack A, Trentin L, Foà R, Ferrarini M, Gattei V, Gentile M. Comparison of ibrutinib and idelalisib plus rituximab in real-life relapsed/resistant chronic lymphocytic leukemia cases. Eur J Haematol 2021; 106:493-499. [PMID: 33378569 DOI: 10.1111/ejh.13573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To compare the capacity of ibrutinib (IB) and idelalisib-rituximab (IDELA-R) of prolonging overall survival (OS) as in CLL patients, previously treated with chemotherapy only. METHODS A real-life cohort of 675 cases has been identified and investigated in the database of the groups participating in the study. RESULTS At an unadjusted univariate analysis, a significant death risk reduction was observed favoring IB (IDELA-R vs IB HR = 0.5, 95% CI = 0.36-0.71) although with some limitations due to the non-randomized and retrospective nature of the study and to the lower number of patients in the IDELA-R group (112 cases) related to the current prescribing practice. To overcome the potential problem of confounding by indication, we adjusted the association between the type of therapy and mortality for all variables significantly associated with OS at Cox univariate analysis. Furthermore, those variables, differently distributed between the two study groups, were introduced into the multivariate Cox model to improve the effectiveness of the analysis. By introducing all these variables into the multiple Cox regression model, we confirmed the protective effect of IB vs IDELA-R (HR = 0.67, 95% CI = 0.45-0.98, P = .04) independent of potential confounders. CONCLUSIONS Although our analysis presents some constraints, that is, the unavailability of additional potential confounders, and the retrospective nature of the study, this observation may be of help for the daily clinical practice, particularly in the absence of randomized trials comparing the two schedules.
Collapse
Affiliation(s)
- Fortunato Morabito
- Biothecnology Research Unit, AO of Cosenza, Cosenza, Italy.,Hematology and Bone Marrow Transplant Unit, Hemato-Oncology Department, Augusta Victoria Hospital, East Jerusalem, Israel
| | - Giovanni Tripepi
- CNR-IFC, Research Unit of Reggio Calabria, Reggio Calabria, Italy
| | - Giovanni Del Poeta
- Division of Hematology, S. Eugenio Hospital and University of Tor Vergata, Rome, Italy
| | | | - Gianluigi Reda
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | - Paolo Sportoletti
- Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Luca Laurenti
- Fondazione Universitaria Policlinico A Gemelli di Roma, Roma, Italy
| | - Marta Coscia
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
| | - Yair Herishanu
- Sourasky Medical Center, Institute of Hematology and Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Sabrina Bossio
- Biothecnology Research Unit, AO of Cosenza, Cosenza, Italy
| | - Marzia Varettoni
- Division of Haematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Roberta Murru
- Hematology and Stem Cell Transplantation Unit, Ospedale A. Businco, Cagliari, Italy
| | - Annalisa Chiarenza
- Division of Hematology, Policlinico, Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Andrea Visentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
| | | | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Daniela Pietrasanta
- Division of Hematology, Azienda Ospedaliera SS Arrigo e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Giacomo Loseto
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Ugo Consoli
- Hematology Department, G. Garibaldi Hospital, Catania, Italy
| | | | - Francesca Maria Rossi
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Hamdi Al-Janazreh
- Hematology and Bone Marrow Transplant Unit, Hemato-Oncology Department, Augusta Victoria Hospital, East Jerusalem, Israel
| | - Ernesto Vigna
- Biothecnology Research Unit, AO of Cosenza, Cosenza, Italy.,Hematology Unit AO of Cosenza, Cosenza, Italy
| | | | | | - Ramona Cassin
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | | | - Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ilaria Angeletti
- Reparto di Oncoematologia Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Annalisa Biagi
- Division of Hematology, S. Eugenio Hospital and University of Tor Vergata, Rome, Italy
| | - Ilaria Del Giudice
- Department of Translational and Precision Medicine, 'Sapienza' University, Rome, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Antonino Neri
- Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy
| | - Gilberto Fronza
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Monti
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Menichini
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Ozren Jaksic
- Department of Hematology, Dubrava Univerisity Hospital, Zagreb, Croatia
| | - Davide Rossi
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Francesco Di Raimondo
- Division of Hematology, Policlinico, Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Antonio Cuneo
- Hematology Section, Department of Medical Sciences, University of Ferrara, Cona, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Aaron Polliack
- Department of Hematology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livio Trentin
- Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy
| | - Robin Foà
- Department of Translational and Precision Medicine, 'Sapienza' University, Rome, Italy
| | - Manlio Ferrarini
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Massimo Gentile
- Biothecnology Research Unit, AO of Cosenza, Cosenza, Italy.,Hematology Unit AO of Cosenza, Cosenza, Italy
| |
Collapse
|
40
|
Guo C, Gao YY, Ju QQ, Zhang CX, Gong M, Li ZL. HELQ and EGR3 expression correlate with IGHV mutation status and prognosis in chronic lymphocytic leukemia. J Transl Med 2021; 19:42. [PMID: 33485349 PMCID: PMC7825181 DOI: 10.1186/s12967-021-02708-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/16/2021] [Indexed: 11/16/2022] Open
Abstract
Background IGHV mutation status is a crucial prognostic biomarker for CLL. In the present study, we investigated the transcriptomic signatures associating with IGHV mutation status and CLL prognosis. Methods The co-expression modules and hub genes correlating with IGHV status, were identified using the GSE28654, by ‘WGCNA’ package and R software (version 4.0.2). The over-representation analysis was performed to reveal enriched cell pathways for genes of correlating modules. Then 9 external cohorts were used to validate the correlation of hub genes expression with IGHV status or clinical features (treatment response, transformation to Richter syndrome, etc.). Moreover, to elucidate the significance of hub genes on disease course and prognosis of CLL patients, the Kaplan–Meier analysis for the OS and TTFT of were performed between subgroups dichotomized by the median expression value of individual hub genes. Results 2 co-expression modules and 9 hub genes ((FCRL1/FCRL2/HELQ/EGR3LPL/LDOC1/ZNF667/SOWAHC/SEPTIN10) correlating with IGHV status were identified by WGCNA, and validated by external datasets. The modules were found to be enriched in NF-kappaB, HIF-1 and other important pathways, involving cell proliferation and apoptosis. The expression of hub genes was revealed to be significantly different, not only between CLL and normal B cell, but also between various types of lymphoid neoplasms. HELQ expression was found to be related with response of immunochemotherapy treatment significantly (p = 0.0413), while HELQ and ZNF667 were expressed differently between stable CLL and Richter syndrome patients (p < 0.0001 and p = 0.0278, respectively). By survival analysis of subgroups, EGR3 expression was indicated to be significantly associated with TTFT by 2 independent cohorts (GSE39671, p = 0.0311; GSE22762, p = 0.0135). While the expression of HELQ and EGR3 was found to be associated with OS (p = 0.0291 and 0.0114 respectively).The Kras, Hedgehog and IL6-JAK-STAT3 pathways were found to be associating with the expression of hub genes, resulting from GSEA. Conclusions The expression of HELQ and EGR3 were correlated with IGHV mutation status in CLL patients. Additionally, the expression of HELQ/EGR3 were prognostic markers for CLL associating with targetable cell signaling pathways.
Collapse
Affiliation(s)
- Chao Guo
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China
| | - Ya-Yue Gao
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China
| | - Qian-Qian Ju
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China
| | - Chun-Xia Zhang
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China
| | - Ming Gong
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China
| | - Zhen-Ling Li
- Department of Hematology, China-Japan Friendship Hospital, Yinghua East Street, Beijing, 100029, China.
| |
Collapse
|
41
|
Condoluci A, Rossi D. Genomic Instability and Clonal Evolution in Chronic Lymphocytic Leukemia: Clinical Relevance. J Natl Compr Canc Netw 2020; 19:227-233. [PMID: 33383567 DOI: 10.6004/jnccn.2020.7623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/15/2020] [Indexed: 11/17/2022]
Abstract
Genomic instability and clonal heterogeneity can influence cancer progression, response to therapy, and relapse. Chronic lymphocytic leukemia (CLL) harbors a variety of clones and subclones that will evolve differently according to intrinsic (microenvironment) and extrinsic (therapy) pressures. Different patterns of clonal evolution have been described, providing insights into the CLL leukemic cell, dynamics, selection, and treatment refractoriness. With the help of genomic technologies allowing a granular resolution of CLL clones, novel synergic therapeutic strategies can be tested with the aim of reaching a genomic-epigenomic ultrapersonalized, tailored approach. These efforts should consider the presence of targetable alterations, continuous cancer reshaping conferring disease refractoriness, and intratumoral clonal equilibrium to possibly avoid clonal selection.
Collapse
Affiliation(s)
- Adalgisa Condoluci
- 1Division of Hematology, Oncology Institute of Southern Switzerland, and.,2Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Davide Rossi
- 1Division of Hematology, Oncology Institute of Southern Switzerland, and.,2Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| |
Collapse
|
42
|
Lee JC, Lamanna N. Is There a Role for Chemotherapy in the Era of Targeted Therapies? Curr Hematol Malig Rep 2020; 15:72-82. [PMID: 32107713 DOI: 10.1007/s11899-020-00563-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The treatment landscape of chronic lymphocytic leukemia has been rapidly evolving over the past few years. The prior standard of care, chemoimmunotherapy, is being replaced by targeted agents, and the utility of chemotherapy has come under question. In this review, we examine recent data comparing chemoimmunotherapy to targeted agents, how these data impact clinical management, and whether there are potential future roles for cytotoxic chemotherapy. RECENT FINDINGS Clinical trials have shown improved clinical outcomes with targeted agents compared to traditional chemoimmunotherapy. Based on these data, the current treatment paradigm primarily favors targeted agents over chemoimmunotherapy, with a few exceptions. However, targeted agents have notable limitations, and thus, there may be a future role of cytotoxic chemotherapy when administered in combination with targeted agents. Although targeted agents have nearly replaced chemoimmunotherapy in the treatment of chronic lymphocytic leukemia, novel combinations utilizing chemotherapy are being developed that may lead to better outcomes.
Collapse
Affiliation(s)
| | - Nicole Lamanna
- Columbia University Medical Center, NY, USA. .,Associate Attending, Leukemia Service, Director of the Chronic Lymphocytic Leukemia Program, Hematologic Malignancies Section, Herbert Irving Comprehensive Cancer Center, New York-Presbyterian/Columbia University Medical Center, NY, USA.
| |
Collapse
|
43
|
Awan FT, Al-Sawaf O, Fischer K, Woyach JA. Current Perspectives on Therapy for Chronic Lymphocytic Leukemia. Am Soc Clin Oncol Educ Book 2020; 40:1-10. [PMID: 32239979 DOI: 10.1200/edbk_279099] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Therapy for chronic lymphocytic leukemia has improved dramatically over the past decade with the introduction of new targeted therapies and a paradigm shift toward targeted therapies for the majority of patients. Better understanding of prognostic factors has helped tailor therapy for individual patients, and work continues to identify optimal therapy for each patient. When therapy is required, most patients will be treated with targeted therapies, either the Bruton tyrosine kinase (BTK) inhibitors ibrutinib or acalabrutinib or the BCL-2 inhibitor venetoclax in combination with obinutuzumab. Without head-to-head comparisons showing differential efficacy among these options, considerations regarding safety, patient preference, and ability to sequence therapy currently influence treatment decisions. Also, clinical trials investigating combinations of these therapies have the potential to further change the standard of care. In this review, we cover the currently available options for the frontline treatment of chronic lymphocytic leukemia (CLL) and discuss safety considerations and toxicity management with each agent as well as novel combination strategies currently under investigation.
Collapse
Affiliation(s)
- Farrukh T Awan
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX
| | - Othman Al-Sawaf
- Department of Internal Medicine and Center of Integrated Oncology Cologne Bonn, University Hospital, Cologne, Germany
| | - Kirsten Fischer
- Department of Internal Medicine and Center of Integrated Oncology Cologne Bonn, University Hospital, Cologne, Germany
| | - Jennifer A Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| |
Collapse
|
44
|
Mauro FR, Molica S, Soddu S, Ilariucci F, Coscia M, Zaja F, Angelucci E, Re F, Liberati AM, Tedeschi A, Reda G, Pietrasanta D, Gozzetti A, Battistini R, Del Poeta G, Musolino C, Nanni M, Piciocchi A, Vignetti M, Neri A, Albano F, Cuneo A, Del Giudice I, Starza ID, De Propris MS, Raponi S, Guarini AR, Foà R. High rate of MRD-responses in young and fit patients with IGHV mutated chronic lymphocytic leukemia treated with front-line fludarabine, cyclophosphamide, and intensified dose of ofatumumab (FCO2). Haematologica 2020; 105:2671-2674. [PMID: 33131259 PMCID: PMC7604632 DOI: 10.3324/haematol.2019.235705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Francesca R. Mauro
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Stefano Molica
- Department of Hematology, Pugliese Ciaccio Hospital, Catanzaro
| | - Stefano Soddu
- Italian Group for Adult Hematologic Diseases (GIMEMA) Foundation, Rome
| | | | - Marta Coscia
- Division of Hematology, A.O.U. Città della Salute e della Scienza di Torino and Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino
| | - Francesco Zaja
- SC Ematologia, Azienda Sanitaria Universitaria Integrata, Trieste
| | - Emanuele Angelucci
- Ematologia e Centro Trapianti, IRCCS Ospedale Policlinico San Martino, Genova
| | | | - Anna Marina Liberati
- Department of Onco-Hematology, University of Perugia, Santa Maria Hospital, Terni
| | | | - Gianluigi Reda
- Department of Hematology, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico of Milan, Milan
| | - Daniela Pietrasanta
- Department of Hematology, SS. Antonio e Biagio e Cesare Arrigo Hospital, Alessandria
| | - Alessandro Gozzetti
- Hematology, Department of Medical Science Surgery and Neurosciences, University of Siena, Siena
| | | | - Giovanni Del Poeta
- Hematology, Department of Biomedicine and Prevention, University Tor Vergata, Rome
| | | | - Mauro Nanni
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Alfonso Piciocchi
- Italian Group for Adult Hematologic Diseases (GIMEMA) Foundation, Rome
| | - Marco Vignetti
- Italian Group for Adult Hematologic Diseases (GIMEMA) Foundation, Rome
| | - Antonino Neri
- Department of Hematology, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico of Milan, Milan
| | - Francesco Albano
- Emergency and Transplantation Department, Hematology Section, University of Bari, Bari
| | - Antonio Cuneo
- Department of Hematology, S. Anna Hospital, Ferrara, Italy
| | - Ilaria Del Giudice
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Irene Della Starza
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Maria Stefania De Propris
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Sara Raponi
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Anna R Guarini
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| | - Robin Foà
- Department of Hematology and Department of Translational and Precision Medicine, 'Sapienza' University, Rome
| |
Collapse
|
45
|
Moia R, Boggione P, Mahmoud AM, Kodipad AA, Adhinaveni R, Sagiraju S, Patriarca A, Gaidano G. Targeting p53 in chronic lymphocytic leukemia. Expert Opin Ther Targets 2020; 24:1239-1250. [PMID: 33016796 DOI: 10.1080/14728222.2020.1832465] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Genomic studies have allowed to identify molecular predictors for chronic lymphocytic leukemia (CLL) treatment tailoring. TP53 disruption is the strongest predictor of chemo-refractoriness and its assessment is the first decisional node in the disease treatment algorithm. AREAS COVERED The review covers the p53 biological pathway, its genetic alterations and clinical implications in CLL, and its druggable targets. The potential therapeutic options for TP53 disrupted patients are described, including: i) agents circumventing TP53 disruption; ii) targeted therapies restoring the physiological function of mutant p53; and iii) medicines potentiating p53 function. EXPERT OPINION The key approach to improve CLL outcome is treatment tailoring in individual patients. BCR and BCL2 inhibitors have significantly improved CLL survival, however TP53 disrupted patients still have a less favorable outcome than wild type cases, possibly because these novel drugs do not directly target p53 and do not restore the function of the disrupted p53 pathway. Emerging innovative molecules in cancer are able to restore the p53 mutant protein and/or potentiate the activity of the p53 wild type protein. If these compounds were confirmed as efficacious also for CLL, they would represent another step forward in the care of high risk CLL patients with TP53 abnormalities.
Collapse
Affiliation(s)
- Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Paola Boggione
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Abdurraouf Mokhtar Mahmoud
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Ahad Ahmed Kodipad
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Ramesh Adhinaveni
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Sruthi Sagiraju
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Andrea Patriarca
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale , Novara, Italy
| |
Collapse
|
46
|
Thompson P. BTK Inhibitors and Chemoimmunotherapy for CLL. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2020; 20 Suppl 1:S22-S24. [PMID: 32862855 DOI: 10.1016/s2152-2650(20)30449-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Affiliation(s)
- Philip Thompson
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, 77030, United States.
| |
Collapse
|
47
|
Davids MS, Fisher DC, Tyekucheva S, McDonough M, Hanna J, Lee B, Francoeur K, Montegaard J, Odejide O, Armand P, Arnason J, Brown JR. A phase 1b/2 study of duvelisib in combination with FCR (DFCR) for frontline therapy for younger CLL patients. Leukemia 2020; 35:1064-1072. [PMID: 32820271 PMCID: PMC7895867 DOI: 10.1038/s41375-020-01010-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Fludarabine, cyclophosphamide, and rituximab (FCR) is highly effective initial therapy for younger patients with chronic lymphocytic leukemia (CLL); however, most eventually relapse. Duvelisib is a delta/gamma PI3K inhibitor approved for relapsed/refractory CLL. We conducted an investigator-initiated, phase 1b/2 study of duvelisib + FCR (DFCR) as initial treatment for CLL patients aged ≤65. A standard 3 + 3 design included two dose levels of duvelisib (25 mg qd and 25 mg bid). Duvelisib was given for 1 week, then with standard FCR added for up to six 28-day cycles, then up to 2 years of duvelisib maintenance. Thirty-two patients were enrolled. The phase 2 dose of duvelisib was identified as 25 mg bid. Hematologic toxicity was common, and all-grade non-hematologic toxicities included transaminitis (28%), febrile neutropenia (22%), pneumonia (19%), and colitis (6%). The best overall response rate by ITT was 88% (56% CR/CRi and 32% PR). The best rate of bone marrow undetectable minimal residual disease (BM-uMRD) by ITT was 66%. The rate of CR with BM-uMRD at end of combination treatment (primary endpoint) was 25%. Three-year PFS and OS are 73 and 93%, respectively. DFCR is active as initial therapy of younger CLL patients. Immune-mediated and infectious toxicities occurred and required active management.
Collapse
Affiliation(s)
- Matthew S Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - David C Fisher
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Svitlana Tyekucheva
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mikaela McDonough
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brandon Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karen Francoeur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Josie Montegaard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Oreofe Odejide
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jon Arnason
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jennifer R Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
48
|
Moia R, Patriarca A, Mahmoud AM, Ferri V, Favini C, Rasi S, Deambrogi C, Gaidano G. Assessing prognosis of chronic lymphocytic leukemia using biomarkers and genetics. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1804860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Andrea Patriarca
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Abdurraouf Mokhtar Mahmoud
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Valentina Ferri
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Chiara Favini
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Silvia Rasi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Clara Deambrogi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale and Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| |
Collapse
|
49
|
Lee J, Wang YL. Prognostic and Predictive Molecular Biomarkers in Chronic Lymphocytic Leukemia. J Mol Diagn 2020; 22:1114-1125. [PMID: 32615167 DOI: 10.1016/j.jmoldx.2020.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of B cells with a variable clinical course. Prognostication is important to place patients into different risk categories for guiding decisions on clinical management, to treat or not to treat. Although several clinical, cytogenetic, and molecular parameters have been established, in the past decade, a tremendous understanding of molecular lesions has been obtained with the advent of high-throughput sequencing. Meanwhile, rapid advances in the understanding of the CLL oncogenic pathways have led to the development of small-molecule targeting signal transducers, Bruton tyrosine kinase and phosphatidylinositol 3-kinase, as well as anti-apoptotic protein BCL2 apoptosis regulator. After an initial response to these targeted therapies, some patients develop resistance and experience disease progression. Novel gene mutations have been identified that account for some of the drug resistance mechanisms. This article focuses on the prognostic and predictive molecular biomarkers in CLL relevant to the molecular pathology practice, beginning with a review of well-established prognostic markers that have already been incorporated into major clinical guidelines, which will be followed by a discussion of emerging biomarkers that are expected to impact clinical practice soon in the future. Special emphasis will be put on predictive biomarkers related to newer targeted therapies in hopes that this review will serve as a useful reference for molecular diagnostic professionals, clinicians, as well as laboratory investigators and trainees.
Collapse
Affiliation(s)
- Jimmy Lee
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Y Lynn Wang
- Department of Pathology, Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
50
|
Morabito F, Gentile M, Monti P, Recchia AG, Menichini P, Skafi M, Atrash M, De Luca G, Bossio S, Al-Janazreh H, Galimberti S, Salah Z, Morabito L, Mujahed A, Hindiyeh M, Dono M, Fais F, Cutrona G, Neri A, Tripepi G, Fronza G, Ferrarini M. TP53 dysfunction in chronic lymphocytic leukemia: clinical relevance in the era of B-cell receptors and BCL-2 inhibitors. Expert Opin Investig Drugs 2020; 29:869-880. [PMID: 32551999 DOI: 10.1080/13543784.2020.1783239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Patients with TP53 dysfunction, assessed by del(17p) or TP53 mutations, respond poorly to chemo-immunotherapy and fare better with the new therapies (BCR and BCL-2 inhibitors); however, it is unclear whether their response is similar to that of patients without anomalies or whether there is currently an adequate determination of TP53 dysfunction. AREA COVERED A literature search was undertaken on clinical trials and real-world experience data on patients with TP53 dysfunction treated with different protocols. Moreover, data on the TP53 biological function and on the tests currently employed for its assessment were reviewed. EXPERT OPINION Although TP53 dysfunction has less negative influence on the new biological therapies, patients with these alterations, particularly those with biallelic inactivation of TP53, have a worst outcome with these therapies than those without alterations. At present, a determination of TP53, particularly with next generation sequencing (NGS) methodologies, may be sufficient for the identifications of the patients unsuitable for chemo-immunotherapy, although integration with del(17p) would be advisable. For the future, more extensive determinations of the TP53 status, including functional assays, may become part of the current armamentarium for a better patient stratification and treatment with newer protocols.
Collapse
Affiliation(s)
- Fortunato Morabito
- Hematology Department and Bone Marrow Transplant Unit, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel.,Biotechnology Research Unit, Aprigliano, AO/ASP , Cosenza, Italy
| | - Massimo Gentile
- Biotechnology Research Unit, Aprigliano, AO/ASP , Cosenza, Italy.,Hematology Unit, Hematology and Oncology Department , Cosenza, Italy
| | - Paola Monti
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | | | - Paola Menichini
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Mamdouh Skafi
- Hematology Department and Bone Marrow Transplant Unit, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel
| | - Moien Atrash
- Hematology Department and Bone Marrow Transplant Unit, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel
| | - Giuseppa De Luca
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Sabrina Bossio
- Biotechnology Research Unit, Aprigliano, AO/ASP , Cosenza, Italy
| | - Hamdi Al-Janazreh
- Hematology Department and Bone Marrow Transplant Unit, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel
| | | | - Zaidoun Salah
- The Lautenberg Center for General and Tumor Immunology, Department of Immunology and Cancer Research-Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School , Jerusalem, Israel
| | - Lucio Morabito
- Humanitas Clinical and Research Center, IRCCS , Rozzano, Italy
| | - Alham Mujahed
- Laboratory Department, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel
| | - Musa Hindiyeh
- Laboratory Department, Cancer Care Center, Augusta Victoria Hospital , Jerusalem, Israel
| | - Mariella Dono
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Franco Fais
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino , Genova, Italy.,Department of Experimental Medicine, University of Genoa , Genoa, Italy
| | - Giovanna Cutrona
- Molecular Pathology Unit, IRCCS Ospedale Policlinico San Martino , Genova, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milan , Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico , Milan, Italy
| | | | - Gilberto Fronza
- Mutagenesis and Cancer Prevention Unit, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Manlio Ferrarini
- Department of Experimental Medicine, University of Genoa , Genoa, Italy
| |
Collapse
|