1
|
Lao Z, Lam KY, Cheung YMC, Teng CL, Radhakrishnan V, Bhurani D, Ko BS, Goh YT. Recommendations for the treatment and management of adult B-Cell acute lymphoblastic leukemia in Asia-Pacific: Outcomes from a pilot initiative. Asia Pac J Clin Oncol 2024; 20:325-334. [PMID: 38148287 DOI: 10.1111/ajco.14041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/27/2023] [Accepted: 12/07/2023] [Indexed: 12/28/2023]
Abstract
The outcomes of adult B-cell acute lymphoblastic leukemia (ALL) remain poor. Recent advancements in the field of leukemia research show potential for improved patient care. However, the adoption of research findings into clinical practice is fraught with practice- and country-specific challenges. The continued addition of new findings warrants critical evaluation for the feasibility of incorporation into clinical practice. A uniform set of evidence-based guidelines can favorably assist physicians in making optimal clinical decisions. Such a resource may also serve as a reference point for strategic planning of initiatives aimed at addressing critical barriers in the optimal management of B-cell ALL. This initiative was undertaken to seek a collaborative perspective and understand the existing challenges. Concordance-based recommendations were outlined through a systematic discussion on various aspects of treatment and management of adult B-cell ALL. The outcomes and experiences gained from this exercise will serve as a foundation for future efforts encompassing the more granular aspects of the management of B-cell ALL across the Asia-Pacific region.
Collapse
Affiliation(s)
- Zhentang Lao
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| | - Kwong Yok Lam
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - Yuk Man Carol Cheung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| | - Chieh-Lin Teng
- Department of Medicine, Division of Hematology/Medical Oncology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Vivek Radhakrishnan
- Division of Haematology Oncology and Haematopoietic Cell Transplantation, Tata Medical Center, Kolkata, India
| | - Dinesh Bhurani
- Department of Hematology and Bone Marrow Transplant, Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
| | - Bor-Sheng Ko
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Yeow Tee Goh
- Department of Haematology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
2
|
Yanagida E, Kubota A, Miyoshi H, Ohshima K, Kawakita T, Murayama T. The case of T-ALL presenting with NK phenotype after COVID-19 vaccination. Pathol Res Pract 2023; 242:154310. [PMID: 36706586 PMCID: PMC9840814 DOI: 10.1016/j.prp.2023.154310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/16/2023]
Abstract
NK-lymphoblastic leukemia/lymphoma (NK-LL) is an extremely rare hematopoietic tumor consisting of natural killer (NK) precursor cells, and their lineage overlaps with T-cells, making it challenging to diagnose. COVID-19 vaccination is recommended for people with a risk of aggravation such as cancer-bearing patients, including hematopoietic tumors. We present a 55-year-old man who had cervical lymph node swelling post vaccination for COVID-19. Hematological malignancy was suspected due to the presence of atypical lymphoid cells with an elevated IL-2R in laboratory data. Tumor cells were positive for CD7, CD56, cyCD3, and terminal deoxynucleotidyl transferase (TdT) evidenced through flow cytometry of the bone marrow and the lymph node. The histopathological findings showed monotonous tumor cell proliferation, the cells being positive for CD3 and TdT in the bone marrow and they were positive for CD3, TdT, and CD56 in lymph node. Even though these findings suggested NK-LL, clonal T-cell receptor (TCR) β gene rearrangement by Southern blot hybridization was observed in the bone marrow. TCRβ rearrangement led to the final diagnosis of T-cell lymphoblastic leukemia (T-ALL). The causal relationship between COVID-19 vaccination and carcinogenesis is not clear, and more cases need to be studied in order to elucidate the relationship between the two factors.
Collapse
Affiliation(s)
- Eriko Yanagida
- Department of Pathology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan; Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan.
| | - Akira Kubota
- Department of Hematology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Toshiro Kawakita
- Department of Hematology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| | - Toshihiko Murayama
- Department of Pathology, National Hospital Organization, Kumamoto Medical Center, Kumamoto, Japan
| |
Collapse
|
3
|
Nedumannil R, Ritchie D, Bajel A, Ng AP, Harrison SJ, Westerman D. Real-world utility of early measurable residual disease assessments by multi-parametric flow cytometry in adult patients with B-lymphoblastic leukemia receiving Hyper-CVAD induction chemotherapy. Eur J Haematol Suppl 2023; 110:168-176. [PMID: 36321745 DOI: 10.1111/ejh.13890] [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: 08/24/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022]
Abstract
Multi-parametric flow cytometry (MFC) has a well-established role in measurable residual disease (MRD) monitoring in patients with B-lymphoblastic leukemia (B-ALL). However, the optimal time-point (TP) for early MRD testing and associated prognostic impact remain undefined in adult B-ALL patients receiving Hyper-CVAD induction chemotherapy. To evaluate the utility of MRD analysis after one cycle (TP1) in comparison to MRD analysis after two cycles (TP2) of induction treatment with Hyper-CVAD chemotherapy, we studied 49 adult B-ALL patients over a 10-year period (2010-2020) who had available bone marrow samples for morphological and MFC MRD assessments at the two separate TPs. Median times to TP1 and TP2 relative to start of treatment were 21 and 45 days, respectively. When censored at transplant, achievement of MRD negativity at TP1 was not associated with a statistically significant improvement in either event-free survival (EFS) (p = .426) or overall survival (OS) (p = .335) when compared to patients with MRD positivity. In contrast, achieving MRD negativity at TP2 was associated with a statistically significant improvement in both EFS (p = ·005) and OS (p = .047) over patients who remained MRD positive. Multivariate analysis demonstrated that KMT2A-rearrangement and MRD positivity at TP2 were the only significant predictors of outcome, correlating with worse EFS and OS. Therefore, in the absence of residual morphologic disease, MRD analysis after one cycle of Hyper-CVAD induction chemotherapy did not provide additional benefit with regard to risk stratification or correlation with survival outcomes when compared to MRD testing after two cycles of Hyper-CVAD in adult B-ALL patients.
Collapse
Affiliation(s)
- Rithin Nedumannil
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Diagnostic Haematology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - David Ritchie
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashish Bajel
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley P Ng
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Simon J Harrison
- Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - David Westerman
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Nogami A, Sasaki K. Therapeutic Advances in Immunotherapies for Hematological Malignancies. Int J Mol Sci 2022; 23:11526. [PMID: 36232824 PMCID: PMC9569660 DOI: 10.3390/ijms231911526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Following the success of immunotherapies such as chimeric antigen receptor transgenic T-cell (CAR-T) therapy, bispecific T-cell engager therapy, and immune checkpoint inhibitors in the treatment of hematologic malignancies, further studies are underway to improve the efficacy of these immunotherapies and to reduce the complications associated with their use in combination with other immune checkpoint inhibitors and conventional chemotherapy. Studies of novel therapeutic strategies such as bispecific (tandem or dual) CAR-T, bispecific killer cell engager, trispecific killer cell engager, and dual affinity retargeting therapies are also underway. Because of these studies and the discovery of novel immunotherapeutic target molecules, the use of immunotherapy for diseases initially thought to be less promising to treat with this treatment method, such as acute myeloid leukemia and T-cell hematologic tumors, has become a reality. Thus, in this coming era of new transplantation- and chemotherapy-free treatment strategies, it is imperative for both scientists and clinicians to understand the molecular immunity of hematologic malignancies. In this review, we focus on the remarkable development of immunotherapies that could change the prognosis of hematologic diseases. We also review the molecular mechanisms, development processes, clinical efficacies, and problems of new agents.
Collapse
Affiliation(s)
- Ayako Nogami
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
- Department of Hematology, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX 77030, USA
| |
Collapse
|
5
|
Tardif M, Souza A, Krajinovic M, Bittencourt H, Tran TH. Molecular-based and antibody-based targeted pharmacological approaches in childhood acute lymphoblastic leukemia. Expert Opin Pharmacother 2021; 22:1871-1887. [PMID: 34011251 DOI: 10.1080/14656566.2021.1931683] [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: 02/06/2023]
Abstract
Introduction: Despite the significant survival improvement in childhood acutelymphoblastic leukemia (ALL), 15-20% of patients continue to relapse; outcomes following relapse remain suboptimal and have room for further improvement. Advances in genomics have shed new insights on the biology of ALL, led to the discovery of novel genomically defined ALL subtypes, refined prognostic significance and revealed new therapeutic vulnerabilities.Areas covered: In this review, the authors provide an overview of the genomic landscape of childhood ALL and highlight recent advances in molecular-based and antibody-based pharmacological approaches in the treatment of childhood ALL, from emerging preclinical evidence to published results of completed clinical trials.Expert opinion: Molecularly targeted therapies and immunotherapies have expanded the horizons of ALL therapy and represent promising therapeutic avenues for high-risk and relapsed/refractory ALL. These novel therapies are now moving into frontline ALL therapy and may define new treatment paradigms that aim to further improve survival and reduce chemotherapy-related toxicities in the management of pediatric ALL.
Collapse
Affiliation(s)
- Magalie Tardif
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Centre, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Amalia Souza
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Centre, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Maja Krajinovic
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Centre, CHU Sainte-Justine, Montréal, Québec, Canada.,Department of Medicine, Université De Montréal, Montréal, Québec, Canada
| | - Henrique Bittencourt
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Centre, CHU Sainte-Justine, Montréal, Québec, Canada.,Department of Medicine, Université De Montréal, Montréal, Québec, Canada
| | - Thai Hoa Tran
- Division of Pediatric Hematology-Oncology, Charles-Bruneau Cancer Centre, CHU Sainte-Justine, Montréal, Québec, Canada.,Department of Medicine, Université De Montréal, Montréal, Québec, Canada
| |
Collapse
|
6
|
Sasaki K, Jabbour E, Short NJ, Jain N, Ravandi F, Pui C, Kantarjian H. Acute lymphoblastic leukemia: A population-based study of outcome in the United States based on the surveillance, epidemiology, and end results (SEER) database, 1980-2017. Am J Hematol 2021; 96:650-658. [PMID: 33709456 DOI: 10.1002/ajh.26156] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/07/2021] [Indexed: 12/22/2022]
Abstract
The treatment in acute lymphoblastic Leukemia (ALL) has evolved and improved dramatically over the past four decades. We assessed the outcome of ALL overall, and the two major subsets of Philadelphia chromosome (Ph)-positive and Ph-negative ALL by age, time periods, ethnicity, median household income, and geographic county area. A total of 12 788 patients diagnosed with ALL from 1980 to 2017 were included. We performed an analysis to better evaluate the outcome evolution in ALL according to time period and patient's demographic factors. The overall 5-year survival rates have improved significantly over time, from 51% before 1990 to 72% since 2010. The survival rates for children (age 0 to 14 years) and adolescents (age 15 to 19 years) have improved from 73% and 55% before 1990 to 93% and 74% since 2010, respectively. Similarly, the rates had improved from 33% to 59% for adults 20 to 29 years old, 24% to 59% for 30 to 39 years old, and 14% to 43% for 40 to 59 years old between the two time periods. The rates remained under 30% in older patients (60+ years). Since 2010, patients with Ph-negative ALL had 5-year survival rate of 73% and those with Ph-positive ALL 50%. African Americans, Hispanic ethnicity, and lower household income were associated with inferior survival. The outcome of patients with ALL showed continued improvement across all age groups in the US. The recent introduction of targeted therapies, together with optimized supportive care, will continue to improve outcomes, particularly in older patients.
Collapse
Affiliation(s)
- Koji Sasaki
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| | - Elias Jabbour
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| | - Nicholas J. Short
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| | - Nitin Jain
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| | - Farhad Ravandi
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| | - Ching‐Hon Pui
- Departments of Oncology and Pathology St. Jude Children's Research Hospital Memphis Tennessee
| | - Hagop Kantarjian
- Department of Leukemia U.T. M.D. Anderson Cancer Center Houston Texas
| |
Collapse
|
7
|
Value of flow cytometry for MRD-based relapse prediction in B-cell precursor ALL in a multicenter setting. Leukemia 2020; 35:1894-1906. [PMID: 33318611 PMCID: PMC8257490 DOI: 10.1038/s41375-020-01100-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/07/2020] [Accepted: 11/15/2020] [Indexed: 11/17/2022]
Abstract
PCR of TCR/Ig gene rearrangements is considered the method of choice for minimal residual disease (MRD) quantification in BCP-ALL, but flow cytometry analysis of leukemia-associated immunophenotypes (FCM-MRD) is faster and biologically more informative. FCM-MRD performed in 18 laboratories across seven countries was used for risk stratification of 1487 patients with BCP-ALL enrolled in the NOPHO ALL2008 protocol. When no informative FCM-marker was available, risk stratification was based on real-time quantitative PCR. An informative FCM-marker was found in 96.2% and only two patients (0.14%) had non-informative FCM and non-informative PCR-markers. The overall 5-year event-free survival was 86.1% with a cumulative incidence of relapse (CIR5y) of 9.5%. FCM-MRD levels on days 15 (HzR 4.0, p < 0.0001), 29 (HzR 2.7, p < 0.0001), and 79 (HzR 3.5, p < 0.0001) associated with hazard of relapse adjusted for age, cytogenetics, and WBC. The early (day 15) response associated with CIR5y adjusted for day 29 FCM-MRD, with higher levels in adults (median 2.4 × 10−2 versus 5.2 × 10−3, p < 0.0001). Undetectable FCM- and/or PCR-MRD on day 29 identified patients with a very good outcome (CIR5y = 3.2%). For patients who did not undergo transplantation, day 79 FCM-MRD > 10−4 associated with a CIR5y = 22.1%. In conclusion, FCM-MRD performed in a multicenter setting is a clinically useful method for MRD-based treatment stratification in BCP-ALL.
Collapse
|
8
|
Ribera JM, Morgades M, Montesinos P, Tormo M, Martínez-Carballeira D, González-Campos J, Gil C, Barba P, García-Boyero R, Coll R, Pedreño M, Ribera J, Mercadal S, Vives S, Novo A, Genescà E, Hernández-Rivas JM, Bergua J, Amigo ML, Vall-Llovera F, Martínez-Sánchez P, Calbacho M, García-Cadenas I, Garcia-Guiñon A, Sánchez-Sánchez MJ, Cervera M, Feliu E, Orfao A. A pediatric regimen for adolescents and young adults with Philadelphia chromosome-negative acute lymphoblastic leukemia: Results of the ALLRE08 PETHEMA trial. Cancer Med 2020; 9:2317-2329. [PMID: 32022463 PMCID: PMC7131850 DOI: 10.1002/cam4.2814] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 11/19/2022] Open
Abstract
Background Pediatric‐based or ‐inspired trials have improved the prognosis of adolescents and young adults (AYA) with Philadelphia chromosome‐negative (Ph‐neg) acute lymphoblastic leukemia (ALL). Methods This study reports the results of treatment of the ALLRE08 trial, a full pediatric trial for AYA aged 15‐30 years with standard‐risk (SR) ALL. Results From 2008 to 2018, 89 patients (38 adolescents [15‐18 years] and 51 young adults [YA, 19‐30 years], median age: 20 [15‐29] years) were enrolled in the ALLRE08 trial. The complete response (CR) was 95%. Twenty‐two patients were transferred to a high‐risk (HR) protocol because of poor marrow response on day 14 (n = 20) or high‐level of end‐induction minimal residual response (MRD ≥ 0.25%, n = 2). Cumulative incidence of relapse (CIR) at 5 years was 35% (95%CI: 23%‐47%), with significant differences between adolescents and YA: 13% (4%‐28%) vs 52% (34%‐67%), P = .012. No treatment‐related mortality was observed in 66/66 patients following the ALLRE08 trial vs 3/23 patients moved to a HR trial. The estimated 5‐year overall survival (OS) was 74% (95%CI: 63%‐85%), with significantly higher rates for adolescents vs YA: 87% (95%CI: 74%‐100%) vs 63% (46%‐80%), P = .021. Although CIR or OS were lower in patients who were transferred to a HR trial, the differences were not statistically significant (CIR: 34% [21%‐47%] vs 37% [14%‐61%]; OS: 78% [66%‐90%] vs 61% [31%;91%]). Conclusion A full pediatric trial is feasible and effective for AYA with Ph‐neg, SR‐ALL, with better results for adolescents than for YA. Outcome of patients with poor early response rescued with a HR trial was not significantly inferior.
Collapse
Affiliation(s)
- Josep-Maria Ribera
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mireia Morgades
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pau Montesinos
- Departments of Clinical Hematology, Hospital Universitari i Politècnic La Fe, CIBERONC, Instituto Carlos III, Valencia, Spain, Madrid, Spain
| | - Mar Tormo
- Departments of Clinical Hematology, Hospital Clínico Valencia, Valencia, Spain
| | | | - José González-Campos
- Departments of Clinical Hematology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Cristina Gil
- Departments of Clinical Hematology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Pere Barba
- Departments of Clinical Hematology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Raimundo García-Boyero
- Departments of Clinical Hematology, Hospital General Universitario de Castellón, Castelló, Spain
| | - Rosa Coll
- Departments of Clinical Hematology, ICO-Hospital Josep Trueta, Girona, Spain
| | - María Pedreño
- Departments of Clinical Hematology, Hospital Dr. Peset, Valencia, Spain
| | - Jordi Ribera
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Santiago Mercadal
- Departments of Clinical Hematology, ICO-Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Susana Vives
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andrés Novo
- Departments of Clinical Hematology, Hospital Son Espases, Palma de Mallorca, Spain
| | - Eulàlia Genescà
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jesús-María Hernández-Rivas
- Departments of Clinical Hematology, Hospital Clínico Universitario, Cancer Research Center (IBMCC-CSIC/USAL), Cytometry Service CIBERONC, Salamanca, Spain
| | - Juan Bergua
- Departments of Clinical Hematology, Hospital San Pedro de Alcántara, Cáceres, Spain
| | - María-Luz Amigo
- Departments of Clinical Hematology, Hospital Morales Meseguer, Murcia, Spain
| | - Ferran Vall-Llovera
- Departments of Clinical Hematology, Hospital Mútua de Terrassa, Terrassa, Spain
| | | | - María Calbacho
- Departments of Clinical Hematology, Hospital Ramón y Cajal, Madrid, Spain
| | | | | | | | - Marta Cervera
- Departments of Clinical Hematology, ICO-Hospital Joan XXIII, Tarragona, Spain
| | - Evarist Feliu
- Departments of Clinical Hematology, ICO-Hospital Germans Trias i Pujol. Josep Carreras Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alberto Orfao
- Departments of Clinical Hematology, Hospital Clínico Universitario, Cancer Research Center (IBMCC-CSIC/USAL), Cytometry Service CIBERONC, Salamanca, Spain
| | | |
Collapse
|
9
|
Deak D, Pop C, Zimta AA, Jurj A, Ghiaur A, Pasca S, Teodorescu P, Dascalescu A, Antohe I, Ionescu B, Constantinescu C, Onaciu A, Munteanu R, Berindan-Neagoe I, Petrushev B, Turcas C, Iluta S, Selicean C, Zdrenghea M, Tanase A, Danaila C, Colita A, Colita A, Dima D, Coriu D, Einsele H, Tomuleasa C. Let's Talk About BiTEs and Other Drugs in the Real-Life Setting for B-Cell Acute Lymphoblastic Leukemia. Front Immunol 2020; 10:2856. [PMID: 31921126 PMCID: PMC6934055 DOI: 10.3389/fimmu.2019.02856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/20/2019] [Indexed: 01/07/2023] Open
Abstract
Background: Therapy for acute lymphoblastic leukemia (ALL) are currently initially efficient, but even if a high percentage of patients have an initial complete remission (CR), most of them relapse. Recent data shows that immunotherapy with either bispecific T-cell engagers (BiTEs) of chimeric antigen receptor (CAR) T cells can eliminate residual chemotherapy-resistant B-ALL cells. Objective: The objective of the manuscript is to present improvements in the clinical outcome for chemotherapy-resistant ALL in the real-life setting, by describing Romania's experience with bispecific antibodies for B-cell ALL. Methods: We present the role of novel therapies for relapsed B-cell ALL, including the drugs under investigation in phase I-III clinical trials, as a potential bridge to transplant. Blinatumomab is presented in a critical review, presenting both the advantages of this drug, as well as its limitations. Results: Bispecific antibodies are discussed, describing the clinical trials that resulted in its approval by the FDA and EMA. The real-life setting for relapsed B-cell ALL is described and we present the patients treated with blinatumomab in Romania. Conclusion: In the current manuscript, we present blinatumomab as a therapeutic alternative in the bridge-to-transplant setting for refractory or relapsed ALL, to gain a better understanding of the available therapies and evidence-based data for these patients in 2019.
Collapse
Affiliation(s)
- Dalma Deak
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Cristina Pop
- Department of Pharmacology, Faculty of Pharmacy, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandra Ghiaur
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Angela Dascalescu
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Ion Antohe
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Bogdan Ionescu
- Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Constantinescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Onaciu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Munteanu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bobe Petrushev
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Turcas
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Sabina Iluta
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Selicean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Catalin Danaila
- Department of Hematology, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania.,Department of Hematology, Regional Institute of Oncology, Iasi, Romania
| | - Anca Colita
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania.,Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Andrei Colita
- Department of Hematology, Coltea Hospital, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Delia Dima
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Daniel Coriu
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.,Department of Hematology, Fundeni Clinical Institute, Bucharest, Romania.,Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Wurzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Department of Hematology/Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
10
|
Della Starza I, Chiaretti S, De Propris MS, Elia L, Cavalli M, De Novi LA, Soscia R, Messina M, Vitale A, Guarini A, Foà R. Minimal Residual Disease in Acute Lymphoblastic Leukemia: Technical and Clinical Advances. Front Oncol 2019; 9:726. [PMID: 31448230 PMCID: PMC6692455 DOI: 10.3389/fonc.2019.00726] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
Introduction: Acute lymphoblastic leukemia (ALL) is the first neoplasm where the assessment of early response to therapy by minimal residual disease (MRD) monitoring has proven to be a fundamental tool to guide therapeutic choices. The most standardized methods to study MRD in ALL are multi-parametric flow cytometry (MFC) and polymerase chain reaction (PCR) amplification-based methods. Emerging technologies hold the promise to improve MRD detection in ALL patients. Moreover, novel therapies, such as monoclonal antibodies, bispecific T-cell engagers, and chimeric antigen receptor T cells (CART) represent exciting advancements in the management of B-cell precursor (BCP)-ALL. Aims: Through a review of the literature and in house data, we analyze the current status of MRD assessment in ALL to better understand how some of its limitations could be overcome by emerging molecular technologies. Furthermore, we highlight the future role of MRD monitoring in the context of personalized protocols, taking into account the genetic complexity in ALL. Results and Conclusions: Molecular rearrangements (gene fusions and immunoglobulin and T-cell receptor-IG/TR gene rearrangements) are widely used as targets to detect residual leukemic cells in ALL patients. The advent of novel techniques, namely next generation flow cytometry (NGF), digital-droplet-PCR (ddPCR), and next generation sequencing (NGS) appear important tools to evaluate MRD in ALL, since they have the potential to overcome the limitations of standard approaches. It is likely that in the forthcoming future these techniques will be incorporated in clinical trials, at least at decisional time points. Finally, the advent of new powerful compounds is further increasing MRD negativity rates, with benefits in long-term survival and a potential reduction of therapy-related toxicities. However, the prognostic relevance in the setting of novel immunotherapies still needs to be evaluated.
Collapse
Affiliation(s)
- Irene Della Starza
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,GIMEMA Foundation, Rome, Italy
| | - Sabina Chiaretti
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria S De Propris
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Loredana Elia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Cavalli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia A De Novi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberta Soscia
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Monica Messina
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonella Vitale
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Anna Guarini
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Yu YB, Zhao T, Wang J, Jia JS, Zhu HH, Jiang H, Huang XJ, Jiang Q. [Improved 4-week complete remission rate by additional use of daunorubicin in the medium term of induction chemotherapy in adult Ph-negative acute lymphoblastic leukemia with high proportion of blasts]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2018; 39:676-678. [PMID: 30180471 PMCID: PMC7342841 DOI: 10.3760/cma.j.issn.0253-2727.2018.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Indexed: 11/18/2022]
|
12
|
Cassaday RD, Stevenson PA, Wood BL, Becker PS, Hendrie PC, Sandmaier BM, Radich JL, Shustov AR. Description and prognostic significance of the kinetics of minimal residual disease status in adults with acute lymphoblastic leukemia treated with HyperCVAD. Am J Hematol 2018; 93:546-552. [PMID: 29318644 DOI: 10.1002/ajh.25030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/01/2018] [Accepted: 01/05/2018] [Indexed: 12/29/2022]
Abstract
HyperCVAD is a commonly-used regimen for adults with newly-diagnosed acute lymphoblastic leukemia (ALL). However, relatively little is known about the application of minimal residual disease (MRD) detection with this treatment. To address this, we studied 142 adults with ALL treated with hyperCVAD over a 10-year period who had MRD assessed by either multi-parameter flow cytometry or (for patients with Philadelphia chromosome positive ALL) reverse transcriptase polymerase chain reaction for the BCR-ABL1 translocation. In a multivariate analysis, patients who achieved MRD negativity (MRDNeg ) at any point had significantly better overall survival (OS; hazard ratio [HR] 0.43; P = .01) and event-free survival (EFS; HR 0.27; P < .01). Of 121 patients with MRD assessed at various points within 90 days of starting hyperCVAD, 50% (n = 61) had achieved MRDNeg . Among those that became MRDNeg , the median time to MRDNeg was 68 days. Time to MRDNeg was significantly associated with EFS (P = .009), but not OS (P = .19), implying increasingly better EFS the earlier MRDNeg is achieved. These data add to our understanding of MRD assessment during treatment with hyperCVAD, aide clinicians with predicting relapse risk, and provide additional historical data on which future clinical trials can be designed.
Collapse
Affiliation(s)
- Ryan D. Cassaday
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| | - Philip A. Stevenson
- Clinical Statistics Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| | - Brent L. Wood
- Department of Laboratory Medicine; University of Washington School of Medicine; Seattle Washington
| | - Pamela S. Becker
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| | - Paul C. Hendrie
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
| | - Brenda M. Sandmaier
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| | - Jerald L. Radich
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| | - Andrei R. Shustov
- Department of Medicine; University of Washington School of Medicine; Seattle Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center; Seattle Washington
| |
Collapse
|
13
|
Brüggemann M, Kotrova M. Minimal residual disease in adult ALL: technical aspects and implications for correct clinical interpretation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:13-21. [PMID: 29222232 PMCID: PMC6142572 DOI: 10.1182/asheducation-2017.1.13] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/21/2017] [Indexed: 06/07/2023]
Abstract
Nowadays, minimal residual disease (MRD) is accepted as the strongest independent prognostic factor in acute lymphoblastic leukemia (ALL). It can be detected by molecular methods that use leukemia-specific or patient-specific molecular markers (fusion gene transcripts, or immunoglobulin/T-cell receptor [IG/TR] gene rearrangements), and by multi-parametric flow cytometry. The sensitivity and specificity of these methods can vary across treatment time points and therapeutic settings. Thus, knowledge of the principles and limitations of each technology is of the utmost importance for correct interpretation of MRD results. Time will tell whether new molecular and flow cytometric high-throughput technologies can overcome the limitations of current standard methods and eventually bring additional benefits. MRD during standard ALL chemotherapy is the strongest overall prognostic indicator and has therefore been used for refining initial treatment stratification. Moreover, MRD positivity after the maintenance phase of treatment may point to an impending relapse and thus enable salvage treatment to be initiated earlier, which could possibly improve treatment results. The prognostic relevance of pretransplantation MRD was shown by several studies, and MRD high-risk patients were shown to benefit from stem cell transplantation (SCT). Also, MRD positivity after SCT correlates with worse outcomes. In addition, MRD information is very instructive in current clinical trials that test novel agents to evaluate their treatment efficacy. Although conventional clinical risk factors lose their independent prognostic significance when combined with MRD information, recently identified genetic markers may further improve the treatment stratification in ALL.
Collapse
Affiliation(s)
- Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Michaela Kotrova
- Department of Hematology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
14
|
Minimal residual disease in adult ALL: technical aspects and implications for correct clinical interpretation. Blood Adv 2017; 1:2456-2466. [PMID: 29296895 DOI: 10.1182/bloodadvances.2017009845] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/21/2017] [Indexed: 12/18/2022] Open
Abstract
Nowadays, minimal residual disease (MRD) is accepted as the strongest independent prognostic factor in acute lymphoblastic leukemia (ALL). It can be detected by molecular methods that use leukemia-specific or patient-specific molecular markers (fusion gene transcripts, or immunoglobulin/T-cell receptor [IG/TR] gene rearrangements), and by multi-parametric flow cytometry. The sensitivity and specificity of these methods can vary across treatment time points and therapeutic settings. Thus, knowledge of the principles and limitations of each technology is of the utmost importance for correct interpretation of MRD results. Time will tell whether new molecular and flow cytometric high-throughput technologies can overcome the limitations of current standard methods and eventually bring additional benefits. MRD during standard ALL chemotherapy is the strongest overall prognostic indicator and has therefore been used for refining initial treatment stratification. Moreover, MRD positivity after the maintenance phase of treatment may point to an impending relapse and thus enable salvage treatment to be initiated earlier, which could possibly improve treatment results. The prognostic relevance of pretransplantation MRD was shown by several studies, and MRD high-risk patients were shown to benefit from stem cell transplantation (SCT). Also, MRD positivity after SCT correlates with worse outcomes. In addition, MRD information is very instructive in current clinical trials that test novel agents to evaluate their treatment efficacy. Although conventional clinical risk factors lose their independent prognostic significance when combined with MRD information, recently identified genetic markers may further improve the treatment stratification in ALL.
Collapse
|