151
|
Stein A, Franklin JL, Chia VM, Arrindell D, Kormany W, Wright J, Parson M, Amouzadeh HR, Choudhry J, Joseph G. Benefit-Risk Assessment of Blinatumomab in the Treatment of Relapsed/Refractory B-Cell Precursor Acute Lymphoblastic Leukemia. Drug Saf 2020; 42:587-601. [PMID: 30565020 PMCID: PMC6475509 DOI: 10.1007/s40264-018-0760-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Blinatumomab is the first-and-only Food and Drug Administration (FDA)-approved cluster of differentiation (CD) 19-directed CD3 bispecific T-cell engager (BiTE®) immunotherapy. It is currently FDA approved for the treatment of adults and children with Philadelphia chromosome-positive (Ph+) and Philadelphia chromosome-negative (Ph-) relapsed/refractory (R/R) B-cell precursor acute lymphoblastic leukemia (ALL) and B-cell precursor ALL with minimal residual disease. Similarly, initial marketing authorization for blinatumomab in the European Union was granted for the treatment of adults with Ph- R/R B-cell precursor ALL. The benefits of treating R/R B-cell precursor ALL patients with blinatumomab include increased overall survival, more favorable hematologic remission and molecular response rates, and a lower incidence rate of selected adverse events when compared with standard-of-care chemotherapy. The key risks associated with blinatumomab treatment include cytokine release syndrome, neurotoxicity, and medication errors. Here, we review the benefits and risks of blinatumomab treatment and describe how these risks can be mitigated.
Collapse
Affiliation(s)
| | - Janet L Franklin
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Victoria M Chia
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Deborah Arrindell
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA.
| | - William Kormany
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Jacqueline Wright
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Mandy Parson
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Hamid R Amouzadeh
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Jessica Choudhry
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - Guiandre Joseph
- Global Development, Observational Research, Global Patient Safety, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| |
Collapse
|
152
|
Adult T-cell acute lymphoblastic leukemias with IL7R pathway mutations are slow-responders who do not benefit from allogeneic stem-cell transplantation. Leukemia 2020; 34:1730-1740. [PMID: 31992840 DOI: 10.1038/s41375-019-0685-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/17/2019] [Accepted: 12/05/2019] [Indexed: 12/26/2022]
Abstract
The prognostic value of IL7-receptor pathway (IL7Rp) mutations in T-cell acute lymphoblastic leukemia (T-ALL) remains unclear. We performed a comprehensive study of 200 adult patients with T-ALL included in the GRAALL2003/2005 protocols to address the clinical significance of IL7Rp mutations. Next-generation sequencing of the IL7Rp (IL7R/JAK1/JAK3/STAT5B) revealed that IL7Rp mutations were frequent in adult T-ALL (28%) particularly in immature/early T-cell progenitor (ETP)-ALL. They were associated with mutations of NOTCH-pathway, PHF6, and PRC2 components but not with K/NRAS. IL7Rp mutated (IL7Rpmut) T-ALL were slow-responders, with a high rate of M2/M3 day-8 marrow compared with IL7Rp non-mutated (IL7RpWT) T-ALL (p = 0.002) and minimal residual disease positivity at 6-weeks (MRD1) (p = 0.008) but no difference in MRD2 positivity at 12-weeks. Despite this, no adverse prognosis was evidenced when censored for allogeneic hematopoietic stem cell transplantation (HSCT). In time-dependent analysis, HSCT did not benefit IL7Rpmut patients whereas it was of marked benefit to IL7RpWT cases. IL7Rp-mutations identify a subgroup of slow-responder T-ALLs which benefit from post-induction chemotherapy regimens but not from HSCT. Our data suggest that prior knowledge of the mutation status of IL7Rp may influence HSCT decision and help to guide therapy reduction.
Collapse
|
153
|
Gökbuget N, Dombret H, Giebel S, Brüggemann M, Doubek M, Foa R, Hoelzer D, Kim C, Martinelli G, Parovichnikova E, Maria Ribera J, Schoonen M, Tuglus C, Zugmaier G, Bassan R. Blinatumomab vs historic standard-of-care treatment for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukaemia. Eur J Haematol 2020; 104:299-309. [PMID: 31876009 PMCID: PMC7079006 DOI: 10.1111/ejh.13375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Survival outcomes from a single-arm phase 2 blinatumomab study in patients with minimal residual disease (MRD)-positive B-cell precursor (BCP)-acute lymphoblastic leukaemia (ALL) were compared with those receiving standard of care (SOC) in a historic data set. METHODS The primary analysis comprised adult Philadelphia chromosome (Ph)-negative patients in first complete haematologic remission (MRD ≥ 10-3 ). Relapse-free survival (RFS) and overall survival (OS) were compared between blinatumomab- and SOC-treatment groups. Baseline differences between groups were adjusted by propensity scores. RESULTS The primary analysis included 73 and 182 patients from the blinatumomab and historic data sets, respectively. When weighted by age to the blinatumomab-treatment group, median RFS was 7.8 months and median OS was 25.9 months in the SOC-treated group. In the blinatumomab study, median RFS was 35.2 months; median OS was not evaluable. Propensity score weighting achieved balance with seven baseline prognostic factors. With adjustment for haematopoietic stem cell transplantation (HSCT) status, a 50% reduction in risk of relapse or death was observed with blinatumomab vs SOC. Median RFS, unadjusted for HSCT status, was 35.2 months with blinatumomab and 8.3 months with SOC. CONCLUSIONS These analyses suggest that blinatumomab improves RFS, and possibly OS, in adults with MRD-positive Ph-negative BCP-ALL vs SOC.
Collapse
Affiliation(s)
| | - Hervé Dombret
- Hôpital Saint-Louis, University Paris Diderot, Paris, France
| | - Sebastian Giebel
- Maria Sklodowska-Curie Institute-Oncology Center, Gliwice, Poland
| | | | - Michael Doubek
- University Hospital and CEITEC Masaryk University, Brno, Czech Republic
| | - Robin Foa
- 'Sapienza' University of Rome, Rome, Italy
| | | | - Christopher Kim
- Center for Observational Research, Amgen Inc, Thousand Oaks, CA, USA
| | | | | | - Josep Maria Ribera
- ICO-Hospital Germans Trias i Pujol, Jose Carreras Research Institute, Barcelona, Spain
| | | | | | | | - Renato Bassan
- UOC Ematologia, Ospedale dell'Angelo, Mestre-Venezia, Italy
| |
Collapse
|
154
|
Shah S, Martin A, Turner M, Cong Z, Zaman F, Stein A. A systematic review of outcomes after stem cell transplantation in acute lymphoblastic leukemia with or without measurable residual disease. Leuk Lymphoma 2020; 61:1052-1062. [DOI: 10.1080/10428194.2019.1709834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
| | - Amber Martin
- EVIDERA, Evidence, Synthesis, Modeling, and Communications, Waltham, MA, USA
| | - Monica Turner
- EVIDERA, Evidence, Synthesis, Modeling, and Communications, Waltham, MA, USA
| | - Ze Cong
- Amgen Inc., Thousand Oaks, CA, USA
| | | | - Anthony Stein
- City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
155
|
Park E, Chen J, Moore A, Mangolini M, Santoro A, Boyd JR, Schjerven H, Ecker V, Buchner M, Williamson JC, Lehner PJ, Gasparoli L, Williams O, Bloehdorn J, Stilgenbauer S, Leitges M, Egle A, Schmidt-Supprian M, Frietze S, Ringshausen I. Stromal cell protein kinase C-β inhibition enhances chemosensitivity in B cell malignancies and overcomes drug resistance. Sci Transl Med 2020; 12:eaax9340. [PMID: 31941829 PMCID: PMC7116365 DOI: 10.1126/scitranslmed.aax9340] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/15/2019] [Indexed: 12/15/2022]
Abstract
Overcoming drug resistance remains a key challenge to cure patients with acute and chronic B cell malignancies. Here, we describe a stromal cell-autonomous signaling pathway, which contributes to drug resistance of malignant B cells. We show that protein kinase C (PKC)-β-dependent signals from bone marrow-derived stromal cells markedly decrease the efficacy of cytotoxic therapies. Conversely, small-molecule PKC-β inhibitors antagonize prosurvival signals from stromal cells and sensitize tumor cells to targeted and nontargeted chemotherapy, resulting in enhanced cytotoxicity and prolonged survival in vivo. Mechanistically, stromal PKC-β controls the expression of adhesion and matrix proteins, required for activation of phosphoinositide 3-kinases (PI3Ks) and the extracellular signal-regulated kinase (ERK)-mediated stabilization of B cell lymphoma-extra large (BCL-XL) in tumor cells. Central to the stroma-mediated drug resistance is the PKC-β-dependent activation of transcription factor EB, regulating lysosome biogenesis and plasma membrane integrity. Stroma-directed therapies, enabled by direct inhibition of PKC-β, enhance the effectiveness of many antileukemic therapies.
Collapse
Affiliation(s)
- Eugene Park
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Jingyu Chen
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Andrew Moore
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Maurizio Mangolini
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Antonella Santoro
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK
| | - Joseph R Boyd
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT 05405, USA
| | - Hilde Schjerven
- Department of Laboratory Medicine, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
- KG Jebsen Centre for B cell Malignancies, IMM, OUH, 0424 Oslo, Norway
| | - Veronika Ecker
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technische Universität München, 81675 Munich, Germany
| | - Maike Buchner
- Institut für Klinische Chemie und Pathobiochemie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technische Universität München, 81675 Munich, Germany
| | - James C Williamson
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Paul J Lehner
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Luca Gasparoli
- University College London (UCL) GOS-ICH, London WC1N 1EH, UK
| | - Owen Williams
- University College London (UCL) GOS-ICH, London WC1N 1EH, UK
| | - Johannes Bloehdorn
- Department of Internal Medicine III, University of Ulm, 89081 Ulm, Germany
| | | | - Michael Leitges
- Faculty of Medicine, Craig L. Dobbin Genetics Research Centre, Memorial University of Newfoundland, St. John's, Newfoundland A1B 3V6, Canada
| | - Alexander Egle
- IIIrd Medical Department with Hematology, Medical Oncology, Hemostaseology, Infectious Diseases and Rheumatology, Oncologic Center, Paracelsus Medical University, Cancer Cluster Salzburg, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute (SCRI) with Laboratory of Immunological and Molecular Cancer Research (LIMCR), 5020 Salzburg, Austria
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Marc Schmidt-Supprian
- German Cancer Consortium, DKFZ, 69120 Heidelberg, Germany
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, 81675 Munich, Germany
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute and Department of Haematology, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AH, UK.
| |
Collapse
|
156
|
Abstract
Bispecific T cell engagers are antibody constructs directed to a tumor-specific target on the one hand and to CD3-positive T cells on the other hand. Blinatumomab is a compound with specificity for the pan-B cell marker CD19. Clinical activity was tested in relapsed and refractory (R/R) non-Hodgkin's Lymphoma (NHL), R/R acute lymphoblastic leukemia (ALL), and ALL patients with minimal residual disease. Trials have already been started in de novo ALL. The most clinically relevant toxicities are neurologic events and cytokine release syndrome as with other T cell-activating therapies. The mechanisms of resistance are not fully understood. Higher leukemia load and later stage disease represent unfavorable factors. Besides, an upregulation of regulatory T cells and inhibitory molecules like PD-1/PD-L1 may have a role as the loss of target by several mechanisms. The future will show whether the use of bispecifics in ALL can change the standard treatment algorithms and whether bispecific T cell engagers will also be successfully used in other malignant entities.
Collapse
Affiliation(s)
- Nicola Gökbuget
- Department of Medicine II, University Hospital Goethe University, Theodor Stern Kai 7, 60590, Frankfurt, Germany.
| |
Collapse
|
157
|
Jabbour E, Gökbuget N, Advani A, Stelljes M, Stock W, Liedtke M, Martinelli G, O’Brien S, Wang T, Laird AD, Vandendries E, Neuhof A, Nguyen K, Dakappagari N, DeAngelo DJ, Kantarjian H. Impact of minimal residual disease status in patients with relapsed/refractory acute lymphoblastic leukemia treated with inotuzumab ozogamicin in the phase III INO-VATE trial. Leuk Res 2020; 88:106283. [DOI: 10.1016/j.leukres.2019.106283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/23/2019] [Indexed: 01/05/2023]
|
158
|
Ali S, Moreau A, Melchiorri D, Camarero J, Josephson F, Olimpier O, Bergh J, Karres D, Tzogani K, Gisselbrecht C, Pignatti F. Blinatumomab for Acute Lymphoblastic Leukemia: The First Bispecific T-Cell Engager Antibody to Be Approved by the EMA for Minimal Residual Disease. Oncologist 2019; 25:e709-e715. [PMID: 32297447 DOI: 10.1634/theoncologist.2019-0559] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/24/2019] [Indexed: 01/26/2023] Open
Abstract
On November 15, 2018, the Committee for Medicinal Products for Human Use (CHMP) recommended the extension of indication for blinatumomab to include the treatment of adults with minimal residual disease (MRD) positive B-cell precursor acute lymphoblastic leukemia (ALL). Blinatumomab was authorized to treat relapsed or refractory B-precursor ALL, and the change concerned an extension of use. On March 29, 2018, the U.S. Food and Drug Administration (FDA) granted accelerated approval to blinatumomab to treat both adults and children with B-cell precursor ALL who are in remission but still have MRD. On July 26, 2018, the CHMP had originally adopted a negative opinion on the extension. The reason for the initial refusal was that although blinatumomab helped to reduce the amount of residual cancer cells in many patients, there was no strong evidence that it led to improved survival. During the re-examination, the CHMP consulted the scientific advisory group. The CHMP agreed with the expert group's conclusion that, although there was no strong evidence of patients living longer, the available data from the main study (MT103-203) indicated a good durable response to blinatumomab, with an overall complete response rate for the primary endpoint full analysis set (defined as all subjects with an Ig or T-cell receptor polymerase chain reaction MRD assay with the minimum required sensitivity of 1 × 10-4 at central lab established at baseline [n = 113]) as 79.6% (90/113; 95% confidence interval, 71.0-86.6), with a median time to complete MRD response of 29.0 days (range, 5-71). Therefore, the CHMP concluded that the benefits of blinatumomab outweigh its risks and recommended granting the change to the marketing authorization. The Committee for Orphan Medicinal Products, following reassessment, considered that significant benefit continued to be met and recommended maintaining the orphan designation and thus 10 years market exclusivity (the Orphan Designation is a legal procedure that allows for the designation of a medicinal substance with therapeutic potential for a rare disease, before its first administration in humans or during its clinical development). The marketing authorization holder for this medicinal product is Amgen Europe B.V. IMPLICATIONS FOR PRACTICE: Immunotherapy with blinatumomab has excellent and sustainable results, offering new hope for patients with minimal residual disease-positive acute lymphoblastic leukemia, a disease with poor prognosis. New recommendations and change of practice for treatment of this patient group are detailed.
Collapse
Affiliation(s)
- Sahra Ali
- European Medicines Agency, Amsterdam, The Netherlands
| | - Alexandre Moreau
- French National Agency for Medicines and Health Products Safety, Saint-Denis Cedex, France
| | - Daniela Melchiorri
- Department of Physiology and Pharmacology, University of Rome, Sapienza, Rome, Italy
| | | | - Filip Josephson
- Medical Products Agency, Department of Efficacy and Safety 3, Uppsala, Sweden
| | | | - Jonas Bergh
- Department of Oncology-Pathology, Karolinska Institutet, BES, Cancer Theme, Karolinska University Hospital Bioclinicum, Solna, Sweden
| | | | | | | | | |
Collapse
|
159
|
Xue Y, Suo P, Huang X, Lu A, Wang Y, Zuo Y, Yan C, Wu J, Kong J, Zhang X, Chen Y, Jia Y, Liu K, Han W, Xu L, Zhang L, Cheng Y. Superior survival of unmanipulated haploidentical haematopoietic stem cell transplantation compared with intensive chemotherapy as post‐remission treatment for children with very high‐risk philadelphia chromosome negative B‐cell acute lymphoblastic leukaemia in first complete remission. Br J Haematol 2019; 188:757-767. [PMID: 31725190 DOI: 10.1111/bjh.16226] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/31/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Yu‐juan Xue
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Pan Suo
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Xiao‐jun Huang
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Ai‐dong Lu
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Yu Wang
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Ying‐xi Zuo
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Chen‐hua Yan
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Jun Wu
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Jun Kong
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Xiao‐hui Zhang
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Yu‐hong Chen
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Yue‐ping Jia
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Kai‐yan Liu
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Wei Han
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Lan‐ping Xu
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| | - Le‐ping Zhang
- Department of Paediatrics Peking University People’s Hospital Peking University Beijing China
| | - Yi‐fei Cheng
- Department of Haematology National Clinical Research Centre for Haematological Disease Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation Peking University People's Hospital Peking University Institute of Haematology Beijing China
| |
Collapse
|
160
|
Zhao J, Song Y, Liu D. Recent advances on blinatumomab for acute lymphoblastic leukemia. Exp Hematol Oncol 2019; 8:28. [PMID: 31709129 PMCID: PMC6833142 DOI: 10.1186/s40164-019-0152-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/31/2019] [Indexed: 01/14/2023] Open
Abstract
Although complete remission rate of B cell acute lymphoblastic leukemia (B-ALL) has improved significantly over the past few decades, patients with relapsed/refractory ALL still have dismal outcome. Tyrosine kinase inhibitors, antibody–drug conjugates and chimeric antigen receptor T cell therapy are changing the therapy landscape for B- ALL. Blinatumomab, a bi-specific T cell engager, has been approved for patients with relapsed/refractory and minimal residual disease positive B-ALL. This review summarized data from recent clinical trials of blinatumomab for B-ALL treatment.
Collapse
Affiliation(s)
- Juanjuan Zhao
- 1Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yongping Song
- 1Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Delong Liu
- 1Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China.,2Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595 USA
| |
Collapse
|
161
|
Gidman W, Shah S, Zhang L, McKendrick J, Cong Z, Cohan D, Ottmann O. Clinicians' Perspectives on Cure in Adult Patients with Acute Lymphoblastic Leukemia with Minimal Residual Disease: A Delphi Study. Adv Ther 2019; 36:3017-3029. [PMID: 31586302 PMCID: PMC6822800 DOI: 10.1007/s12325-019-01099-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Indexed: 02/07/2023]
Abstract
Hematologic complete remission (CR) is achievable for most adults with B cell precursor acute lymphoblastic leukemia (BCP-ALL). However, minimal residual disease (MRD) in patients with hematologic CR is associated with increased risk of relapse, shorter survival, and poorer transplantation outcomes. This study explored the concept of cure in adults with Philadelphia chromosome-negative (Ph−) BCP-ALL by MRD status at first hematologic CR (CR1) to inform evaluation of the clinical and economic benefits of new agents, where the concept of cure is important but long-term data are not available. The study used modified Delphi methodology involving clinicians experienced in the treatment of adult ALL. Participants completed a questionnaire, which was followed by country-specific panel discussions to discuss results and identify consensus on concepts and definitions. Clinicians from France (n = 4), Germany (n = 4), and the UK (n = 5) took part. Participants described cure in terms of the probability of future relapse. Relapse-free survival (RFS) was the preferred outcome measure to describe cure for the three patient groups considered (patients with MRD at CR1; patients who become negative for MRD after further treatment; patients who continue to have MRD). Consensus was reached on definitions of cure: that cure would begin to be considered at 3 years’ RFS and/or would be highly likely at 5 years’ RFS. Participants agreed that patients with MRD should usually undergo hematopoietic stem cell transplantation to have the best chance of survival; consensus was reached that alternatives are required when transplantation is not an option. Panels agreed that patients who achieve cure have a higher mortality rate and lower health-related quality of life than the general population. This study provides quantitative and qualitative information on the concept of cure in Ph− BCP-ALL in CR by MRD status applicable to interpreting the value of new therapies. Funding: Amgen. Plain Language Summary: Plain language summary available for this article.
Collapse
|
162
|
Subdural Hygroma: A Morbid Complication of Intrathecal Chemotherapy in a Patient with B-Cell Acute Lymphocytic Leukemia. Case Rep Hematol 2019; 2019:3494056. [PMID: 31662916 PMCID: PMC6791203 DOI: 10.1155/2019/3494056] [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: 05/04/2019] [Accepted: 08/28/2019] [Indexed: 11/17/2022] Open
Abstract
A subdural hygroma is an accumulation of cerebrospinal fluid in the subdural space that may occur secondary to trauma and surgery, or for iatrogenic reasons, such as a lumbar puncture. Lumbar puncture is a procedure used commonly for intrathecal chemotherapy for patients with B-cell acute lymphocytic leukemia (B-ALL) though subdural hygroma is a very rare complication. We present a case of a fatal, refractory subdural hygroma in a patient with B-ALL.
Collapse
|
163
|
Czyz A, Nagler A. The Role of Measurable Residual Disease (MRD) in Hematopoietic Stem Cell Transplantation for Hematological Malignancies Focusing on Acute Leukemia. Int J Mol Sci 2019; 20:ijms20215362. [PMID: 31661875 PMCID: PMC6862140 DOI: 10.3390/ijms20215362] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 01/17/2023] Open
Abstract
The significance of measurable residual disease (MRD) in hematopoietic stem cell transplantation (HSCT) is well recognized in different hematological malignancies, but the evidence indicate that pre-transplant MRD status is of particular importance in acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). In ALL, inadequate response at the level of MRD is a commonly accepted risk factor for relapse and thus an indication for allogeneic HSCT. Similarly, growing evidence from the literature strongly suggest that MRD detected by multiparameter flow cytometry or molecular techniques should be also used for risk stratification in AML at the time of HSCT. Despite the well-defined association of MRD and outcomes of HSCT in acute leukemias, there are still many open issues such as the role of additional pre-transplant consolidation for MRD eradication, the ability of HSCT to overcome negative influence of MRD positivity on survival, the impact of conditioning regimen intensity on MRD clearance post HSCT, and transplantation outcomes or the selection of optimal donor with regards to MRD status. In addition, the role of MRD assessment in guiding post-transplant maintenance treatment should also be addressed in prospective trials. These open issues mostly awaiting further clinical studies will be discussed in our current review.
Collapse
Affiliation(s)
- Anna Czyz
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Ludwik Pasteur 4, 50-367 Wroclaw, Poland.
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, Derech Sheba 2, 52-621 Ramat Gan, Israel.
| |
Collapse
|
164
|
Gavrilina OA, Troitskaya VV, Baskhaeva GA, Lukyanova IA, Zarubina KI, Parovichnikova EN. APPLICATION OF POSITRON EMISSION TOMOGRAPHY / COMPUTER TOMOGRAPHY FOR EVALUATING THE RESPONSE TO CHEMOTHERAPY IN PATIENTS WITH ACUTE LYMPHOBLASTIC LEUKEMIA / LYMPHOBLASTIC LYMPHOMA. RUSSIAN JOURNAL OF HEMATOLOGY AND TRANSFUSIOLOGY 2019. [DOI: 10.35754/0234-5730-2019-64-2-138-149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction.No recommendations are currently available on the use of positron emission tomography / computer tomography (PET/CT) for evaluating the response to chemotherapy in patients with acute lymphoblastic leukosis / lymphoblastic lymphoma (ALL/LBL).Aim. The aim of this research was to study the ability of tumour cells to accumulate radiopharmaceuticals during PET/CT in patients with ALL/LBL, as well as to evaluate the prognostic value of PET/CT results performed after completion of consolidation therapy with/without autologous hematopoietic blood stem cell transplantation (auto-HSCT) in patients with Ph-negative ALL/LLL who underwent therapy according to the protocols of a Russian research group ALL-2009/ALL-2016.Materials and methods.PET/CT was performed in 3 patients with various variants of a newly diagnosed ALL before the onset of therapy and after the completion of induction therapy. In 10 patients with Ph-negative ALL/LLL, a PET study was performed after consolidation had been completed according to the ALL-2009/ALL-2016 protocol.Results. The results of PET/CT in 3 patients with different variants of newly detected ALL/LBL were analysed. All patients showed a metabolic activity of 18F-FDG in all morphologically and immunohistochemically (immunophenotypically) confirmed lesions. An analysis of the PET/CT results in 10 patients with Ph-negative ALL/LBL after completion of consolidation therapy with/without auto-HSCT showed that all patients had achieved a PET-negative disease remission. With a median follow-up of 20.5 months (from 15 to 44 months), only one out of 10 patients demonstrated isolated neurorecurrence 10 months after achieving remission. The remaining 9 patients, under a median relapse-free survival rate of 19 months (from 14 to 43 months), demonstrated complete clinical and hematological remission.Conclusion. Specific medullary and extramedullary lesions in ALL/LBL are capable of accumulating 18F-FDG in PET, which allows the method under study to be used for evaluating the completeness of remission in extramedullary lesions. The prognostic feasibility of PET/CT under the involvement of the central nervous system remains to be studied.
Collapse
|
165
|
Frisch A, Ofran Y. How I diagnose and manage Philadelphia chromosome-like acute lymphoblastic leukemia. Haematologica 2019; 104:2135-2143. [PMID: 31582548 PMCID: PMC6821607 DOI: 10.3324/haematol.2018.207506] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/03/2019] [Indexed: 01/31/2023] Open
Abstract
Advances in our understanding of mechanisms of leukemogenesis and driver mutations in acute lymphoblastic leukemia (ALL) lead to a more precise and informative sub-classification, mainly of B-cell ALL. In parallel, in recent years, novel agents have been approved for the therapy of B-cell ALL, and many others are in active clinical research. Among the newly recognized disease subtypes, Philadelphia-chromosome-like ALL is the most heterogeneous and thus, diagnostically challenging. Given that this subtype of B-cell ALL is associated with a poorer prognosis, improvement of available therapeutic approaches and protocols is a burning issue. Herein, we summarize, in a clinically relevant manner, up-to-date information regarding diagnostic strategies developed for the identification of patients with Philadelphia-chromosome-like ALL. Common therapeutic dilemmas, presented as several case scenarios, are also discussed. It is currently acceptable that patients with B-cell ALL, treated with an aim of cure, irrespective of their age, be evaluated for a Philadelphia-chromosome-like signature as early as possible. Following Philadelphia-chromosome-like recognition, a higher risk of resistance or relapse must be realized and treatment should be modified based on the patient’s specific genetic driver and clinical features. However, while active targeted therapeutic options are limited, there is much more to do than just prescribe a matched inhibitor to the identified mutated driver genes. In this review, we present a comprehensive evidence-based approach to the diagnosis and management of Philadelphia-chromosome-like ALL at different time-points during the disease course.
Collapse
Affiliation(s)
- Avraham Frisch
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa
| | - Yishai Ofran
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa .,Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
166
|
Bassan R, Brüggemann M, Radcliffe HS, Hartfield E, Kreuzbauer G, Wetten S. A systematic literature review and meta-analysis of minimal residual disease as a prognostic indicator in adult B-cell acute lymphoblastic leukemia. Haematologica 2019; 104:2028-2039. [PMID: 30890593 PMCID: PMC6886415 DOI: 10.3324/haematol.2018.201053] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 03/14/2019] [Indexed: 11/09/2022] Open
Abstract
Minimal (or 'measurable') residual disease in acute lymphoblastic leukemia appears to be a prognostic indicator, with potential value in informing individualized treatment decisions. Complete understanding of the strength of the association between minimal residual disease and long-term outcomes is, however, lacking. A systematic literature review and meta-analysis were performed to elucidate the clinical significance of minimal residual disease with respect to relapse-free survival and overall survival in precursor B-cell acute lymphoblastic leukemia. A total of 23 articles and abstracts, most published between 2012 and 2016, were identified for inclusion in the primary meta-analysis. Typically, patients were in their first complete remission at the time of minimal residual disease assessment; in two studies, all patients were in their second, or later, complete remission. The primary analysis revealed improved relapse-free survival across all studies for patients who achieved minimal residual disease negativity (random effects hazard ratio, 2.34; 95% confidence interval, 1.91-2.86). Improved overall survival for patients who achieved minimal residual disease negativity was also observed (hazard ratio, 2.19; 95% confidence interval, 1.63-2.94). There was no observed difference in the impact of minimal residual disease status in subgroups based on disease stage, minimal residual disease sensitivity threshold level, Philadelphia chromosome status, histological phenotype, risk group, minimal residual disease testing location, minimal residual disease timing after induction, or minimal residual disease detection method. Despite heterogeneity in study design and patient populations between the contributing studies, these data provide a compelling argument for minimal residual disease as a clinical tool for assessing prognosis and guiding treatment decisions in precursor B-cell acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Renato Bassan
- Complex Operative Unit of Haematology, dell'Angelo Hospital and Santissimi Giovanni and Paolo Hospital, Mestre and Venice, Italy
| | - Monika Brüggemann
- Department of Medicine II, Schleswig-Holstein University Hospital, Kiel, Germany
| | | | | | | | | |
Collapse
|
167
|
DeFilipp Z, Advani AS, Bachanova V, Cassaday RD, Deangelo DJ, Kebriaei P, Rowe JM, Seftel MD, Stock W, Tallman MS, Fanning S, Inamoto Y, Kansagra A, Johnston L, Nagler A, Sauter CS, Savani BN, Perales MA, Carpenter PA, Larson RA, Weisdorf D. Hematopoietic Cell Transplantation in the Treatment of Adult Acute Lymphoblastic Leukemia: Updated 2019 Evidence-Based Review from the American Society for Transplantation and Cellular Therapy. Biol Blood Marrow Transplant 2019; 25:2113-2123. [PMID: 31446198 DOI: 10.1016/j.bbmt.2019.08.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 01/05/2023]
Abstract
The role of hematopoietic cell transplantation (HCT) in adults with acute lymphoblastic leukemia (ALL) is reviewed and critically evaluated in this systematic evidence-based review. Specific criteria were used for searching the published literature and for grading the quality and strength of the evidence and the strength of the recommendations. A panel of ALL experts developed consensus on the treatment recommendations based on the evidence. Allogeneic HCT offers a survival benefit in selected patients with ALL, and this review summarizes the standard indications as well as the areas of controversy. There is now greater experience with pediatric-inspired chemotherapy regimens that has transformed upfront therapy for adult ALL, resulting in higher remission rates and overall survival. This in turn has increased the equipoise around decision making for ALL in first complete remission (CR1) when there is no measurable residual disease (MRD) at the end of induction and/or consolidation. Randomized studies are needed for adults with ALL to compare allogeneic HCT in CR1 with pediatric-inspired chemotherapy alone. Indications for transplantation in the evolving landscape of MRD assessments and novel targeted and immune therapeutics remain important areas of investigation.
Collapse
Affiliation(s)
- Zachariah DeFilipp
- Blood and Marrow Transplant Program, Massachusetts General Hospital, Boston, MA.
| | - Anjali S Advani
- Department of Medical Oncology & Hematology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Veronika Bachanova
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN
| | - Ryan D Cassaday
- Division of Clinical Research, Fred Hutchinson Cancer Research Center and Department of Medicine, University of Washington, Seattle, WA, USA
| | - Daniel J Deangelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jacob M Rowe
- Department of Hematology, Shaare Zedek Medical Center, Jerusalem, Israel
| | | | - Wendy Stock
- Department of Hematology and Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Martin S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY
| | - Suzanne Fanning
- Hematology and Medical Oncology, Greenville Health System Cancer Institute, Greenville, SC
| | - Yoshihiro Inamoto
- Division of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Ankit Kansagra
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Laura Johnston
- Department of Medicine, Stanford University Medical Center, Stanford, CA
| | - Arnon Nagler
- Hematology and Bone Marrow Transplantation Division, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Craig S Sauter
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY
| | - Paul A Carpenter
- Division of Clinical Research, Fred Hutchinson Cancer Research Center and Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Richard A Larson
- Department of Hematology and Oncology, Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Daniel Weisdorf
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN
| |
Collapse
|
168
|
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: 79] [Impact Index Per Article: 13.2] [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
|
169
|
Paul S, Rausch CR, Welch MA, Kantarjian HM, Jabbour EJ. SOHO State of the Art Update and Next Questions: Advances in the Treatment of Adult Acute Lymphoblastic Leukemia. CLINICAL LYMPHOMA MYELOMA AND LEUKEMIA 2019; 19:471-479. [DOI: 10.1016/j.clml.2019.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022]
|
170
|
Long-term outcomes of modified BFM-95 regimen in adults with newly diagnosed standard-risk acute lymphoblastic leukemia: a retrospective single-center study. Int J Hematol 2019; 110:458-465. [PMID: 31321731 DOI: 10.1007/s12185-019-02703-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
The standard treatment for adult acute lymphoblastic leukemia (ALL) is undefined. Patients with newly diagnosed standard-risk ALL at a single institution were retrospectively analyzed. From 2010 to 2017, 46 patients were treated using the modified Berlin-Frankfurt-Münster (BFM)-ALL-95 regimen. Hematologic and molecular complete remission (CR) rates of 91.3% and 76.1% were achieved. The 5-year event-free survival (EFS) and overall survival (OS) rates for all patients in the cohort were 58.0% (95% confidence interval, 42.1-73.9%) and 66.7% (95% CI, 51.4-82.0%), respectively. No patient presented with central nervous system involvement after CR in this study. This condition could be related to four doses of high-dose methotrexate (MTX) every 3 months during the maintenance phase. Multivariate analysis revealed that minimal residual disease positive and time interval between induction IA and Protocol M of more than 70 days were independent adverse factors for EFS and OS. One or more instances of grade 4 myelosuppression occurred during induction therapy. Nonhematological side effects were mild. No toxicity-related deaths were observed in the entire cohort. The data indicated that the modified regimen is well tolerated and can produce the promising outcomes in Chinese adults with standard-risk ALL.
Collapse
|
171
|
Li ZR, Zhao T, Liu YR, Wang YZ, Xu LP, Zhang XH, Wang Y, Jiang H, Chen YY, Chen H, Han W, Yan CH, Wang J, Jia JS, Huang XJ, Jiang Q. [Minimal residual disease in adults with Philadelphia chromosome negative acute lymphoblastic leukemia in high-risk]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:554-560. [PMID: 32397017 PMCID: PMC7364904 DOI: 10.3760/cma.j.issn.0253-2727.2019.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Indexed: 11/21/2022]
Abstract
Objective: To explore the significance of minimal residual disease (MRD) in predicting prognosis and guiding therapy of adults with Philadelphia-chromosome negative acute lymphoblastic leukemia (Ph(-) ALL) in high-risk. Methods: Data of newly diagnosed adults with Ph(-) ALL in high-risk who achieved CR were reviewed. Variables associated with outcome were identified by COX regression model and Landmark analysis. Results: A total of 177 patients, 99 (56%) cases male with a median age of 40 years (range, 16-65 years) were included in this study. Of them, 95 (54%) patients received allo-HSCT in CR(1). Multivariate analyses showed that MRD negativity after the first cycle of consolidation (HR=0.52, 95%CI 0.30-0.89, P=0.017) and achieving CR within 4 weeks (HR=0.43, 95%CI 0.24-0.79, P=0.006) were the factors significantly-associated with longer DFS, and allo-HSCT was associated with both longer DFS (HR=0.13, 95%CI 0.08-0.22, P<0.001) and OS (HR=0.24, 95%CI 0.15-0.41, P<0.001) . Landmark analysis was performed on 121 patients, of 85 patients achieving MRD negativity after the first cycle of consolidation, multivariate analyses showed that MRD negativity after the third cycle of consolidation was significantly-associated with longer DFS (HR=0.18, 95%CI 0.05-0.64, P=0.008) and OS (HR=0.14, 95%CI 0.04-0.50, P=0.003) . For the patients achieving MRD negativity after both the first and the third cycles of consolidation, the 3-year DFS rate in the allo-HSCT cohort had a higher trend compared with that in the chemotherapy cohort (75.2% vs 51.3%, P=0.082) , however, the 3-year OS rates in the 2 cohorts were similar (72.7% vs 68.7%, P=0.992) . In those with MRD positivity after the first and/or the third cycle of consolidation, 3-year DFS (64.8% vs 33.3%, P=0.006) and OS (77.0% vs 33.3%, P=0.028) rates in the allo-HSCT cohort were significantly higher than those in the chemotherapy cohort, and similar to those in the cohort achieving MRD negativity after both the first and the third cycles of consolidation and receiving allo-HSCT. Conclusions: MRD negativity after the first cycle of consolidation was a predictor for better outcome in adults with Ph(-) ALL in high-risk. The survival advantage of the allo-HSCT cohort was not pronounced compared with that in the chemotherapy cohort even in those with high-risk features but achieving MDR negativity after both the first and third cycles of consolidation. However, allo-HSCT could be a good option for the patients with MRD positivity after the first and/or the third cycle of consolidation.
Collapse
Affiliation(s)
- Z R Li
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing 100044, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Nagafuji K, Miyamoto T, Eto T, Ogawa R, Okumura H, Takase K, Kawano N, Miyazaki Y, Fujisaki T, Wake A, Ohno Y, Kurokawa T, Kamimura T, Takamatsu Y, Yokota S, Akashi K. Prospective evaluation of minimal residual disease monitoring to predict prognosis of adult patients with Ph-negative acute lymphoblastic leukemia. Eur J Haematol 2019; 103:164-171. [PMID: 31132205 DOI: 10.1111/ejh.13268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVE We investigated whether minimal residual disease (MRD) status in adult patients with Philadelphia chromosome (Ph)-negative acute lymphoblastic leukemia (ALL) is useful for decision on clinical indications for allogeneic hematopoietic stem cell transplantation (HSCT). METHODS We prospectively monitored MRD after induction and consolidation therapy in adult patients with Ph-negative ALL. RESULTS Among 103 adult ALL patients enrolled, 59 were Ph-negative, and MRD status was assessed in 51 patients. The probability of 3-year overall survival (OS) and disease-free survival (DFS) was 69% (95%CI 54-80) and 50% (95%CI 36-63), respectively. Patients who were MRD-negative after induction therapy (n = 15) had a significantly better 3-year DFS compared with those who were MRD-positive (n = 30; 73% vs 41%, P = 0.018). Patients who were MRD-positive after induction but became MRD-negative after consolidation chemotherapy C in the first course (n = 11) showed a significantly worse 3-year DFS compared with patients who were MRD-negative after induction chemotherapy A in the first course (45% vs 73%, P = 0.025). CONCLUSIONS These results indicate that DFS of about 70% can be expected in MRD-negative patients after induction therapy, and the patients did not benefit from HSCT in 1CR. This study was registered with the UMIN Clinical Trials Registry (UMIN-CTR), number UMIN000001519.
Collapse
Affiliation(s)
- Koji Nagafuji
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Toshihiro Miyamoto
- Department of Hematology/Oncology, Kyushu University Hospital, Fukuoka, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Ryosuke Ogawa
- Department of Hematology and Oncology, Japan Community Healthcare Organization Kyushu Hospital, Kitakyushu, Japan
| | - Hirokazu Okumura
- Department of Hematology, Toyama Prefectural Central Hospital, Toyama, Japan
| | - Ken Takase
- Department of Hematology and Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Noriaki Kawano
- Department of Hematology, Miyazaki Prefectural Hospital, Miyazaki, Japan
| | | | - Tomoaki Fujisaki
- Department of Hematology, Matsuyama Red Cross Hospital, Matsuyama, Japan
| | - Atsushi Wake
- Department of Hematology, Toranomon Hospital Kajigaya, Kawasaki, Japan
| | - Yuju Ohno
- Department of Hematology, Kitakyushu Municipal Medical Center, Kitakyushu, Japan
| | - Toshiro Kurokawa
- Department of Hematology, Toyama Red Cross Hospital, Toyama, Japan
| | - Tomohiko Kamimura
- Department of Hematology, HaraSanshin General Hospital, Fukuoka, Japan
| | - Yasushi Takamatsu
- Division of Medical Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | | | - Koichi Akashi
- Department of Hematology/Oncology, Kyushu University Hospital, Fukuoka, Japan
| |
Collapse
|
173
|
Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. Next-Generation Sequencing in Acute Lymphoblastic Leukemia. Int J Mol Sci 2019; 20:ijms20122929. [PMID: 31208040 PMCID: PMC6627957 DOI: 10.3390/ijms20122929] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/04/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common childhood cancer and accounts for about a quarter of adult acute leukemias, and features different outcomes depending on the age of onset. Improvements in ALL genomic analysis achieved thanks to the implementation of next-generation sequencing (NGS) have led to the recent discovery of several novel molecular entities and to a deeper understanding of the existing ones. The purpose of our review is to report the most recent discoveries obtained by NGS studies for ALL diagnosis, risk stratification, and treatment planning. We also report the first efforts at NGS use for minimal residual disease (MRD) assessment, and early studies on the application of third generation sequencing in cancer research. Lastly, we consider the need for the integration of NGS analyses in clinical practice for genomic patients profiling from the personalized medicine perspective.
Collapse
Affiliation(s)
- Nicoletta Coccaro
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Luisa Anelli
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Antonella Zagaria
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Giorgina Specchia
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy.
| |
Collapse
|
174
|
Shimizu H, Doki N, Kanamori H, Sakura T, Mori T, Machida S, Takahashi S, Ohwada C, Fujisawa S, Yano S, Hagihara M, Kanda Y, Onoda M, Gotoh M, Kako S, Taguchi J, Usuki K, Kawai N, Aotsuka N, Okamoto S. Prognostic impact of cytogenetic abnormalities in adult patients with Philadelphia chromosome-negative ALL who underwent an allogeneic transplant. Bone Marrow Transplant 2019; 54:2020-2026. [PMID: 31186516 DOI: 10.1038/s41409-019-0585-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/15/2019] [Accepted: 05/16/2019] [Indexed: 12/16/2022]
Abstract
Although cytogenetic abnormalities at diagnosis are recognized as an important prognostic factor in patients with Philadelphia chromosome (Ph)-negative acute lymphoblastic leukemia (ALL), the prognostic impact has not been evaluated in allogeneic stem cell transplant (allo-SCT) recipients. Thus, we assessed 373 Ph-negative ALL patients who underwent allo-SCT. The high-risk (HR) group included those with t(4;11), t(8;14), low hypodiploidy, and complex karyotype, and the standard risk (SR) group included all other karyotypes. Among the 204 patients who underwent a transplant during the first remission (167 in the SR group and 37 in the HR group), the overall survival (OS) rates were similar between these groups (64.1% vs. 80.0% at 5 years, respectively; p = 0.12). Conversely, among the 106 patients who underwent a transplant while not in remission (84 in the SR group and 22 in the HR group), patients in the SR group showed a significantly superior OS rate compared to the HR group (15.4% vs. 4.5% at 5 years, respectively; p = 0.022). These results suggested that treatment outcomes of Ph-negative ALL patients with HR cytogenetic abnormalities may improve following allo-SCT, especially in the first remission. Innovative transplant approaches are warranted in patients who are not in remission.
Collapse
Affiliation(s)
| | - Noriko Doki
- Division of Hematology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Heiwa Kanamori
- Department of Hematology, Kanagawa Cancer Center, Yokohama, Japan
| | - Toru Sakura
- Leukemia Research Center, Saiseikai Maebashi Hospital, Gunma, Japan
| | - Takehiko Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shinichiro Machida
- Department of Hematology and Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Satoshi Takahashi
- Division of Molecular Therapy, The Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Chikako Ohwada
- Division of Hematology, Department of Clinical Cell Biology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shin Fujisawa
- Department of Hematology, Yokohama City University Medical Center, Yokohama, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Maki Hagihara
- Department of Hematology and Clinical Immunology, Yokohama City University Hospital, Yokohama, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Masahiro Onoda
- Department of Internal Medicine, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Moritaka Gotoh
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Shinichi Kako
- Division of Hematology, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Jun Taguchi
- Department of Hematology, Shizuoka Red Cross Hospital, Shizuoka, Japan
| | - Kensuke Usuki
- Division of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Nobutaka Kawai
- Department of Hematology, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Nobuyuki Aotsuka
- Department of Hematology, Narita Red Cross Hospital, Narita, Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
175
|
Outcomes of adult acute lymphoblastic leukemia in the era of pediatric-inspired regimens: a single-center experience. Int J Hematol 2019; 110:295-305. [DOI: 10.1007/s12185-019-02678-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 01/23/2023]
|
176
|
Modvig S, Madsen HO, Siitonen SM, Rosthøj S, Tierens A, Juvonen V, Osnes LTN, Vålerhaugen H, Hultdin M, Thörn I, Matuzeviciene R, Stoskus M, Marincevic M, Fogelstrand L, Lilleorg A, Toft N, Jónsson OG, Pruunsild K, Vaitkeviciene G, Vettenranta K, Lund B, Abrahamsson J, Schmiegelow K, Marquart HV. Minimal residual disease quantification by flow cytometry provides reliable risk stratification in T-cell acute lymphoblastic leukemia. Leukemia 2019; 33:1324-1336. [PMID: 30552401 DOI: 10.1038/s41375-018-0307-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 01/22/2023]
Abstract
Minimal residual disease (MRD) measured by PCR of clonal IgH/TCR rearrangements predicts relapse in T-cell acute lymphoblastic leukemia (T-ALL) and serves as risk stratification tool. Since 10% of patients have no suitable PCR-marker, we evaluated flowcytometry (FCM)-based MRD for risk stratification. We included 274 T-ALL patients treated in the NOPHO-ALL2008 protocol. MRD was measured by six-color FCM and real-time quantitative PCR. Day 29 PCR-MRD (cut-off 10-3) was used for risk stratification. At diagnosis, 93% had an FCM-marker for MRD monitoring, 84% a PCR-marker, and 99.3% (272/274) had a marker when combining the two. Adjusted for age and WBC, the hazard ratio for relapse was 3.55 (95% CI 1.4-9.0, p = 0.008) for day 29 FCM-MRD ≥ 10-3 and 5.6 (95% CI 2.0-16, p = 0.001) for PCR-MRD ≥ 10-3 compared with MRD < 10-3. Patients stratified to intermediate-risk therapy on day 29 with MRD 10-4-<10-3 had a 5-year event-free survival similar to intermediate-risk patients with MRD < 10-4 or undetectable, regardless of method for monitoring. Patients with day 15 FCM-MRD < 10-4 had a cumulative incidence of relapse of 2.3% (95% CI 0-6.8, n = 59). Thus, FCM-MRD allows early identification of patients eligible for reduced intensity therapy, but this needs further studies. In conclusion, FCM-MRD provides reliable risk prediction for T-ALL and can be used for stratification when no PCR-marker is available.
Collapse
Affiliation(s)
- S Modvig
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - H O Madsen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - S M Siitonen
- Helsinki University Ctrl. Hospital, Helsinki, Finland
| | - S Rosthøj
- Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - A Tierens
- Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Hospital of Oslo, Oslo, Norway
| | - V Juvonen
- Department of Clinical Chemistry and Laboratory Division, University of Turku and Turku University Hospital, Turku, Finland
| | - L T N Osnes
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - H Vålerhaugen
- Department of Pathology, Laboratory of Molecular Pathology, Oslo University Hospital, Oslo, Norway
| | - M Hultdin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - I Thörn
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - R Matuzeviciene
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- Centre of Laboratory Medicine, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - M Stoskus
- Hematology, Oncology and Transfusion Medicine Centre, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - M Marincevic
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - L Fogelstrand
- Department of Clinical Chemistry, Sahlgrenska University Hospital, and Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - A Lilleorg
- Department of Clinical Immunology, North Estonia Medical Centre, Tallinn, Estonia
| | - N Toft
- Department of Hematology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - O G Jónsson
- Children's Hospital, Landspitali University Hospital, Reykjavik, Iceland
| | - K Pruunsild
- Tallinn Children's Hospital, Tallinn, Estonia
| | - G Vaitkeviciene
- Children's Hospital, Affiliate of Vilnius University Hospital Santariskiu Klinikos, Vilnius, Lithuania
| | - K Vettenranta
- Department of Pediatrics, Helsinki University Children's Hospital and University of Helsinki, Helsinki, Finland
| | - B Lund
- Department of Pediatrics, St. Olavs University Hospital and Department of Clinical and Molecular Medicine, NTNU, Trondheim, Norway
| | - J Abrahamsson
- Institution of Clinical Sciences, Department of Pediatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - K Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- The Institute of Clinical medicine, The Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - H V Marquart
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
177
|
Maffini E, Saraceni F, Lanza F. Treatment of Adult Patients with Relapsed/Refractory B-Cell Philadelphia-Negative Acute Lymphoblastic Leukemia. Clin Hematol Int 2019; 1:85-93. [PMID: 34595415 PMCID: PMC8432388 DOI: 10.2991/chi.d.190503.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/26/2019] [Indexed: 12/27/2022] Open
Abstract
The majority of adult patients affected by B-cell acute lymphoblastic leukemia (B-ALL) will relapse after an initial response, while approximately 20% will display primary resistant disease. Patients suffering from relapsed/refractory B-ALL have a very poor outcome. Allogeneic hematopoietic cell transplantation (HCT) still represents the only curative approach, but is not so frequently feasible, because of patient's fitness, donor availability, and the ability to achieve a remission prior to HCT. The estimated remission rates with conventional cytotoxic agents are around 30%, but they are short-lived. These disappointing results led to the introduction of new immunologic-based treatments-blinatumomab and inotuzumab. They produced a substantial improvement in terms of response rates, with the ability, in most cases, to induce a minimal residual disease (MRD)-negative status. Similarly, T cells engineered to express a CD19-specific chimeric antigen receptor (CAR-T) have yielded sensational results among patients with relapsed/refractory B-ALL, with unexpectedly high MRD-negative complete remissions rates. However, the first studies looking at long-term outcomes after CAR-T infusions told us that a significant fraction of such responses are not durable, and may benefit from a consolidation approach such as an allogeneic HCT.
Collapse
Affiliation(s)
- Enrico Maffini
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Saraceni
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| | - Francesco Lanza
- Hematology Unit, Romagna Transplant Network, Hospital of Ravenna, Viale Randi n. 5, 48121 Ravenna, Italy
| |
Collapse
|
178
|
Giebel S, Marks DI, Boissel N, Baron F, Chiaretti S, Ciceri F, Cornelissen JJ, Doubek M, Esteve J, Fielding A, Foa R, Gorin NC, Gökbuget N, Hallböök H, Hoelzer D, Paravichnikova E, Ribera JM, Savani B, Rijneveld AW, Schmid C, Wartiovaara-Kautto U, Mohty M, Nagler A, Dombret H. Hematopoietic stem cell transplantation for adults with Philadelphia chromosome-negative acute lymphoblastic leukemia in first remission: a position statement of the European Working Group for Adult Acute Lymphoblastic Leukemia (EWALL) and the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation (EBMT). Bone Marrow Transplant 2019; 54:798-809. [PMID: 30385870 DOI: 10.1038/s41409-018-0373-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 12/24/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) in first complete remission is a standard of care for adult patients with Philadelphia chromosome (Ph)-negative acute lymphoblastic leukemia (ALL) and high risk of relapse. However, the stratification systems vary among study groups. Inadequate response at the level of minimal residual disease is the most commonly accepted factor indicating the need for alloHSCT. In this consensus paper on behalf of the European Working Group for Adult Acute Lymphoblastic Leukemia and the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation, we summarize available evidence and reflect current clinical practice in major European study groups regarding both indications for HSCT and particular aspects of the procedure including the choice of donor, source of stem cells and conditioning. Finally, we propose recommendations for daily clinical practice as well as for planning of prospective trials.
Collapse
Affiliation(s)
- Sebastian Giebel
- Maria Sklodowska-Curie Institute-Cancer Center, Gliwice Branch, Gliwice, Poland.
| | - David I Marks
- University Hospitals Bristol National Health Service Foundation Trust, Bristol, UK
| | | | | | | | | | - Jan J Cornelissen
- Erasmus MC Cancer Institute University Medical Center, Rotterdam, The Netherlands
| | | | | | - Adele Fielding
- North London Cancer Network, Univ. College London Hosp, London, UK
| | | | - Norbert-Claude Gorin
- EBMT Acute Leukemia Working Party Office, Paris, France
- Hospital Saint-Antoine, Paris, France
| | - Nicola Gökbuget
- Maria Sklodowska-Curie Institute-Cancer Center, Gliwice Branch, Gliwice, Poland
- Hopital St. Louis, Paris, France
| | | | - Dieter Hoelzer
- University Hospital, Goethe University, Frankfurt, Germany
| | - Elena Paravichnikova
- FGBU Hematology Research Center, Russia Federation Ministry of Public Health, Moscow, Russia
| | - Josep-Maria Ribera
- ICO-Hospital Germans Trias I Pujol, Jose Carreras Research Institute, Badalona, Spain
| | - Bipin Savani
- Vanderbilt University Medical Center, Nashville, USA
| | - Anita W Rijneveld
- Erasmus MC Cancer Institute University Medical Center, Rotterdam, The Netherlands
| | - Christoph Schmid
- Klinikum Augsburg, Ludwig-Maximilians-Universitaet, Munich-Augsburg, Germany
| | | | - Mohamad Mohty
- North London Cancer Network, Univ. College London Hosp, London, UK
- EBMT Acute Leukemia Working Party Office, Paris, France
| | - Arnon Nagler
- North London Cancer Network, Univ. College London Hosp, London, UK
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | | |
Collapse
|
179
|
Rafei H, Kantarjian HM, Jabbour EJ. Recent advances in the treatment of acute lymphoblastic leukemia. Leuk Lymphoma 2019; 60:2606-2621. [DOI: 10.1080/10428194.2019.1605071] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Hind Rafei
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M. Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias J. Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
180
|
Foster LH, Lum LG. Treatment of hematological malignancies with T cell redirected bispecific antibodies: current status and future needs. Expert Opin Biol Ther 2019; 19:707-720. [PMID: 31081696 DOI: 10.1080/14712598.2019.1604672] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Enthusiasm for developing therapeutic bispecific antibodies (BsAbs) for cancer applications has become intense in the past decade facilitated by advances in molecular biology, hybridoma technology, and protein engineering. The central strategy in BsAb engineering is to combine the specificities directed at effector cells, and at a tumor target associated antigen (TAA) into a single construct. AREAS COVERED This article highlights the clinical use of BsAbs to target effector cells to multiple myeloma (MM), non-Hodgkin lymphoma (NHL), acute myeloid leukemia (AML), and acute lymphoblastic leukemia (ALL). We discuss the successes, challenges, and future strategies. Secondary literature search was performed using Pubmed, clinicaltrials.gov and non-proprietary internet search engines. EXPERT OPINION The use of BsAb constructs to target hematologic malignancies has achieved limited success to date. There continues to be a high level of enthusiasm for developing and applying new constructs to overcome the challenges in engineering and clinical application for hematologic malignancies.
Collapse
Affiliation(s)
- Laahn H Foster
- a Division of Hematology/Oncology , University of Virginia , Charlottesville , VA , USA
| | - Lawrence G Lum
- a Division of Hematology/Oncology , University of Virginia , Charlottesville , VA , USA
| |
Collapse
|
181
|
Affiliation(s)
- Paul S Gaynon
- Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
182
|
|
183
|
Curran E, Stock W. Taking a "BiTE out of ALL": blinatumomab approval for MRD-positive ALL. Blood 2019; 133:1715-1719. [PMID: 30796026 PMCID: PMC6634959 DOI: 10.1182/blood-2018-12-852376] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Blinatumomab, a bispecific T-cell engager (BiTE) associated with improved survival in relapsed or refractory acute lymphoblastic leukemia (ALL), was recently approved for treatment of minimal residual disease (MRD). MRD is an important predictor of survival in ALL, and recent studies suggest that achievement of MRD-negativity with blinatumomab improves outcomes in patients with ALL. However, further research is needed to determine how to optimally incorporate blinatumomab, and other novel therapies, into current therapies for ALL.
Collapse
Affiliation(s)
- Emily Curran
- Section of Hematology/Oncology, Department of Medicine, and University of Chicago Comprehensive Cancer Center, University of Chicago, Chicago, IL
| | - Wendy Stock
- Section of Hematology/Oncology, Department of Medicine, and University of Chicago Comprehensive Cancer Center, University of Chicago, Chicago, IL
| |
Collapse
|
184
|
Xu M, Liu H, Liu Y, Ma X, Qiu H, Fu C, Tang X, Han Y, Chen S, Wu D, Sun A. Gene mutations and pretransplant minimal residual disease predict risk of relapse in adult patients after allogeneic hematopoietic stem-cell transplantation for T cell acute lymphoblastic leukemia. Leuk Lymphoma 2019; 60:2744-2753. [PMID: 30950667 DOI: 10.1080/10428194.2019.1597270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Mingzhu Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Hong Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Yuejun Liu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Xiao Ma
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Huiying Qiu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Chengcheng Fu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Xiaowen Tang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Yue Han
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Suning Chen
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Depei Wu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| | - Aining Sun
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Suzhou Institute of Blood and Marrow Transplantation, Suzhou, China
| |
Collapse
|
185
|
Richard-Carpentier G, Kantarjian H, Jabbour E. Recent Advances in Adult Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2019; 14:106-118. [PMID: 30879177 DOI: 10.1007/s11899-019-00503-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW This article reviews the recent advances in the pathophysiology and management of acute lymphoblastic leukemia (ALL) in adults. RECENT FINDINGS Addition of rituximab to standard chemotherapy improves survival in the frontline treatment of B cell ALL, and measurable residual disease (MRD) is the most important prognostic factor. Tyrosine kinase inhibitors (TKI), particularly ponatinib, in combination with Hyper-CVAD significantly improve outcomes in Ph + ALL challenging the benefit of allogeneic stem cell transplant in first line for these patients. Blinatumomab, inotuzumab ozogamicin, and chimeric antigen receptor (CAR) T cells are better options than chemotherapy alone for the treatment of relapsed or refractory ALL. Combination of these agents with chemotherapy and their incorporation in the frontline setting show promises to improve cure rates of ALL. Development of monoclonal antibodies, CAR T, and potent TKI has improved the outcome of ALL. Advances in our understanding of ALL biology are expected to bring new therapeutic strategies in the upcoming years.
Collapse
Affiliation(s)
- Guillaume Richard-Carpentier
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 428, Houston, TX, 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 428, Houston, TX, 77030, USA
| | - Elias Jabbour
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, 1515 Holcombe Boulevard, Box 428, Houston, TX, 77030, USA.
| |
Collapse
|
186
|
Standardisation and consensus guidelines for minimal residual disease assessment in Philadelphia-positive acute lymphoblastic leukemia (Ph + ALL) by real-time quantitative reverse transcriptase PCR of e1a2 BCR-ABL1. Leukemia 2019; 33:1910-1922. [PMID: 30858550 DOI: 10.1038/s41375-019-0413-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022]
Abstract
Minimal residual disease (MRD) is a powerful prognostic factor in acute lymphoblastic leukemia (ALL) and is used for patient stratification and treatment decisions, but its precise role in Philadelphia chromosome positive ALL is less clear. This uncertainty results largely from methodological differences relating to the use of real-time quantitative PCR (qRT-PCR) to measure BCR-ABL1 transcript levels for MRD analysis. We here describe the first results by the EURO-MRD consortium on standardization of qRT-PCR for the e1a2 BCR-ABL1 transcript in Ph + ALL, designed to overcome the lack of standardisation of laboratory procedures and data interpretation. Standardised use of EAC primer/probe sets and of centrally prepared plasmid standards had the greatest impact on reducing interlaboratory variability. In QC1 the proportion of analyses with BCR-ABL1/ABL1 ratios within half a log difference were 40/67 (60%) and 52/67 (78%) at 10-3 and 36/67 (53%) and 53/67 (79%) at 10-4BCR-ABL1/ABL1. Standardized RNA extraction, cDNA synthesis and cycler platforms did not improve results further, whereas stringent application of technical criteria for assay quality and uniform criteria for data interpretation and reporting were essential. We provide detailed laboratory recommendations for the standardized MRD analysis in routine diagnostic settings and in multicenter clinical trials for Ph + ALL.
Collapse
|
187
|
PAX5 biallelic genomic alterations define a novel subgroup of B-cell precursor acute lymphoblastic leukemia. Leukemia 2019; 33:1895-1909. [PMID: 30842609 DOI: 10.1038/s41375-019-0430-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 11/08/2022]
Abstract
Chromosomal rearrangements and specific aneuploidy patterns are initiating events and define subgroups in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Here we analyzed 250 BCP-ALL cases and identified a novel subgroup ('PAX5-plus', n = 19) by distinct DNA methylation and gene expression profiles. All patients in this subgroup harbored mutations in the B-lineage transcription factor PAX5, with p.P80R as hotspot. Mutations either affected two independent codons, consistent with compound heterozygosity, or suffered LOH predominantly through chromosome 9p aberrations. These biallelic events resulted in disruption of PAX5 transcriptional programs regulating B-cell differentiation and tumor suppressor functions. Homozygous CDKN2A/B deletions and RAS-activating hotspot mutations were highly enriched as cooperating events in the genomic profile of PAX5-plus ALL. Together, this defined a specific pattern of triple alterations, exclusive to the novel subgroup. PAX5-plus ALL was observed in pediatric and adult patients. Although restricted by the limited sample size, a tendency for more favorable clinical outcome was observed, with 10 of 12 adult PAX5-plus patients achieving long-term survival. PAX5-plus represents the first BCP-ALL subgroup defined by sequence alterations in contrast to gross chromosomal events and exemplifies how deregulated differentiation (PAX5), impaired cell cycle control (CDKN2A/B) and sustained proliferative signaling (RAS) cooperatively drive leukemogenesis.
Collapse
|
188
|
Gökbuget N, Dombret H, Giebel S, Bruggemann M, Doubek M, Foà R, Hoelzer D, Kim C, Martinelli G, Parovichnikova E, Rambaldi A, Ribera JM, Schoonen M, Stieglmaier JM, Zugmaier G, Bassan R. Minimal residual disease level predicts outcome in adults with Ph-negative B-precursor acute lymphoblastic leukemia. ACTA ACUST UNITED AC 2019; 24:337-348. [PMID: 30757960 DOI: 10.1080/16078454.2019.1567654] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Detectable minimal residual disease (MRD) after therapy for acute lymphoblastic leukemia (ALL) is the strongest predictor of hematologic relapse. This study evaluated outcomes of patients with B-cell precursor ALL with MRD of ≥10-4 Methods: Study population was from ALL study groups in Europe managed in national study protocols 2000-2014. MRD was measured by polymerase chain reaction or flow cytometry. Patients were age ≥15 years at initial ALL diagnosis. Patients were excluded if exposed to blinatumomab within 18 months of baseline or prior alloHSCT. RESULTS Of 272 patients in CR1, baseline MRD was ≥10-1, 10-2 to <10-1, 10-3 to <10-2, and 10-4 to <10-3 in 15 (6%), 71 (26%), 109 (40%), and 77 (28%) patients, respectively. Median duration of complete remission (DoR) was 18.5 months (95% confidence interval [CI], 11.9-27.2), median relapse-free survival (RFS) was 12.4 months (95% CI, 10.0-19.0) and median overall survival (OS) was 32.5 months (95% CI, 23.6-48.0). Lower baseline MRD level (P ≤ .0003) and white blood cell count <30,000/µL at diagnosis (P ≤ .0053) were strong predictors for better RFS and DoR. Allogeneic hematopoietic stem cell transplantation (alloHSCT) was associated with longer RFS (hazard ratio [HR], 0.59; 95% CI, 0.41-0.84) and DoR (HR, 0.43; 95% CI, 0.29-0.64); the association with OS was not significant (HR, 0.72; 95% CI, 0.50-1.05). DISCUSSION In conclusion, RFS, DoR, and OS are relatively short in patients with MRD-positive ALL, particularly at higher MRD levels. AlloHSCT may improve survival but has limitations. Alternative approaches are needed to improve outcomes in MRD-positive ALL.
Collapse
Affiliation(s)
- Nicola Gökbuget
- a Department of Medicine II, Department of Hematology/Oncology , University Hospital , Frankfurt , Germany
| | - Hervé Dombret
- b Hôpital Saint-Louis, University Paris Diderot , Paris , France
| | - Sebastian Giebel
- c Maria Sklodowska Curie Memorial Cancer Center , Gliwice , Poland
| | - Monika Bruggemann
- d Department of Hematology and Oncology , University Hospital Schleswig-Holstein, Campus Kiel , Kiel , Germany
| | - Michael Doubek
- e Department of Internal Medicine, Hematology and Oncology , University Hospital , Brno , Czech Republic
| | - Robin Foà
- f "Sapienza" University of Rome , Rome , Italy
| | - Dieter Hoelzer
- a Department of Medicine II, Department of Hematology/Oncology , University Hospital , Frankfurt , Germany
| | | | - Giovanni Martinelli
- h Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | | | - Alessandro Rambaldi
- j Dipartimento di Oncologia ed Ematologia , Università degli Studi di Milano and Ospedale Papa Giovanni XXIII , Bergamo , Italy
| | - Josep-Maria Ribera
- k ICO-Hospital Germans Trias I Pujol, Josep Carreras Research Institute , Barcelona , Spain
| | | | | | | | | |
Collapse
|
189
|
King AC, Pappacena JJ, Tallman MS, Park JH, Geyer MB. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia. Leuk Res 2019; 79:27-33. [PMID: 30831480 DOI: 10.1016/j.leukres.2019.02.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 11/28/2022]
Abstract
Incorporation of ABL-targeted oral tyrosine kinase inhibitors (TKIs) into frontline therapeutic regimens has improved outcomes for adults with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph + ALL). However, patients with persistent minimal residual disease (MRD) exhibit increased risk of relapse. Combining consolidative chemotherapy with TKIs may increase rates of infectious complications, organ toxicity, hospitalization, and non-relapse mortality. Blinatumomab has demonstrated single-agent activity in patients with relapsed B-ALL or persistent MRD, including Ph + B-ALL. We have used blinatumomab concomitantly with commercially available TKIs as consolidative therapy to spare toxicities of conventional chemotherapy. We evaluated 11 adults with previously treated Ph + B-ALL who received blinatumomab concurrent with TKI (ponatinib, n = 5; dasatinib, n = 4; nilotinib, n = 1; imatinib, n = 1) to eradicate MRD or sustain MRD-negativity. Eight of 9 patients with MRD achieved BCR-ABL1 negativity (complete molecular response, CMR) after a median of one cycle; 2/2 patients without measurable disease durably maintained CMR. Cytokine release syndrome (all grade 1-2) was observed in 3/11 patients; one patient experienced transient grade 1 neurologic toxicity. Transient grade 2 transaminitis was observed in 6/11 patients, including 4/5 recipients of blinatumomab + ponatinib. This small series suggests blinatumomab + TKI is a safe and effective consolidation strategy for patients with Ph + ALL to achieve or maintain CMR.
Collapse
Affiliation(s)
- Amber C King
- Department of Pharmacy, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Jeremy J Pappacena
- Department of Pharmacy, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Mark B Geyer
- Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA; Cellular Therapeutics Center, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
190
|
Short NJ, Jabbour E, Albitar M, de Lima M, Gore L, Jorgensen J, Logan AC, Park J, Ravandi F, Shah B, Radich J, Kantarjian H. Recommendations for the assessment and management of measurable residual disease in adults with acute lymphoblastic leukemia: A consensus of North American experts. Am J Hematol 2019; 94:257-265. [PMID: 30394566 PMCID: PMC6572728 DOI: 10.1002/ajh.25338] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
Measurable residual disease (MRD) that persists after initial therapy is a powerful predictor of relapse and survival in acute lymphoblastic leukemia (ALL). However, the optimal use of this information to influence therapeutic decisions is controversial. Herein, we comprehensively review the role of MRD assessment in adults with ALL, including methods to quantify residual leukemia cells during remission, prognostic impact of MRD across ALL subtypes, and available therapeutic approaches to eradicate MRD. This review presents consensus statements and provides an evidence-based framework for practicing hematologists and oncologists to use MRD information to make rational treatment decisions in adult patients with ALL.
Collapse
Affiliation(s)
| | - Elias Jabbour
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Lia Gore
- Children’s Hospital Colorado and University of Colorado Cancer Center, Aurora, CO
| | | | - Aaron C. Logan
- University of California San Francisco, San Francisco, CA
| | - Jae Park
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Farhad Ravandi
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | |
Collapse
|
191
|
Stratmann JA, von Rose AB, Koschade S, Wendelin K, Köhler F, Heinsch M, Schiller K, Haferlach C, Wattad M, Rieder H, Serve H, Gökbuget N, Steffen B. Clinical and genetic characterization of de novo double-hit B cell precursor leukemia/lymphoma. Ann Hematol 2019; 98:647-656. [PMID: 30613837 DOI: 10.1007/s00277-018-03590-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/25/2018] [Indexed: 12/22/2022]
Abstract
The 2016 revised World Health Organization (WHO) classification of lymphoid neoplasms included the category of high-grade B cell lymphomas (HGBLs) with combined MYC and BCL2 and/or BCL6 rearrangements (double-hit, DH). However, the clinical features of B cell precursor leukemia (BCP-ALL) that harbor DH genetics remain widely unknown. We performed a retrospective analysis of the German Multicenter Study Group for Adult ALL registry and a literature search for de novo DH-BCP-ALLs. We identified 6 patients in the GMALL registry and 11 patients published in the literature between 1983 and June 2018. Patients of all ages (range, 15-86 years) are affected. There is a high incidence of meningeal disease and other extramedullary disease manifestations. Current treatment approaches are mainly ALL-based and are sufficient to induce first complete remissions, but progression-free survival is only 4.0 months (95% CI, 1.5-6.5 months) and all patients succumb to their disease, once relapsed, with a median survival of 5.0 months (95% CI, 3.1-6.9 months), despite intensive salvage and targeted therapy approaches. Of all patients, only two that attained an initial complete remission were alive at data cutoff. In all cases, the BCL2 gene was rearranged to be in proximity to the IGH locus, whereas MYC had various translocation partners juxtaposed. There was no significant survival difference between IG and non-IG translocation partners (HR, 1.03; 95% CI, 0.33-3.2; p = 0.89). In conclusion, de novo DH-BCP-ALL is an aggressive B cell malignancy with deleterious outcome. Physicians have to be aware of this rare disease subset due to the atypical clinical behavior and especially because latest classification systems do not cover this sub-entity.
Collapse
Affiliation(s)
- Jan A Stratmann
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany.
| | - Aaron Becker von Rose
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany
| | - Sebastian Koschade
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany
| | - Knut Wendelin
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Prof.-Ernst-Nathan-Str. 1, 90419, Nuremberg, Germany
| | - Friedemann Köhler
- Department of. Internal Medicine II, Schwarzwald-Baar Clinic Villingen-Schwenningen, Klinikstr. 11, 78052, Villingen-Schwenningen, Germany
| | - Michael Heinsch
- Department of Internal Medicine II, St Johannes Hospital, An der Abtei 7-11, 47166, Duisburg, Germany
| | - Kilian Schiller
- Department of Radiation Oncology, Technical University of Munich (TUM), Ismaninger Strasse 22, 81675, Munich, Germany
| | - Claudia Haferlach
- Munich Leukemia Laboratory (MLL), Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Mohamed Wattad
- Department of Hematology and Oncology, Hospital Essen-Werden, Pattbergstraße 1-3, 45239, Essen, Germany
| | - Harald Rieder
- Institute for Human Genetics, Medical Faculty, Heinrich-Heine-University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Hubert Serve
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany
| | - Nicola Gökbuget
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany
| | - Björn Steffen
- Department of Hematology and Oncology, Johann Wolfgang Goethe University of Frankfurt, Theodor Stern Kai 7, 60596, Frankfurt am Main, Germany
| |
Collapse
|
192
|
Hefazi M, Litzow MR. Recent Advances in the Biology and Treatment of T Cell Acute Lymphoblastic Leukemia. Curr Hematol Malig Rep 2018; 13:265-274. [PMID: 29948644 DOI: 10.1007/s11899-018-0455-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW This article provides an overview of the current knowledge regarding the biology and treatment of T cell acute lymphoblastic leukemia (T-ALL) and highlights the most recent findings in this field over the past 5 years. RECENT FINDINGS Remarkable progress has been made in the genomic landscape of T-ALL over the past few years. The discovery of activating mutations of NOTCH1 and FBXW7 in a majority of patients has been a seminal observation, with several early phase clinical trials currently exploring these as potential therapeutic targets. Characterization of early T cell precursor ALL, incorporation of minimal residual disease assessment into therapeutic protocols, and use of pediatric-intensive regimens along with judicious use of allogeneic HCT have significantly improved risk stratification and treatment outcomes. Improved risk stratification and the use of novel targeted therapies based on recent genomic discoveries are expected to change the therapeutic landscape of T-ALL and hopefully improve the outcomes of this historically poor prognosis disease.
Collapse
Affiliation(s)
- Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
193
|
Cao P, Yu Y, Wang W, Xu H, He Y. Fluorescence in situ hybridization comparison of the prognostic factors in adult and pediatric acute lymphoblastic leukemia: A retrospective analysis of 282 cases. Exp Ther Med 2018; 16:4674-4684. [PMID: 30542419 PMCID: PMC6257547 DOI: 10.3892/etm.2018.6821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 08/31/2018] [Indexed: 11/13/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) affects both children and adults. However, the prognosis of the two cohorts is quite different. The present aim was to review and evaluate one potential cause of why survival is poorer in adult ALL than pediatric ALL via fluorescence in situ hybridization (FISH). Clinical significant features were analyzed in 282 ALL cases. FISH was performed to study mixed lineage leukemia (MLL) translocation and the Philadelphia (Ph) chromosome in newly diagnosed patients, and was used to detect trisomy 4 or 10 and the translocation ETS leukemia-acute myeloid leukemia 1 (TEL-AML1) fusion gene. The overall survival/event-free survival (OS/EFS) outcome of adult ALL and pediatric ALL was analyzed using Kaplan-Meier analysis. Adult ALL had a higher median leukocyte count and lower hemoglobin level than pediatric ALL. FISH revealed that Ph positivity (Ph+) was associated with the high-risk feature of older age. In pediatric ALL, trisomy 4 or 10 was present in 71/207 cases (34.3%), while the TEL-AML1 fusion gene was present in 16/207 cases (7.7%). By contrast, there were very few such positive cases in adult ALL. Survival analysis revealed that, in adult ALL, the 3-year OS and EFS rates were higher in the Ph-negative group than in the Ph+ group. Adult or pediatric ALL is an independent prognostic factor of OS. The present analysis of the clinical and biological features between adult and pediatric ALL indicates that adult ALL has a poorer prognosis than pediatric ALL based on Ph+ status and presence of trisomy 4 or 10. Ph+ ALL is an independent prognosis factor of ALL. FISH may serve an important role in the comparison of prognostic factors in adult and pediatric ALL.
Collapse
Affiliation(s)
- Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China.,Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yalan Yu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei Wang
- Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - He Xu
- Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yuxiang He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
194
|
Barba P, Morgades M, Montesinos P, Gil C, Fox M, Ciudad J, Moreno M, González‐Campos J, Genescà E, Martínez‐Carballeira D, Martino R, Vives S, Guardia R, Mercadal S, Artola M, Cladera A, Tormo M, Esteve J, Bergua J, Vall‐Llovera F, Ribera J, Martínez‐Sanchez P, Amigo M, Bermúdez A, Calbacho M, Hernández‐Rivas J, Feliu E, Orfao A, Ribera J. Increased survival due to lower toxicity for high‐risk T‐cell acute lymphoblastic leukemia patients in two consecutive pediatric‐inspired PETHEMA trials. Eur J Haematol 2018; 102:79-86. [DOI: 10.1111/ejh.13178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Pere Barba
- Hospital Universitari Vall Hebron Universitat Autònoma de Barcelona Barcelona Spain
| | - Mireia Morgades
- ICO Badalona‐Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute Universitat Autònoma de Barcelona Badalona Spain
| | | | | | - María‐Laura Fox
- Hospital Universitari Vall Hebron Universitat Autònoma de Barcelona Barcelona Spain
| | - Juana Ciudad
- Centro de Investigación del Cáncer (CIC, IBMCC USAL‐CSIC), Servicio General de Citometría, Instituto de Investigación Biomédica de Salamanca (IBSAL) Universidad de Salamanca Salamanca Spain
| | | | | | - Eulàlia Genescà
- Josep Carreras Leukaemia Research Institute (IJC) Badalona Spain
| | | | | | - Susana Vives
- ICO Badalona‐Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute Universitat Autònoma de Barcelona Badalona Spain
| | | | - Santiago Mercadal
- ICO L'Hospitalet‐ Hospital Duran i Reynals L'Hospitalet de Llobregat Spain
| | | | | | - Mar Tormo
- Hospital Clínico Universitario de Valencia Valencia Spain
| | - Jordi Esteve
- Hospital Clínic Universitari de Barcelona Barcelona Spain
| | - Juan Bergua
- Hospital San Pedro de Alcántara Cáceres Spain
| | | | - Jordi Ribera
- Josep Carreras Leukaemia Research Institute (IJC) Badalona Spain
| | | | | | | | | | | | - Evaristo Feliu
- ICO Badalona‐Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute Universitat Autònoma de Barcelona Badalona Spain
| | - Alberto Orfao
- Centro de Investigación del Cáncer (CIC, IBMCC USAL‐CSIC), Servicio General de Citometría, Instituto de Investigación Biomédica de Salamanca (IBSAL) Universidad de Salamanca Salamanca Spain
| | - Josep‐María Ribera
- ICO Badalona‐Hospital Germans Trias i Pujol, Josep Carreras Leukemia Research Institute Universitat Autònoma de Barcelona Badalona Spain
| |
Collapse
|
195
|
Burt R, Warcel D, Fielding AK. Blinatumomab, a bispecific B-cell and T-cell engaging antibody, in the treatment of B-cell malignancies. Hum Vaccin Immunother 2018; 15:594-602. [PMID: 30380973 PMCID: PMC6605719 DOI: 10.1080/21645515.2018.1540828] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/22/2018] [Indexed: 01/28/2023] Open
Abstract
Blinatumomab (Blincyto, Amgen), a bi-specific antibody, is a first-in-class, targeted immunotherapy agent for treatment of B-cell malignancies with a novel mechanism of action which involves in-vivo engagement of the patient's T cells with CD19-expressing tumour cells. Clinical trials have demonstrated its efficacy in relapsed B-cell Acute Lymphoblastic Leukaemia (B-ALL) and B-cell Non-Hodgkin's Lymphoma including in patients who are refractory to chemotherapy. This review summarises the development and design of Blinatumomab, the outcome of clinical studies demonstrating its efficacy and how to manage the administration, practically, including relevant toxicities. We compare and contrast it to other emerging agents for treatment of B-cell malignancies.
Collapse
|
196
|
Aldoss I, Pullarkat V. Indications for allogeneic hematopoietic cell transplantation for adults with Philadelphia-chromosome negative acute lymphoblastic leukemia in first complete remission: all about MRD? Bone Marrow Transplant 2018; 54:3-5. [DOI: 10.1038/s41409-018-0398-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 10/31/2018] [Indexed: 12/25/2022]
|
197
|
Testing for minimal residual disease in adults with acute lymphoblastic leukemia in Europe: a clinician survey. BMC Cancer 2018; 18:1100. [PMID: 30419861 PMCID: PMC6233570 DOI: 10.1186/s12885-018-5002-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/28/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In acute lymphoblastic leukemia (ALL), the presence of minimal residual disease (MRD) after induction/consolidation chemotherapy is a strong prognostic factor for subsequent relapse and mortality. Accordingly, European clinical guidelines and protocols recommend testing patients who achieve a complete hematological remission (CR) for MRD for the purpose of risk stratification. The aim of this study was to provide quantitative information regarding real-world clinical practice for MRD testing in five European countries. METHODS A web-based survey was conducted in March/April 2017 in France, Germany, Italy, Spain, and the UK. The survey was developed after consultation with specialist clinicians and a review of published literature. Eligible clinicians (20 per country; 23 in Spain) were board-certified in hemato-oncology or hematology, had at least five years' experience in their current role after training, had treated at least two patients with B-cell precursor ALL in the 12 months before the survey or at least five patients in the last five years, and had experience of testing for MRD in clinical practice. RESULTS MRD testing is now standard practice in the treatment of adult ALL across the five European countries, with common use of recent treatment protocols which specify testing. Respondents estimated that, among clinicians in their country who conduct MRD testing, 73% of patients in first CR (CR1) and 63% of patients in second or later CR (CR2+) are tested for MRD. The median time point reported as most commonly used for the first MRD test, to establish risk status and to determine a treatment plan was four weeks after the start of induction therapy. The timing and frequency of tests is similar across countries. An average of four or five post-CR1 tests per patient in the 12 months after the first MRD test were reported across countries. CONCLUSIONS This comprehensive study of MRD testing patterns shows consistent practice across France, Germany, Italy, Spain, and the UK with respect to the timing and frequency of MRD testing, aligning with use of national protocols. MRD testing is used in clinical practice also in patients who reach CR2 + .
Collapse
|
198
|
Yafour N, Couturier MA, Azarnoush S, Girault S, Hermet E, Masouridi Levrat S, Schmidt A, Michallet M, Etancelin P, Guillaume T, Malard F, Sirvent A, Yakoub-Agha I, Poiré X. [Second allogeneic hematopoietic stem cell transplant: Guidelines from the francophone Society of bone marrow transplantation and cellular therapy (SFGM-TC)]. Bull Cancer 2018; 106:S40-S51. [PMID: 30409466 DOI: 10.1016/j.bulcan.2018.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/31/2018] [Indexed: 01/10/2023]
Abstract
Disease recurrence and graft dysfunction after allogeneic hematopoietic stem cell transplantation (allo-HSCT) currently remain among the major causes of treatment failure in malignant and non-malignant hematological diseases. A second allo-HSCT is a valuable therapeutic option to salvage those situations. During the 8th annual harmonization workshops of the french Society of bone marrow transplantation and cellular therapy (SFGM-TC), a designated working group reviewed the literature in order to elaborate unified guidelines on feasibility, indications, donor choice and conditioning in the case of a second allo-HSCT. In case of relapse, a second allo-HSCT with reduced intensity or non-myeloablative conditioning is a reasonable option, particularly in patients with a good performance status (Karnofsky/Lansky>80%), low co-morbidity score (EBMT score≤3), a longer remission duration after the first allo-HSCT (>6 months), and who present low disease burden at the time of second allo-HSCT. Matched related donors tend to be associated with better outcomes. In the presence of graft dysfunction (primary and secondary graft rejection), an immunoablative conditioning regimen is recommended. A donor change remains a valid option, especially in the absence of graft-versus-host disease after the first allo-HSCT.
Collapse
Affiliation(s)
- Nabil Yafour
- Établissement Hospitalier et Universitaire 1er-Novembre 1954, service d'hématologie et de thérapie cellulaire, BP 4166, 31000 Ibn Rochd, Oran, Algérie; Université d'Oran 1, Ahmed Ben Bella, faculté de médecine, Oran, Algérie.
| | - Marie Anne Couturier
- Hôpital Morvan, institut cancérologie-hématologie, CHRU Brest, 2, avenue Foch, 29200 Brest, France
| | - Saba Azarnoush
- Université Paris Diderot, hôpital Robert-Debré, service d'immuno-hématologie pédiatrique, 48, boulevard Sérurier, 75019 Paris, France
| | - Stéphane Girault
- CHU Limoges, hématologie clinique et thérapie cellulaire, 2, avenue Martin-Luther-King, 87042 Limoges, France
| | - Eric Hermet
- Université d'Auvergne EA3846, CIC-501, CHU Estaing, service de thérapie cellulaire et d'hématologie clinique adulte, Clermont-Ferrand, 58, rue Montalembert, 63000 Clermont-Ferrand, France
| | - Stavroula Masouridi Levrat
- Geneva university hospitals, division of hematology, department of medical specialties, rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Suisse
| | - Aline Schmidt
- CHU d'Angers, maladies du sang, 4, rue Larrey, 49100 Angers, France; Université d'Angers, Inserm U892/CNRS 6299, 49035 Angers, France
| | - Mauricette Michallet
- Centre hospitalier Lyon Sud, hématologie clinique, 165, chemin du Grand-Revoyet, 69495 Pierre-Bénite cedex Lyon, France
| | - Pascaline Etancelin
- Centre Henri-Becquerel, laboratoire de génétique oncologique, rue d'Amiens, 76000 Rouen, France
| | - Thierry Guillaume
- CHU de Nantes, Hôtel-Dieu, service d'hématologie, 1, place Ricordeau, 44000 Nantes, France
| | - Florent Malard
- AP-HP, hôpital Saint-Antoine, service d'hématologie et de thérapie cellulaire, 75012 Paris, France
| | - Anne Sirvent
- CHU Montpellier, hôpital Arnaud-de-Villeneuve, service de l'onco-hématologie pédiatrique, 371, avenue du Doyen-Gaston-Giraud, 34090 Montpellier, France
| | - Ibrahim Yakoub-Agha
- CHU de Lille, maladies du sang, unité d'Allogreffe de CSH, 59000 Lille, France; Université de Lille 2, LIRIC, Inserm U995, 59000 Lille, France
| | - Xavier Poiré
- Cliniques universitaires Saint-Luc, service d'hématologie, 10, avenue Hippocrate, 1200 Bruxelles, Belgique
| |
Collapse
|
199
|
Aldoss I, Kamal MO, Forman SJ, Pullarkat V. Adults with Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia: Considerations for Allogeneic Hematopoietic Cell Transplantation in First Complete Remission. Biol Blood Marrow Transplant 2018; 25:e41-e45. [PMID: 30292743 DOI: 10.1016/j.bbmt.2018.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 09/27/2018] [Indexed: 10/28/2022]
Abstract
Philadelphia chromosome-like (Ph-like) acute lymphoblastic leukemia (ALL) is a subset of high-risk B cell ALLs. A large proportion of Ph-like ALL cases carry activating kinase mutations that could potentially allow them to be targeted by tyrosine kinase inhibitors. Ph-like ALL is not an uncommon entity, especially among adults, with a frequency exceeding 20%, including in older patients (>60 years old) with ALL. Ph-like ALL is associated with inferior outcomes across all ages, and studies have consistently shown a higher incidence of persistent postinduction minimal residual disease in patients carrying Ph-like ALL compared with other subgroups of ALL, and this translates into inferior leukemia-related outcomes. The inferior outcome of conventional chemotherapy for Ph-like ALL in adults raises the fundamental question of whether all adults with Ph-like ALL require an allogeneic hematopoietic cell transplantation (HCT) in first complete remission (CR1) regardless of other presenting features and treatment response parameters. Here we present and discuss several scenarios in which adults with Ph-like ALL underwent or were considered for HCT in CR1 for various reasons. Although the decision to proceed with HCT was clear and indisputable in some of these situations, in others we struggled with the decision to transplant in CR1 because of the lack of published data regarding the efficacy of allogeneic HCT as consolidation for Ph-like ALL. We emphasize the urgent need for developing well-designed studies to address this important question.
Collapse
Affiliation(s)
- Ibrahim Aldoss
- Department of Hematology and Hematopoietic cell transplantation, City of Hope Medical Center, Duarte, California.
| | - Muhammad O Kamal
- Department of Hematology and Oncology, Loma Linda Medical Center, Loma Linda, California
| | - Stephen J Forman
- Department of Hematology and Hematopoietic cell transplantation, City of Hope Medical Center, Duarte, California
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic cell transplantation, City of Hope Medical Center, Duarte, California
| |
Collapse
|
200
|
Hefazi M, Litzow MR. Recent advances in the biology and treatment of B-cell acute lymphoblastic leukemia. Blood Lymphat Cancer 2018; 8:47-61. [PMID: 31360093 PMCID: PMC6467350 DOI: 10.2147/blctt.s170351] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is a hematologic malignancy arising from precursors of the lymphoid lineage. Conventional cytotoxic chemotherapies have resulted in high cure rates of up to 90% in pediatric ALL, but the outcomes for adult patients remain suboptimal with 5-year survival rates of only 30%-40%. Over the last decade, major advances have been made in our understanding and management of ALL. Identification of new prognostic genomic markers and incorporation of minimal residual diseases' assessment into therapeutic protocols have improved risk stratification and treatment strategies. The use of pediatric-inspired regimens for adolescent and young adults, and the advent of tyrosine kinase inhibitors and novel targeted therapies, including monoclonal antibodies and chimeric antigen receptor T cells, have redefined the therapeutic paradigm of ALL, and significantly improved the outcomes. In this article, we will provide an overview of the current knowledge regarding the biology and treatment of ALL, and highlight recent diagnostic and therapeutic advances made in this area over the past 5 years.
Collapse
Affiliation(s)
- Mehrdad Hefazi
- Division of Hematology, Mayo Clinic, Rochester, MN, USA,
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN, USA,
| |
Collapse
|