101
|
Pochon C, Oger E, Michel G, Dalle JH, Salmon A, Nelken B, Bertrand Y, Cavé H, Cayuela JM, Grardel N, Macintyre E, Margueritte G, Méchinaud F, Rohrlich P, Paillard C, Demeocq F, Schneider P, Plantaz D, Poirée M, Eliaou JF, Semana G, Drunat S, Jonveaux P, Bordigoni P, Gandemer V. Follow-up of post-transplant minimal residual disease and chimerism in childhood lymphoblastic leukaemia: 90 d to react. Br J Haematol 2014; 169:249-61. [DOI: 10.1111/bjh.13272] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 11/23/2014] [Indexed: 01/24/2023]
Affiliation(s)
- Cécile Pochon
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| | - Emmanuel Oger
- Clinical Pharmacology Department; Pharmacoepidemiology Team; University Hospital of Rennes; Rennes France
| | - Gérard Michel
- Department of Paediatric Haematology; University Hospital of La Timone; Marseille France
| | - Jean-Hugues Dalle
- Department of Paediatric Haematology; University Hospital of Robert Debré; Paris France
| | - Alexandra Salmon
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| | - Brigitte Nelken
- Department of Paediatric Haematology/Oncology; University Hospital of Jeanne de Flandre; Lille France
| | - Yves Bertrand
- Department of Paediatric Haematology; Hospices Civils de Lyon; Lyon France
| | - Hélène Cavé
- Department of Genetics; University Hospital of Robert Debré and Paris-Diderot University; Paris France
| | | | - Nathalie Grardel
- Laboratory of Haematology; University Hospital of Calmette; Lille France
| | | | - Geneviève Margueritte
- Department of Paediatric Haematology/Oncology; University Hospital of Villeneuve; Montpellier France
| | - Françoise Méchinaud
- Department of Paediatric Haematology/Oncology; University Hospital of Nantes; Nantes France
| | - Pierre Rohrlich
- Department of Paediatric Haematology/Oncology; University Hospital of Besançon; Besançon France
| | - Catherine Paillard
- Department of Paediatric Haematology/Oncology; University Hospital of Hautepierre; Strasbourg France
| | - François Demeocq
- Department of Paediatric Haematology/oncology; University Hospital of Clermont-Ferrand; Clermont-Ferrand France
| | - Pascale Schneider
- Department of Paediatric Haematology; University Hospital of Rouen; Rouen France
| | - Dominique Plantaz
- Department of Paediatric Haematology/oncology; University Hospital of La Tronche; Grenoble France
| | - Marilyne Poirée
- Department of Paediatric Haematology/oncology; University Hospital of Archet II; Nice France
| | - Jean-François Eliaou
- Laboratory of Immunology; University Hospital of Montpellier; Montpellier France
| | - Gilbert Semana
- Laboratory of Immunology; French blood transfusion centre; Rennes France
| | - Séverine Drunat
- Department of Genetics; University Hospital of Robert Debré and Paris-Diderot University; Paris France
| | | | - Pierre Bordigoni
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| | - Virginie Gandemer
- Department of paediatric Haematology/Oncology; University Hospital of Rennes; Rennes France
| |
Collapse
|
102
|
Schrappe M. Detection and management of minimal residual disease in acute lymphoblastic leukemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:244-249. [PMID: 25696862 DOI: 10.1182/asheducation-2014.1.244] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The detection of minimal residual disease (MRD) has become part of the state-of-the-art diagnostics to guide treatment both in pediatric and adult acute lymphoblastic leukemia (ALL). This applies to the treatment of de novo and recurrent ALL. In high-risk ALL, MRD detection is considered an important tool to adjust therapy before and after hematopoietic stem cell transplantation. Precise quantification and quality control is instrumental to avoid false treatment assignment. A new methodological approach to analyzing MRD has become available and is based on next-generation sequencing. In principle, this technique will be able to detect a large number of leukemic subclones at a much higher speed than before. Carefully designed prospective studies need to demonstrate concordance or even superiority compared with those techniques in use right now: detection of aberrant expression of leukemia-specific antigens by flow cytometry of blood or bone marrow, or detection of specific rearrangements of the T-cell receptor or immunoglobulin genes by real-time quantitative polymerase chain reaction using DNA of leukemic cells. In some cases with known fusion genes, such as BCR/ABL, reverse transcriptase-polymerase chain reaction has been used as additional method to identify leukemic cells by analyzing RNA in patient samples. MRD detection may be used to modulate treatment intensity once it has been demonstrated at well-defined informative checkpoints that certain levels of MRD can reliably predict the risk of relapse. In addition, MRD is used as end point to determine the activity of a given agent or treatment protocol. If activity translates into antileukemic efficacy, MRD may be considered a surrogate clinical end point.
Collapse
Affiliation(s)
- Martin Schrappe
- Department of Pediatrics, Christian-Albrechts-University of Kiel, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
103
|
Spinelli O, Tosi M, Guinea Montalvo ML, Peruta B, Parolini M, Scattolin AM, Maino E, Viero P, Rambaldi A, Bassan R. Prognostic impact of minimal residual disease in adult acute lymphoblastic leukemia. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY While adult acute lymphoblastic leukemia (ALL) is curable in 40–50% of the patients, the individual prognosis is rather unpredictable due to associated biological and clinical risk factors. In both B- and T-precursor ALL, minimal residual disease (MRD) represents the most sensitive prognostic marker, useful to support critical treatment decisions, ranging from allogeneic stem cell transplantation in patients with inadequate MRD response to chemotherapy only in MRD responsive ones. This optimized risk-adapted strategy allows to spare transplant-associated morbidity and mortality in patients curable by chemotherapy. Further progress is expected from the integration of the MRD-based strategy with improved pediatric-type regimens and novel targeting agents for discrete ALL subsets. These changes are increasing the cure rate to above 50%.
Collapse
Affiliation(s)
- Orietta Spinelli
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Manuela Tosi
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Barbara Peruta
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Margherita Parolini
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Anna Maria Scattolin
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Elena Maino
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Piera Viero
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| | - Alessandro Rambaldi
- Hematology & Bone Marrow Transplant Unit of Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Renato Bassan
- Hematology & Bone Marrow Transplant Unit, Ospedale dell'Angelo e SS. Giovanni e Paolo, Via Paccagnella 11, 30174 Mestre-Venezia, Mestre-Venezia, Italy
| |
Collapse
|
104
|
Marks DI, Alonso L, Radia R. Allogeneic Hematopoietic Cell Transplantation in Adult Patients with Acute Lymphoblastic Leukemia. Hematol Oncol Clin North Am 2014; 28:995-1009. [DOI: 10.1016/j.hoc.2014.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
105
|
Sutton R, Shaw PJ, Venn NC, Law T, Dissanayake A, Kilo T, Haber M, Norris MD, Fraser C, Alvaro F, Revesz T, Trahair TN, Dalla-Pozza L, Marshall GM, O'Brien TA. Persistent MRD before and after allogeneic BMT predicts relapse in children with acute lymphoblastic leukaemia. Br J Haematol 2014; 168:395-404. [DOI: 10.1111/bjh.13142] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/26/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Rosemary Sutton
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Peter J. Shaw
- Oncology Unit; The Children's Hospital at Westmead; Westmead NSW Australia
| | - Nicola C. Venn
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Tamara Law
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Anuruddhika Dissanayake
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Tatjana Kilo
- Oncology Unit; The Children's Hospital at Westmead; Westmead NSW Australia
| | - Michelle Haber
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Murray D. Norris
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
| | - Chris Fraser
- Royal Children's Hospital; Brisbane Qld Australia
| | - Frank Alvaro
- John Hunter Children's Hospital; Newcastle NSW Australia
| | - Tamas Revesz
- Women and Children's Hospital; Adelaide SA Australia
| | - Toby N. Trahair
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
- Kids Cancer Centre; Sydney Children's Hospital; Randwick NSW Australia
| | | | - Glenn M. Marshall
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
- Kids Cancer Centre; Sydney Children's Hospital; Randwick NSW Australia
| | - Tracey A. O'Brien
- Children's Cancer Institute Australia; Lowy Cancer Research Centre, UNSW; Randwick NSW Australia
- Kids Cancer Centre; Sydney Children's Hospital; Randwick NSW Australia
| |
Collapse
|
106
|
Tian H, Chen GH, Xu Y, Ma X, Chen F, Yang Z, Jin ZM, Qiu HY, Sun AN, Wu DP. Impact of pre-transplant disease burden on the outcome of allogeneic hematopoietic stem cell transplant in refractory and relapsed acute myeloid leukemia: a single-center study. Leuk Lymphoma 2014; 56:1353-61. [DOI: 10.3109/10428194.2014.961016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
107
|
Fuster JL. Current approach to relapsed acute lymphoblastic leukemia in children. World J Hematol 2014; 3:49-70. [DOI: 10.5315/wjh.v3.i3.49] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 05/31/2014] [Accepted: 06/18/2014] [Indexed: 02/05/2023] Open
Abstract
Recurrent acute lymphoblastic leukaemia (ALL) is a common disease for pediatric oncologists and accounts for more deaths from cancer in children than any other malignancy. Although most patients achieve a second remission, about 50% of relapsed ALL patients do not respond to salvage therapy or suffer a second relapse and most children with relapse die. Treatment must be tailored after relapse of ALL, since outcome will be influenced by well-established prognostic features, including the timing and site of disease recurrence, the disease immunophenotype, and early response to retrieval therapy in terms of minimal residual disease (MRD). After reinduction chemotherapy, high risk (HR) patients are clear candidates for allogeneic stem cell transplantation (SCT) while standard risk patients do better with conventional chemotherapy and local therapy. Early MRD response assessment is currently applied to identify those patients within the more heterogeneous intermediate risk group who should undergo SCT as consolidation therapy. Recent evidence suggests distinct biological mechanisms for early vs late relapse and the recognition of the involvement of certain treatment resistance related genes as well cell cycle regulation and B-cell development genes at relapse, provides the opportunity to search for novel target therapies.
Collapse
|
108
|
Bassan R, Spinelli O, Oldani E, Intermesoli T, Tosi M, Peruta B, Borlenghi E, Pogliani EM, Di Bona E, Cassibba V, Scattolin AM, Romani C, Ciceri F, Cortelezzi A, Gianfaldoni G, Mattei D, Audisio E, Rambaldi A. Different molecular levels of post-induction minimal residual disease may predict hematopoietic stem cell transplantation outcome in adult Philadelphia-negative acute lymphoblastic leukemia. Blood Cancer J 2014; 4:e225. [PMID: 25014772 PMCID: PMC4219445 DOI: 10.1038/bcj.2014.48] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- R Bassan
- 1] UOC Ematologia, Ospedale dell'Angelo, Mestre-Venezia, Italy [2] USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - O Spinelli
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - E Oldani
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - T Intermesoli
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - M Tosi
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - B Peruta
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - E Borlenghi
- Divisione di Ematologia, Spedali Civili, Brescia, Italy
| | - E M Pogliani
- Clinica Ematologica, Ospedale San Gerardo, Monza, Italy
| | - E Di Bona
- UOC Ematologia, Ospedale S. Bortolo, Vicenza, Italy
| | - V Cassibba
- Divisione di Ematologia, Ospedale Civile, Bolzano, Italy
| | - A M Scattolin
- UOC Ematologia, Ospedale dell'Angelo, Mestre-Venezia, Italy
| | - C Romani
- UO Ematologia, Ospedale Oncologico di Riferimento Regionale, Cagliari, Italy
| | - F Ciceri
- UO Ematologia, Istituto Scientifico H.S. Raffaele, Milano, Italy
| | - A Cortelezzi
- UO Ematologia I, IRCCS Ca' Granda Ospedale Maggiore Policlinico and Università degli Studi, Milano, Italy
| | - G Gianfaldoni
- Ematologia, Università di Firenze, AOU Careggi, Firenze, Italy
| | - D Mattei
- SC Ematologia, ASO S. Croce e Carle, Cuneo, Italy
| | - E Audisio
- Ematologia 2, Ospedale San Giovanni Battista/Università, Torino, Italy
| | - A Rambaldi
- USC Ematologia, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
109
|
Shah NN, Borowitz MJ, Robey NC, Gamper CJ, Symons HJ, Loeb DM, Wayne AS, Chen AR. Feasibility of treating post-transplantation minimal residual disease in children with acute leukemia. Biol Blood Marrow Transplant 2014; 20:1000-7. [PMID: 24680975 PMCID: PMC4077899 DOI: 10.1016/j.bbmt.2014.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/19/2014] [Indexed: 12/11/2022]
Abstract
Outcomes are poor for patients with hematologic malignancies who experience overt relapse after allogeneic hematopoietic stem cell transplantation (HCT). Data on outcomes of post-transplantation minimal residual disease (MRD) are limited. In this single-institution, retrospective cohort analysis of children with acute leukemia and myelodysplastic syndrome, we document the pattern of relapse with a primary focus on outcomes of post-transplantation MRD. Forty of 93 patients (43%) who underwent a first allogeneic HCT and had systematic pretransplantation and post-transplantation MRD evaluations at +30, +60, +90, +180 days and +1 and +2 years post-transplantation experienced relapse. The median time to relapse was 4.8 months post-transplantation, with a median survival of 4 months post-relapse. Despite frequent, systematic, routine post-HCT disease restaging evaluation, 31 patients (78%) presented with overt disease at the time of relapse. Seven patients with acute leukemia who had post-transplantation MRD presented at a median of 1 month post-transplantation. Owing to rapid disease progression or treatment-related mortality, there was no improvement in survival in those patients whose leukemia was detected in a state of MRD post-transplantation. Our results suggest that early intervention strategies targeting post-transplantation MRD for relapse prevention in acute leukemia may not be feasible.
Collapse
Affiliation(s)
- Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland.
| | | | - Nancy C Robey
- Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | | | - Heather J Symons
- Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | - David M Loeb
- Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Alan S Wayne
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Allen R Chen
- Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
110
|
Tucunduva L, Ruggeri A, Sanz G, Furst S, Cornelissen J, Linkesch W, Mannone L, Ribera JM, Veelken H, Yakoub-Agha I, González Valentín ME, Schots R, Arcese W, Montesinos P, Labopin M, Gluckman E, Mohty M, Rocha V. Impact of minimal residual disease on outcomes after umbilical cord blood transplantation for adults with Philadelphia-positive acute lymphoblastic leukaemia: an analysis on behalf of Eurocord, Cord Blood Committee and the Acute Leukaemia working party of the European group for Blood and Marrow Transplantation. Br J Haematol 2014; 166:749-57. [PMID: 24961645 DOI: 10.1111/bjh.12970] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 04/21/2014] [Indexed: 11/30/2022]
Abstract
The status of umbilical cord blood transplantation (UCBT) in adults with Philadelphia-positive acute lymphoblastic leukaemia (Ph+ALL) and the impact of minimal residual disease (MRD) before transplant are not well established. We analysed 98 patients receiving UCBT for Ph+ALL in first (CR1) or second (CR2) complete remission (CR1, n = 79; CR2, n = 19) with MRD available before UCBT (92% analysed by reverse transcription polymerase chain reaction). Median age was 38 years and median follow-up was 36 months; 63% of patients received myeloablative conditioning and 42% received double-unit UCBT. Eighty-three patients were treated with at least one tyrosine kinase inhibitor before UCBT. MRD was negative (-) in 39 and positive (+) in 59 patients. Three-year cumulative incidence of relapse was 34%; 45% in MRD+ and 16% in MRD- patients (P =0·013). Three-year cumulative incidence of non-relapse mortality was 31%; it was increased in patients older than 35 years (P = 0·02). Leukaemia-free survival (LFS) at 3 years was 36%; 27% in MRD+ and 49% in MRD- patients (P = 0·05), and 41% for CR1 and 14% for CR2 (P = 0·008). Multivariate analysis identified only CR1 as being associated with improved LFS. In conclusion, MRD+ before UCBT is associated with increased relapse. Strategies to decrease relapse in UCBT recipients with Ph+ALL and MRD+ are needed.
Collapse
Affiliation(s)
- Luciana Tucunduva
- Eurocord APHP, University Paris-Diderot, Hospital Saint Louis, Paris, France; Centro de Oncologia, Hospital Sirio Libanes, Sao Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Pulsipher MA, Wayne AS, Schultz KR. New frontiers in pediatric Allo-SCT: novel approaches for children and adolescents with ALL. Bone Marrow Transplant 2014; 49:1259-65. [PMID: 24933210 DOI: 10.1038/bmt.2014.114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/06/2014] [Accepted: 02/13/2014] [Indexed: 11/09/2022]
Abstract
Although most children with ALL can be cured by chemotherapy approaches, allogeneic hematopoietic cell transplant (HCT) therapy offers a better chance of cure to selected high-risk patients in first remission and most children who relapse. Although transplant-related mortality has decreased significantly in the past decade, relapse remains high after HCT for ALL; developing strategies to decrease relapse and improve survival are vital. Recent studies have shown that relapse risk can be accurately defined using measurements of minimal residual disease (MRD) both pre- and post-HCT and by knowing whether patients get GVHD in the first 2 months after transplant. With these risk definitions in hand, investigators are now applying novel agents and immunotherapeutic methods in attempt to lower MRD before transplant and modulate the GVL effect after transplant. With powerful new immunological approaches coming on line, the transplant process itself will likely expand to include pre and/or post-HCT interventions aimed at reducing relapse.
Collapse
Affiliation(s)
- M A Pulsipher
- Division of Hematology and Hematological Malignancies, Primary Children's Hospital, University of Utah School of Medicine/Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - A S Wayne
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, The Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - K R Schultz
- Department of Pediatrics, University of BC, BC Children's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
112
|
Early assessment of minimal residual disease in AML by flow cytometry during aplasia identifies patients at increased risk of relapse. Leukemia 2014; 29:377-86. [PMID: 24912430 DOI: 10.1038/leu.2014.186] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 05/11/2014] [Accepted: 05/27/2014] [Indexed: 01/28/2023]
Abstract
In acute myeloid leukemia (AML), assessment of minimal residual disease (MRD) by flow cytometry (flow MRD) after induction and consolidation therapy has been shown to provide independent prognostic information. However, data on the value of earlier flow MRD assessment are lacking. Therefore, the value of flow MRD detection was determined during aplasia in 178 patients achieving complete remission after treatment according to AMLCG (AML Cooperative Group) induction protocols. Flow MRD positivity during aplasia predicted poor outcome (5-year relapse-free survival (RFS) 16% vs 43%, P<0.001) independently from age and cytogenetic risk group (hazard ratio for MRD positivity 1.71; P=0.009). Importantly, the prognosis of patients without detectable MRD was neither impacted by morphological blast count during aplasia nor by MRD status postinduction. Early flow MRD was also evaluated in the context of existing risk factors. Flow MRD was prognostic within the intermediate cytogenetic risk group (5-year RFS 15% vs 37%, P=0.016) as well as for patients with normal karyotype and NPM1 mutations (5-year RFS 13% vs 49%, P=0.02) or FLT3-ITD (3-year RFS rates 9% vs 44%, P=0.016). Early flow MRD assessment can improve current risk stratification approaches by prediction of RFS in AML and might facilitate adaptation of postremission therapy for patients at high risk of relapse.
Collapse
|
113
|
Walter RB, Gyurkocza B, Storer BE, Godwin CD, Pagel JM, Buckley SA, Sorror ML, Wood BL, Storb R, Appelbaum FR, Sandmaier BM. Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation. Leukemia 2014; 29:137-44. [PMID: 24888275 PMCID: PMC4254901 DOI: 10.1038/leu.2014.173] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/01/2014] [Accepted: 05/07/2014] [Indexed: 12/02/2022]
Abstract
Minimal residual disease (MRD) is associated with adverse outcome in AML after myeloablative (MA) hematopoietic cell transplantation (HCT). We compared this association with that seen after nonmyeloablative (NMA) conditioning in 241 adults receiving NMA (n=86) or MA (n=155) HCT for AML in first remission with pre-HCT bone marrow aspirates assessed by flow cytometry. NMA patients were older and had more comorbidities and secondary leukemias. Three-year relapse estimates were 28% and 57% for MRDneg and MRDpos NMA patients, and 22% and 63% for MA patients. Three-year overall survival (OS) estimates were 48% and 41% for MRDneg and MRDpos NMA patients and 76% and 25% for MA patients. This similar OS after NMA conditioning was largely accounted for by higher non-relapse mortality (NRM) in MRDneg (30%) compared to MRDpos (10%) patients, whereas the reverse was found for MRDneg (7%) and MRDpos (23%) MA patients. A statistically significant difference between MA and NMA patients in the association of MRD with OS (P<0.001) and NRM (P=0.002) but not relapse (P=0.17) was confirmed. After adjustment, the risk of relapse was 4.51-times (P<0.001) higher for MRDpos patients. These data indicate that the negative impact of MRD on relapse risk is similar after NMA and MA conditioning.
Collapse
Affiliation(s)
- R B Walter
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA [3] Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - B Gyurkocza
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - B E Storer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - C D Godwin
- Department of Medicine, Residency Program, University of Washington, Seattle, WA, USA
| | - J M Pagel
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - S A Buckley
- Department of Medicine, Residency Program, University of Washington, Seattle, WA, USA
| | - M L Sorror
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - B L Wood
- Division of Hematopathology, Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - R Storb
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - F R Appelbaum
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - B M Sandmaier
- 1] Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA [2] Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
114
|
Faulk K, Gore L, Cooper T. Overview of therapy and strategies for optimizing outcomes in de novo pediatric acute myeloid leukemia. Paediatr Drugs 2014; 16:213-27. [PMID: 24639021 DOI: 10.1007/s40272-014-0067-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Although acute myelogenous leukemia (AML) accounts for <20 % of leukemia in children, it is responsible for over half of all pediatric leukemia deaths. Improvement in event-free survival rates, now over 50 %, are due largely to intensification of chemotherapy, aggressive supportive care, development of risk stratification based on cytogenetic and molecular markers, and improved salvage regimens. Despite this improvement over the past few decades, the survival rates have recently plateaued, and further improvement will need to take into account advances in molecular characterization of AML, development of novel agents, and better understanding of host factors influencing toxicity and response to chemotherapy. This article reviews the epidemiology and biology trends in diagnosis and treatment of pediatric acute myelogenous leukemia.
Collapse
Affiliation(s)
- Kelly Faulk
- Department of Pediatrics, University of Colorado School of Medicine/Anschutz Medical Campus, Aurora, CO, USA
| | | | | |
Collapse
|
115
|
Balduzzi A, Di Maio L, Silvestri D, Songia S, Bonanomi S, Rovelli A, Conter V, Biondi A, Cazzaniga G, Valsecchi MG. Minimal residual disease before and after transplantation for childhood acute lymphoblastic leukaemia: is there any room for intervention? Br J Haematol 2014; 164:396-408. [PMID: 24422724 DOI: 10.1111/bjh.12639] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/18/2013] [Indexed: 11/26/2022]
Abstract
Eighty-two children and adolescents who underwent allogeneic transplantation for acute lymphoblastic leukaemia in remission (period 2001-2011, median follow-up 4·9 years) had been assessed for minimal residual disease (MRD) by real-time quantitative polymerase chain reaction before and at 1, 3, 6, 9 and 12 months after transplantation. Five-year event-free survival (EFS) and cumulative incidence of relapse were 77·7% [standard error (SE) 5·7] and 11·4% (SE 4·4), respectively, for patients with pre-transplant MRD <1 × 10(-4) (68%), versus 30·8% (SE 9·1; P < 0·001) and 61·5% (SE 9·5; P < 0·001), respectively, for those with MRD ≥1 × 10(-4) (32%). Pre-transplant MRD ≥1 × 10(-4) was associated with a 9·2-fold risk of relapse [95% confidence interval (CI) 3·54-23·88; P < 0·001] compared with patients with MRD <1 × 10(-4). Patients who received additional chemotherapy pre-transplant to reduce MRD had a fivefold reduction of risk of failure (hazard ratio 0·19, CI 0·05-0·70, P = 0·01). Patients who experienced MRD positivity post-transplant did not necessarily relapse (5-year EFS 40·3%, SE 9·3), but had a 2·5-fold risk of failure (CI 1·05-5·75; P = 0·04) if any MRD was detected in the first 100 d, which increased to 7·8-fold (CI 2·2-27·78; P = 0·002) if detected after 6 months. Anticipated immunosuppression-tapering according to MRD may have improved outcome, nevertheless all patients with post-transplant MRD ≥1 × 10(-3) ultimately relapsed, regardless of immunosuppression discontinuation or donor-lymphocyte-infusion. In conclusion, MRD before transplantation had the strongest impact on relapse and MRD positivity after transplantation, mostly if detected early and at low levels, did not necessarily imply relapse. Additional intensified chemotherapy and modulation of immunosuppression may reduce relapse risk and improve ultimate outcome.
Collapse
Affiliation(s)
- Adriana Balduzzi
- Clinica Pediatrica, Università degli Studi di Milano-Bicocca, Ospedale San Gerardo, Monza, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Kaspers G. How I treat paediatric relapsed acute myeloid leukaemia. Br J Haematol 2014; 166:636-45. [PMID: 24837715 DOI: 10.1111/bjh.12947] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/10/2014] [Indexed: 11/28/2022]
Abstract
The prognosis of paediatric acute myeloid leukaemia (AML) has improved significantly over the recent decades, but still about one-third of patients relapse. These patients have a relatively poor prognosis, with a probability of long-term survival from relapse of about 35%. This can only be achieved with very intensive chemotherapy and, usually, allogeneic stem cell transplantation, leading to very significant toxicity and even treatment-related mortality. Major improvements in the treatment of paediatric relapsed AML thus are required still, and several possibilities are discussed. In case of a suspected relapse, a comprehensive diagnostic work-up has to be undertaken, because significant changes in the biological features of the AML cells may have occurred between initial diagnosis and relapse. This review discusses many practical issues that one encounters in the treatment of children with relapsed AML. It will also be of interest for those involved in translational research in AML.
Collapse
Affiliation(s)
- Gertjan Kaspers
- Paediatric Oncology/Haematology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
117
|
Talano JM, Pulsipher MA, Symons HJ, Militano O, Shereck EB, Giller RH, Hancock L, Morris E, Cairo MS. New frontiers in pediatric Allo-SCT. Bone Marrow Transplant 2014; 49:1139-45. [PMID: 24820213 DOI: 10.1038/bmt.2014.89] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 01/02/2023]
Abstract
The inaugural meeting of 'New Frontiers in Pediatric Allogeneic Stem Cell Transplantation' organized by the Pediatric Blood and Transplant Consortium (PBMTC) was held at the American Society of Pediatric Hematology and Oncology Annual Meeting. This meeting provided an international platform for physicians and investigators active in the research and utilization of pediatric Allo-SCT in children and adolescents with malignant and non-malignant disease (NMD), to share information and develop future collaborative strategies. The primary objectives of the conference included: (1) to present advances in Allo-SCT in pediatric ALL and novel pre and post-transplant immunotherapy; (2) to highlight new strategies in alternative allogeneic stem cell donor sources for children and adolescents with non-malignant hematological disorders; (3) to discuss timing of immune reconstitution after Allo-SCT and methods of facilitating more rapid recovery of immunity; (4) to identify strategies of utilizing Allo-SCT in pediatric myeloproliferative disorders; (5) to develop diagnostic and therapeutic approaches to hematological complications post pediatric Allo-SCT; (6) to enhance the understanding of new novel cellular therapeutic approaches to pediatric malignant and non-malignant hematological disorders; and (7) to discuss optimizing drug therapy in pediatric recipients of Allo-SCT. This paper will provide a brief overview of the conference.
Collapse
Affiliation(s)
- J M Talano
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M A Pulsipher
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - H J Symons
- Department of Oncology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - O Militano
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - E B Shereck
- Oregon Health and Science University, Portland, OR, USA
| | - R H Giller
- Children's Hospital Colorado, Aurora, CO, USA
| | - L Hancock
- Pediatric Blood and Marrow Transplant Consortium, Monrovia, CA, USA
| | - E Morris
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - M S Cairo
- 1] Department of Pediatrics, New York Medical College, Valhalla, NY, USA [2] Department of Medicine, New York Medical College, Valhalla, NY, USA [3] Department of Pathology, New York Medical College, Valhalla, NY, USA [4] Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, USA [5] Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
118
|
Hasle H. A critical review of which children with acute myeloid leukaemia need stem cell procedures. Br J Haematol 2014; 166:23-33. [PMID: 24749666 DOI: 10.1111/bjh.12900] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/03/2014] [Indexed: 12/21/2022]
Abstract
The last decades have seen parallel improvements in chemotherapy-based and haematopoietic stem cell transplantation (HSCT) regimens for acute myeloid leukaemia (AML) in children. There has been no consensus on indication for HSCT. Reserving HSCT for high-risk and relapsed patients spare many patients from the long-term toxicity of this treatment. The results of matched unrelated donor HSCT equal family donor transplantation and the presence of a matched sibling should no longer be a transplant indication. Minimal residual disease measured by flow cytometry may identify poor responders benefitting from HSCT in first complete remission (CR1) and those with a favourable response to induction therapy who do not need HSCT even with adverse cytogenetic aberrations. FLT3-internal tandem duplication without NPM1 mutation has a very high relapse rate despite favourable response and HSCT is indicated in CR1 in these cases. Finding the optimal indications for HSCT is a delicate balance between risk of relapse and late effects.
Collapse
Affiliation(s)
- Henrik Hasle
- Department of Paediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark
| |
Collapse
|
119
|
Abstract
Survival rates for children with acute myeloid leukemia (AML) exceed 60 % when modern, intensified chemotherapeutic regimens and enhanced supportive care measures are employed. Despite well-recognized improvements in outcomes, primary refractory or relapsed pediatric AML yields significant morbidity and mortality, and improved understanding of this obstinate population along with refined treatment protocols are urgently needed. Although a significant number of patients with refractory or relapsed disease will achieve remission, long-term survival rates remain poor, and efforts to identify therapies which will improve OS are under continuous investigation. The current fundamental goal of such investigation is the achievement of as complete a remission as possible without dose-limiting toxicities, and the progression to hematopoietic stem cell transplantation thereafter. In this review the scope of the problem of relapsed and refractory AML as well as current and emerging chemotherapy options will be discussed.
Collapse
|
120
|
Factors predictive of relapse of acute leukemia in children after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2014; 20:1033-9. [PMID: 24691222 DOI: 10.1016/j.bbmt.2014.03.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 03/25/2014] [Indexed: 12/17/2022]
Abstract
The presence of minimal residual disease (MRD) before transplantation is the most important prognostic risk factor predictive of post-transplantation relapse in hematologic malignancies. However, MRD alone does not adequately predict relapse in all patients. To improve upon the ability to identify patients likely to relapse, we evaluated risk factors, in addition to MRD, that may be associated with development of post-transplantation relapse. In this single institution, retrospective cohort study of children with acute leukemia or myelodysplastic syndrome who had undergone a first allogeneic transplantation and had pretransplantation MRD evaluation, 40 of 93 patients (43%) experienced relapse. Univariate analysis demonstrated that African American race, high initial white blood cell count, central nervous system (CNS) disease at diagnosis, short first complete remission, nonmyeloablative (NMA) conditioning, lack of remission, and MRD before transplantation were associated with worse relapse-free survival (RFS). In a Cox multivariable analysis, CNS disease (P = .009), lack of remission (P = .01), and NMA conditioning (P = .04) were independently associated with inferior RFS. Among those in a morphologic complete remission who underwent a myeloablative transplantation, having both CNS disease at diagnosis (specifically in acute lymphoblastic leukemia) and MRD positivity was an independent risk factor predictive of relapse, which has not been previously reported. Results from our study support the existence of risk factors complimentary to pretransplantation MRD. Validation in a larger independent homogenous cohort is needed to develop a prognostic tool for clinical use to predict post-transplantation relapse.
Collapse
|
121
|
Pulsipher MA, Langholz B, Wall DA, Schultz KR, Bunin N, Carroll WL, Raetz E, Gardner S, Gastier-Foster JM, Howrie D, Goyal RK, Douglas JG, Borowitz M, Barnes Y, Teachey DT, Taylor C, Grupp SA. The addition of sirolimus to tacrolimus/methotrexate GVHD prophylaxis in children with ALL: a phase 3 Children's Oncology Group/Pediatric Blood and Marrow Transplant Consortium trial. Blood 2014; 123:2017-25. [PMID: 24497539 PMCID: PMC3968388 DOI: 10.1182/blood-2013-10-534297] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/08/2014] [Indexed: 11/20/2022] Open
Abstract
Sirolimus has activity against acute lymphoblastic leukemia (ALL) in xenograft models and efficacy in preventing acute graft-versus-host disease (aGVHD). We tested whether addition of sirolimus to GVHD prophylaxis of children with ALL would decrease aGVHD and relapse. Patients were randomized to tacrolimus/methotrexate (standard) or tacrolimus/methotrexate/sirolimus (experimental). The study met futility rules for survival after enrolling 146 of 259 patients. Rate of Grade 2-4 aGVHD was 31% vs 18% (standard vs experimental, P = .04), however, grade 3-4 aGVHD was not different (13% vs 10%, P = .28). Rates of veno-occlusive disease (VOD) and thrombotic microangiopathy (TMA) were lower in the nonsirolimus arm (9% vs 21% VOD, P = .05; 1% vs 10% TMA, P = .06). At 2 years, event free survival (EFS) and overall survival (OS) were 56% vs 46%, and 65% vs 55% (standard vs experimental), respectively (P = .28 and .23). Multivariate analysis showed increased relapse risk in children with ≥0.1% minimal residual disease (MRD) pretransplant, and decreased risk in patients with grades 1-3 aGVHD (P = .04). Grades 1-3 aGVHD were associated with improved EFS (P = .02), whereas grade 4 aGVHD and extramedullary disease at diagnosis led to inferior OS. Although addition of sirolimus decreased aGVHD, survival was not improved. This study is registered with ClinicalTrials.gov as #NCT00382109.
Collapse
Affiliation(s)
- Michael A Pulsipher
- Division of Hematology and Hematological Malignancies, Huntsman Cancer Institute/University of Utah School of Medicine, Primary Children's Hospital, Salt Lake City, UT
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Impact of minimal residual disease, detected by flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute lymphoblastic leukemia. LEUKEMIA RESEARCH AND TREATMENT 2014; 2014:421723. [PMID: 24778882 PMCID: PMC3981457 DOI: 10.1155/2014/421723] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/28/2014] [Accepted: 02/09/2014] [Indexed: 01/16/2023]
Abstract
In this retrospective study, we evaluated the impact of pre- and posttransplant minimal residual disease (MRD) detected by multiparametric flow cytometry (MFC) on outcome in 160 patients with ALL who underwent myeloablative allogeneic hematopoietic cell transplantation (HCT). MRD was defined as detection of abnormal B or T cells by MFC with no evidence of leukemia by morphology (<5% blasts in marrow) and no evidence of extramedullary disease. Among 153 patients who had pre-HCT flow data within 50 days before transplant, MRD pre-HCT increased the risk of relapse (hazard ratio (HR) = 3.64; 95% confidence interval (CI), 1.87-7.09; P = .0001) and mortality (HR = 2.39; 95% CI, 1.46-3.90, P = .0005). Three-year estimates of relapse were 17% and 38% and estimated 3-year OS was 68% and 40% for patients without and with MRD pre-HCT, respectively. 144 patients had at least one flow value post-HCT, and the risk of relapse among those with MRD was higher than that among those without MRD (HR = 7.47; 95% CI, 3.30-16.92, P < .0001). The risk of mortality was also increased (HR = 3.00; 95% CI, 1.44-6.28, P = .004). These data suggest that pre- or post-HCT MRD, as detected by MFC, is associated with an increased risk of relapse and death after myeloablative HCT for ALL.
Collapse
|
123
|
Ommen HB, Hokland P, Haferlach T, Abildgaard L, Alpermann T, Haferlach C, Kern W, Schnittger S. Relapse kinetics in acute myeloid leukaemias with MLL translocations or partial tandem duplications within the MLL gene. Br J Haematol 2014; 165:618-28. [PMID: 24611505 DOI: 10.1111/bjh.12792] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/10/2014] [Indexed: 01/03/2023]
Abstract
Correct action upon re-emergence of minimal residual disease in acute myeloid leukaemia (AML) patients has not yet been established. The applicability of demethylating agents and use of allogeneic stem cell transplantation will be dependent on pre-relapse AML growth rates. We here delineate molecular growth kinetics of AML harbouring MLL partial tandem duplication (MLL-PTD; 37 cases) compared to those harbouring MLL translocations (43 cases). The kinetics of MLL-PTD relapses was both significantly slower than those of MLL translocation positive ones (median doubling time: MLL-PTD: 24 d, MLL-translocations: 12 d, P = 0·015, Wilcoxon rank sum test), and displayed greater variation depending on additional mutations. Thus, MLL-PTD+ cases with additional RUNX1 mutations or FLT3-internal tandem duplication relapsed significantly faster than cases without one of those two mutations (Wilcoxon rank sum test, P = 0·042). As rapid relapses occurred in all MLL subgroups, frequent sampling are necessary to obtain acceptable relapse detection rates and times from molecular relapse to haematological relapse (blood sampling every second month: MLL-PTD: 75%/50 d; MLL translocations: 85%/25 d). In conclusion, in this cohort relapse kinetics is heavily dependent on AML subtype as well as additional genetic aberrations, with possibly great consequences for the rational choice of pre-emptive therapies.
Collapse
Affiliation(s)
- Hans B Ommen
- Department of Haematology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Gandemer V, Pochon C, Oger E, Dalle JHH, Michel G, Schmitt C, de Berranger E, Galambrun C, Cavé H, Cayuela JM, Grardel N, Macintyre E, Margueritte G, Méchinaud F, Rorhlich P, Lutz P, Demeocq F, Schneider P, Plantaz D, Poirée M, Bordigoni P. Clinical value of pre-transplant minimal residual disease in childhood lymphoblastic leukaemia: the results of the French minimal residual disease-guided protocol. Br J Haematol 2014; 165:392-401. [DOI: 10.1111/bjh.12749] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 12/06/2013] [Indexed: 01/24/2023]
Affiliation(s)
- Virginie Gandemer
- Department of Paediatric Haematology/Oncology; University Hospital of Rennes; Rennes France
| | - Cécile Pochon
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| | - Emmanuel Oger
- Clinical Pharmacology Department; Pharmacoepidemiology Team; University Hospital of Rennes; Rennes France
| | - Jean-Hugues H. Dalle
- Department of Paediatric Haematology; University Hospital of Robert Debré; Paris France
| | - Gérard Michel
- Department of Paediatric Haematology; University Hospital of La Timone; Marseille France
| | - Claudine Schmitt
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| | - Eva de Berranger
- Department of Paediatric Haematology/Oncology; University Hospital of Jeanne de Flandre; Lille France
| | - Claire Galambrun
- Department of Paediatric Haematology; Hospices Civils de Lyon; Lyon France
| | - Hélène Cavé
- Department of Genetics; University Hospital of Robert Debré and Paris-Diderot University; Paris France
| | | | - Nathalie Grardel
- Laboratory of Haematology; University Hospital of Calmette; Lille France
| | | | - Geneviève Margueritte
- Department of Paediatric Haematology/Oncology; University Hospital of Villeneuve; Montpellier France
| | - Françoise Méchinaud
- Department of Paediatric Haematology/Oncology; University Hospital of Nantes; Nantes France
| | - Pierre Rorhlich
- Department of Paediatric Haematology/Oncology; University Hospital of Besançon; Besançon France
| | - Patrick Lutz
- Department of Paediatric Haematology/Oncology; University Hospital of Hautepierre; Strasbourg France
| | - François Demeocq
- Department of Paediatric Haematology/oncology; University Hospital of Clermont-Ferrand; Clermont-Ferrand France
| | - Pascale Schneider
- Department of Paediatric Haematology; University Hospital of Rouen; Rouen France
| | - Dominique Plantaz
- Department of Paediatric Haematology/oncology; University Hospital of La Tronche; Grenoble France
| | - Marilyne Poirée
- Department of Paediatric Haematology/oncology; University Hospital of Archet II; Nice France
| | - Pierre Bordigoni
- Department of Paediatric Haematology/oncology; University Hospital of Nancy; Nancy France
| |
Collapse
|
125
|
Zhou Y, Slack R, Jorgensen JL, Wang SA, Rondon G, de Lima M, Shpall E, Popat U, Ciurea S, Alousi A, Qazilbash M, Hosing C, O'Brien S, Thomas D, Kantarjian H, Medeiros LJ, Champlin RE, Kebriaei P. The effect of peritransplant minimal residual disease in adults with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2014; 14:319-26. [PMID: 24548609 DOI: 10.1016/j.clml.2014.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/05/2014] [Accepted: 01/06/2014] [Indexed: 12/19/2022]
Abstract
BACKGROUND Allogeneic HSCT is highly effective for treating ALL. However, many ALL patients relapse after HSCT. There has been a continuing effort to improve identification of patients at high risk of relapse, with the goal of early intervention to improve outcome. PATIENTS AND METHODS In this retrospective analysis, we examined the effect of MRD on the risk of hematologic relapse in 149 adult patients with ALL in morphologic remission undergoing allogeneic HSCT. MRD was assessed at the time of HSCT and after HSCT. RESULTS Patients with pretransplant MRD had a trend for shorter progression-free survival (PFS) at 2 years compared with patients without MRD, nearing statistical significance; 28% versus 47%, P = .08, on univariate analysis. This trend remained on multivariate analysis with better PFS in patients without MRD at the time of HSCT, hazard ratio (HR), 0.62 (95% confidence interval, 0.37-1.04); P = .07. Additionally, emergence of MRD after HSCT was a strong predictor for overt hematologic relapse (HR, 4; P < .001) with a median latency interval of 3.8 months. CONCLUSION These findings demonstrate the predictive value of monitoring for MRD around the time of transplant in adult patients with ALL.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Hematopathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Rebecca Slack
- Department of Biostatistics, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Sa A Wang
- Department of Hematopathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Gabriela Rondon
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Marcos de Lima
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Stefan Ciurea
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Muzaffar Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Chitra Hosing
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Susan O'Brien
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Deborah Thomas
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
126
|
A clofarabine-based bridging regimen in patients with relapsed ALL and persistent minimal residual disease (MRD). Bone Marrow Transplant 2013; 49:440-2. [PMID: 24317126 DOI: 10.1038/bmt.2013.195] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/06/2013] [Accepted: 10/21/2013] [Indexed: 11/09/2022]
Abstract
In patients with relapsed ALL, minimal residual disease (MRD) identified prior to allogeneic hematopoietic cell transplantation (HCT) is a strong predictor of relapse. We report our experience using a combination of reduced-dosing clofarabine, CY and etoposide as a 'bridge' to HCT in eight patients with high risk or relapsed ALL and pre-HCT MRD. All patients had detectable MRD (>0.01%, flow cytometry) at the start of therapy with all eight achieving MRD reduction following one cycle. The regimen was well tolerated with seven grade 3/4 toxicities occurring among four of the eight patients. Five patients (62.5%) are alive, one died from relapse (12.5%) and two from transplant-related mortality (25%). The combination of reduced-dose clofarabine, CY and etoposide as bridging therapy appears to be well tolerated in patients with relapsed ALL and is effective in reducing pre-HCT MRD.
Collapse
|
127
|
Abstract
SUMMARY Predictive/prognostic factors in acute leukemia continue to be sought, in order to refine treatment strategies. Minimal residual disease (MRD) testing has been shown to be a statistically significant factor by multivariate analysis in both acute lymphoblastic leukemia (ALL) and acute myeloid leukemia. Its utility in guiding therapy has been more extensively studied in pediatric ALL, with some protocols having instituted MRD testing into therapeutic algorithms. The clinical impact of MRD testing in ALL and acute myeloid leukemia will be presented, including both molecular and flow cytometric methodologies, with a more focused discussion of the strategy, methodology and interpretation of MRD testing by multiparametric flow cytometry.
Collapse
Affiliation(s)
- Lorinda Soma
- University of Washington, Department of Laboratory Medicine, Division of Hematopathology, Room NW120, Box 357110, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Brent Wood
- University of Washington, Department of Laboratory Medicine, Division of Hematopathology, Room NW120, Box 357110, 1959 NE Pacific Street, Seattle, WA 98195, USA
| |
Collapse
|
128
|
Wang L, Gao L, Xu S, Gong S, Chen L, Lü S, Chen J, Qiu H, Xu X, Ni X, Song X, Zhang W, Yang J, Liu M, Hu X, Wang J. FISH+CD34+CD38- cells detected in newly diagnosed acute myeloid leukemia patients can predict the clinical outcome. J Hematol Oncol 2013; 6:85. [PMID: 24517186 PMCID: PMC4028871 DOI: 10.1186/1756-8722-6-85] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/16/2013] [Indexed: 01/16/2023] Open
Abstract
Background In acute myeloid leukemia (AML), the leukemia initiating cells (LICs) or leukemia stem cells (LSCs) is found within the CD34+CD38- cell compartment. The LICs subpopulation survives chemotherapy and is most probable the cause of minimal residual disease (MRD), which in turn is thought to cause relapse. The aim of this study was to determine the prognostic value of the percentage of LICs in blasts at diagnosis. Design and methods The percentage of LICs in the blast population was determined at diagnosis using a unique Flow-FISH analysis, which applies fluorescent in situ hybridization (FISH) analysis on flow cytometry sorted cells to distinguish LICs within the CD34+CD38- cell compartment. Fourty-five AML patients with FISH-detectable cytogenetic abnormalities treated with standardized treatment program were retrospectively included in the study. Correlations with overall survival (OS), events-free survival (EFS) and cumulative incidence of relapse (CIR) were evaluated with univariate and multivariate analysis. Results The percentage of LICs is highly variable in patients with acute myeloid leukemia, ranged from 0.01% to 52.8% (median, 2.1%). High LIC load (≥1%) negatively affected overall survival (2-year OS: 72.57% vs. 16.75%; P = 0.0037) and events-free survival (2-year EFS: 67.23% vs. 16.33%; P = 0.0018), which was due to an increased cumulative incidence of relapse (2-year CIR: 56.7% vs. 18.0%; P = 0.021). By multivariate analysis, high LIC load retained prognostic significance for OS and EFS. Conclusions In the present study, we established the Flow-FISH protocol as a useful method to distinguish normal and leukemic cells within the CD34+CD38- cell subpopulation. The high percentage of LICs at diagnosis was significantly correlated with increased risk of poor clinical outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Xiaoxia Hu
- Institute of Hematology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | | |
Collapse
|
129
|
Is minimal residual disease monitoring clinically relevant in adults with acute myelogenous leukemia? Curr Hematol Malig Rep 2013; 8:109-15. [PMID: 23563936 DOI: 10.1007/s11899-013-0157-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the past year, there has been increasing attention towards understanding the clinical relevance of minimal residual disease (MRD) assessment. The monitoring of MRD levels at various stages of therapy has considerable potential to impact the guidance of treatment for AML patients and improve outcomes. Thus, efforts have increased to address important concerns regarding MRD measurements. These concerns include: (1) what should be monitored; (2) what methodologies should be used; (3) whether such methodologies are standardized across laboratories; (4) how prognostic levels are defined; (5) when MRD should be monitored; and (6) what treatment options are available for MRD positive patients. In this review, we will discuss the methodologies available for MRD and the studies available to date aiming to address the concerns around the use of MRD measurements for AML patients.
Collapse
|
130
|
Mateos MK, O’Brien TA, Oswald C, Gabriel M, Ziegler DS, Cohn RJ, Russell SJ, Barbaric D, Marshall GM, Trahair TN. Transplant-related mortality following allogeneic hematopoeitic stem cell transplantation for pediatric acute lymphoblastic leukemia: 25-year retrospective review. Pediatr Blood Cancer 2013; 60:1520-7. [PMID: 23733511 PMCID: PMC3798104 DOI: 10.1002/pbc.24559] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/15/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND Over the last 25 years, donor source, conditioning, graft-versus-host disease prevention and supportive care for children undergoing hematopoeitic stem cell transplantation (HSCT) have changed dramatically. HSCT indications for acute lymphoblastic leukemia (ALL) now include high-risk patients in first and subsequent remission. There is a large burden of infectious and pre-HSCT morbidities, due to myelosuppressive therapy required for remission induction. We hypothesized that, despite these trends, overall survival (OS) had increased. PROCEDURE A retrospective audit of allogeneic pediatric HSCT for ALL was performed in our institution over 25 years. Outcomes for 136 HSCTs were analyzed in three consecutive 8-year periods (Period 1: 1/1/1984-31/8/1992, Period 2: 1/9/1992-30/4/2001, Period 3: 1/5/2001-31/12/2009). RESULTS Despite a significant increase in unrelated donor HSCT, event-free and OS over 25 years improved significantly. (EFS 31.6-64.8%, P = 0.0027; OS 41.8-78.9%, P < 0.0001) Concurrently, TRM dropped from 33% to 5% (P = 0.0004) whilst relapse rate was static (P = 0.07). TRM reduced significantly for matched sibling and unrelated cord blood transplantation (UCT) in Period 3 compared with earlier periods (P = 0.036, P = 0.0098, respectively). Factors leading to improved survival in patients undergoing UCT include better matching, higher total nucleated cell doses, and significantly faster neutrophil engraftment. Length of initial HSCT admission was similar over time. CONCLUSION EFS and OS have increased significantly despite heightened HSCT complexity. This survival gain was due to TRM reduction. Contemporary patients have benefited from refined donor selection and improved supportive care. Overall rates of leukemic relapse post-HSCT are unchanged, and remain the focus for improvement.
Collapse
Affiliation(s)
- Marion K Mateos
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Tracey A O’Brien
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Cecilia Oswald
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia
| | | | - David S Ziegler
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Richard J Cohn
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Susan J Russell
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Draga Barbaric
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia
| | - Glenn M Marshall
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia
| | - Toby N Trahair
- Kids Cancer Centre, Sydney Children’s HospitalRandwick, NSW, Australia,School of Women and Children’s Health, University of New South WalesNSW, Australia,* Correspondence to: Toby N. Trahair, Pediatric Hematologist/Oncologist Sydney Children’s Hospital, Randwick, NSW, Australia 2031., E-mail:
| |
Collapse
|
131
|
Risk-adapted donor lymphocyte infusion based on chimerism and donor source in pediatric leukemia. Blood Cancer J 2013; 3:e137. [PMID: 23995046 PMCID: PMC3763390 DOI: 10.1038/bcj.2013.39] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 11/08/2022] Open
Abstract
Donor lymphocyte infusion (DLI) is commonly used to treat leukemia relapse following stem cell transplantation. In florid relapse, however, the efficacy of DLI is limited with substantial risk of severe graft-versus-host disease (GvHD). Here, we develop a novel risk-adapted strategy characterized by pre-emptive DLI initiated at the time of mixed chimerism, a small starting dose based on donor source, dose-escalation guided by real-time chimerism monitoring and withholding of DLI immediately in patients achieving full donor chimerism. A total of 178 DLIs were given to 38 patients with mixed chimerism; thereafter, 33 patients (86.8%) had donor chimerism successfully increased, including 30 (78.9%) who had chimerism fully converted back to 100% donor. Cumulative incidence of relapse was significantly lower (P=0.00004) and overall survival higher (P=0.0003) in patients with chimerism fully corrected as compared with those of patients whose chimerism remained mixed. Only 13.2% of the patients developed acute grade III-IV GvHD with no associated mortality. In conclusion, the risk-adapted DLI strategy is useful in minimizing the risk of childhood leukemia relapse, GvHD and death.
Collapse
|
132
|
Ustun C, Wiseman AC, Defor TE, Yohe S, Linden MA, Oran B, Burke M, Warlick E, Miller JS, Weisdorf D. Achieving stringent CR is essential before reduced-intensity conditioning allogeneic hematopoietic cell transplantation in AML. Bone Marrow Transplant 2013; 48:1415-20. [PMID: 23933764 DOI: 10.1038/bmt.2013.124] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/09/2013] [Accepted: 07/10/2013] [Indexed: 12/22/2022]
Abstract
Reduced-intensity conditioning (RIC) allogeneic hematopoietic cell transplantation (allo-HCT) can cure patients with AML in CR. However, relapse after RIC allo-HCT may indicate heterogeneity in the stringency of CR. Strict definition of CR requires no evidence of leukemia by both morphologic and flow cytometric criteria. We re-evaluated 85 AML patients receiving RIC allo-HCT in CR to test if a strict definition of CR had direct implications for the outcome. These patients had leukemia immunophenotype documented at diagnosis and analyzed at allo-HCT. Eight (9.4%) had persistent leukemia by flow cytometric criteria at allo-HCT. The patients with immunophenotypic persistent leukemia had a significantly increased relapse (hazard ratio (HR): 3.7; 95% confidence interval (CI): 1.3-10.3, P=0.01) and decreased survival (HR: 2.9; 95% CI: 1.3-6.4, P<0.01) versus 77 patients in CR by both morphology and flow cytometry. However, the pre-allo-HCT bone marrow (BM) blast count (that is, 0-4%) was not significantly associated with risks of relapse or survival. These data indicate the presence of leukemic cells, but not the BM blast count affects survival. A strict morphologic and clinical lab flow cytometric definition of CR predicts outcomes after RIC allo-HCT, and therefore is critical to achieve at transplantation.
Collapse
Affiliation(s)
- C Ustun
- 1] Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA [2] Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood 2013; 122:1813-21. [PMID: 23847197 DOI: 10.1182/blood-2013-06-506725] [Citation(s) in RCA: 298] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Minimal residual disease (MRD) before myeloablative hematopoietic cell transplantation (HCT) is associated with adverse outcome in acute myeloid leukemia (AML) in first complete remission (CR1). To compare this association with that for patients in second complete remission (CR2) and to examine the quantitative impact of MRD, we studied 253 consecutive patients receiving myeloablative HCT for AML in CR1 (n = 183) or CR2 (n = 70) who had pre-HCT marrow aspirates analyzed by 10-color flow cytometry. Three-year estimates of overall survival were 73% (64%-79%) and 32% (17%-48%) for MRDneg and MRDpos CR1 patients, respectively, and 73% (57%-83%) and 44% (21%-65%) for MRDneg and MRDpos CR2 patients, respectively. Similar estimates of relapse were 21% (14%-28%) and 58% (41%-72%) for MRDneg and MRDpos CR1 patients, respectively, and 19% (9%-31%) and 68% (41%-85%) for MRDneg and MRDpos CR2 patients, respectively. Among the MRDpos patients, there was no statistically significant evidence that increasing levels of MRD were associated with increasing risks of relapse and death. After multivariable adjustment, risks of death and relapse were 2.61 times and 4.90 times higher for MRD(pos) patients (P < .001). Together, our findings indicate that the negative impact of pre-HCT MRD is similar for AML in CR1 and CR2 with even minute levels (≤ 0.1%) as being associated with adverse outcome.
Collapse
|
134
|
Abstract
PURPOSE OF REVIEW In patients with acute myeloid leukemia (AML), measuring response to treatment is essential to guide clinical decisions. Methods for detecting disease beyond the resolution limit of morphology (i.e., minimal residual disease, MRD) are now widely available. We here discuss their merits and the results of side-to-side comparisons. RECENT FINDINGS The ever-increasing comprehension of the molecular genetics of AML has led to the identification of targets for molecular monitoring of MRD in the majority of AML cases. Likewise, virtually all cases express aberrant immunophenotypes suitable for MRD monitoring by flow cytometry, a progress bolstered by powerful new-generation instruments. The clinical significance of MRD monitoring by either approach has been corroborated by recent results. However, with few exceptions, most of the studies continue to rely on retrospectively determined cut-off levels and time points. Moreover, when applied in parallel, the two approaches have yielded contradictory results. SUMMARY MRD monitoring can help predicting the risk of relapse better than morphology and also provide endpoints for clinical testing of experimental agents. MRD can be applied to guide therapy but one must carefully consider the characteristics of the methods used and the degree of expertise of the laboratory performing the test.
Collapse
|
135
|
Abstract
With steadily improved cure rates for children with newly diagnosed acute lymphoblastic leukaemia (ALL), treating relapsed ALL has become increasingly challenging largely due to resistance to salvage therapy. Improved biological understanding of mechanisms of relapse and drug resistance, the identification of actionable molecular targets by studying leukaemic cell and host genetics, precise risk stratification with minimum residual disease measurement, and the development of new therapeutic drugs and approaches are needed to improve outcomes of relapsed patients. Molecularly targeted therapies and innovative immunotherapeutic approaches that include specialised monoclonal antibodies and cellular therapies hold promise of enhanced leukaemia cell killing with non-overlapping toxicities. Advances in preparative regimens, donor selection, and supportive care should improve the success of haemopoietic stem-cell transplantation for high-risk patients.
Collapse
Affiliation(s)
- Deepa Bhojwani
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38015, USA.
| | | |
Collapse
|
136
|
Abstract
Acute lymphoblastic leukaemia occurs in both children and adults but its incidence peaks between 2 and 5 years of age. Causation is multifactorial and exogenous or endogenous exposures, genetic susceptibility, and chance have roles. Survival in paediatric acute lymphoblastic leukaemia has improved to roughly 90% in trials with risk stratification by biological features of leukaemic cells and response to treatment, treatment modification based on patients' pharmacodynamics and pharmacogenomics, and improved supportive care. However, innovative approaches are needed to further improve survival while reducing adverse effects. Prognosis remains poor in infants and adults. Genome-wide profiling of germline and leukaemic cell DNA has identified novel submicroscopic structural genetic changes and sequence mutations that contribute to leukaemogenesis, define new disease subtypes, affect responsiveness to treatment, and might provide novel prognostic markers and therapeutic targets for personalised medicine.
Collapse
Affiliation(s)
- Hiroto Inaba
- Department of Oncology, St Jude Children's Research Hospital and University of Tennessee Health Science Center, Memphis, TN 38105, USA.
| | | | | |
Collapse
|
137
|
Campana D, Leung W. Clinical significance of minimal residual disease in patients with acute leukaemia undergoing haematopoietic stem cell transplantation. Br J Haematol 2013; 162:147-61. [DOI: 10.1111/bjh.12358] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 03/08/2013] [Indexed: 01/04/2023]
Affiliation(s)
- Dario Campana
- Department of Paediatrics; Yong Loo Lin School of Medicine; National University of Singapore; Singapore Singapore
| | - Wing Leung
- Department of Bone Marrow Transplantation and Cellular Therapy; St Jude Children's Research Hospital; Memphis TN USA
- Department of Pediatrics; College of Medicine; University of Tennessee Health Science Center; Memphis TN USA
| |
Collapse
|
138
|
Estey EH. Acute myeloid leukemia: 2013 update on risk-stratification and management. Am J Hematol 2013; 88:318-27. [PMID: 23526416 DOI: 10.1002/ajh.23404] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 01/23/2013] [Indexed: 12/11/2022]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) results from accumulation of abnormal blasts in the marrow. These cells interfere with normal hematopoiesis, can escape into the peripheral blood, and infiltrate CSF and lung. It is likely that many different mutations, epigenetic aberrations, or abnormalities in micro RNA expression can produce the same morphologic disease with these differences responsible for the very variable response to therapy, which is AMLs principal feature. DIAGNOSIS This rests on demonstration that the marrow or blood has > 20% blasts of myeloid lineage. Blast lineage is assessed by multiparameter flow cytometry, with CD33 and CD13 being surface markers typically expressed by myeloid blasts. It should be realized that clinical/prognostic considerations, not the blast % per se, should be the main factor determining how a patient is treated. RISK STRATIFICATION Two features determine risk: the probability of treatment-related mortality (TRM) and, more important, even in patients aged >75 with Zubrod performance status 1, the probability of resistance to standard therapy despite not incurring TRM. The chief predictor of resistance is cytogenetics, with a monosomal karyotype (MK) denoting the disease is essentially incurable with standard therapy even if followed by a standard allogeneic transplant (HCT). The most common cytogentic finding is a normal karyotype(NK) and those of such patients with an NPM1 mutation but no FLT3 internal tandem duplication (ITD), or with a CEBPA mutation, have a prognosis similar to that of patients with the most favorable cytogenetics (inv 16 or t[8;21]) (60-70% cure rate). In contrast, NK patients with a FLT3 ITD have only a 30-40% chance of cure even after HCT. Accordingly analyses of NPM1, FLT3, and CEBPA should be part of routine evaluation, much as is cytogenetics. Risk is best assessed considering several variables simultaneously rather than, for example, only age. Increasing evidence indicates that other mutations and abnormalities in microRNA (miRNA) expression also affect resistance as do post treatment factors, in particular the presence of minimal residual disease. These newer mutations and MRD are discussed in this update. RISK-ADAPTED THERAPY Patients with inv (16) or t(8;21) or who are NPM1+/FLT3ITD-can receive standard therapy (daunorubicin + cytarabine) and should not receive HCT in first CR. It seems likely that use of a daily daunorubicin dose of 90 mg/m(2) will further improve outcome in these patients. There appears no reason to use doses of cytarabine > 1 g/m(2) (for example bid X 6 days), as opposed to the more commonly used 3 g/m(2) . Patients with an unfavorable karyotype (particularly MK) are unlikely to benefit from standard therapy (even with dose escalation) and are thus prime candidates for clinical trials of new drugs or new approaches to HCT; the latter should be done in first CR. Patients with intermediate prognoses (for example NK and NPM and FLT3ITD negative) should also receive HCT in first CR and can plausibly receive either investigational or standard induction therapy, with the same prognostic information about standard therapy leading one patient to choose the standard and another an investigational option. This update discusses results with newer agents: quizartinib and crenolanib, gemtuzumab ozogamicin, clofarabine and cladribine, azacitidine and decitabine, volasertib, and means to prevent relapse after allogeneic transplant. The diagnosis of AML essentially is made as it was in 2012. Thus this review will emphasize new developments in risk stratification and treatment using as references many papers published in 2012.
Collapse
Affiliation(s)
- Elihu H. Estey
- Division of Hematology; University of Washington
- Clinical Research Division; Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
139
|
Fagioli F, Quarello P, Zecca M, Lanino E, Rognoni C, Balduzzi A, Messina C, Favre C, Foà R, Ripaldi M, Rutella S, Basso G, Prete A, Locatelli F. Hematopoietic stem cell transplantation for children with high-risk acute lymphoblastic leukemia in first complete remission: a report from the AIEOP registry. Haematologica 2013; 98:1273-81. [PMID: 23445874 DOI: 10.3324/haematol.2012.079707] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Children with high-risk acute lymphoblastic leukemia in first complete remission can benefit from allogeneic hematopoietic stem cell transplantation. We analyzed the outcome of 211 children with high-risk acute lymphoblastic leukemia in first complete remission who were given an allogeneic transplant between 1990 and 2008; the outcome of patients who, despite having an indication for transplantation and a suitable donor, did not receive the allograft for different reasons in the same time period was not analyzed. Sixty-nine patients (33%) were transplanted between 1990 and 1999, 58 (27%) between 2000 and 2005, and 84 (40%) between 2005 and 2008. A matched family donor was employed in 138 patients (65%) and an unrelated donor in 73 (35%). The 10-year probabilities of overall and disease-free survival were 63.4% and 61%, respectively. The 10-year cumulative incidences of transplantation-related mortality and relapse were 15% and 24%, respectively. After 1999, no differences in either disease-free survival or transplant-related mortality were observed in patients transplanted from unrelated or matched family donors. In multivariate analysis, grade IV acute graft-versus-host disease was an independent factor associated with worse disease-free survival. By contrast, grade I acute graft-versus-host disease and age at diagnosis between 1 and 9 years were favorable prognostic variables. Our study, not intended to evaluate whether transplantation is superior to chemotherapy for children with acute lymphoblastic leukemia in first complete remission and high-risk features, shows that the allograft cured more than 60% of these patients; in the most recent period, the outcome of recipients of grafts from matched family and unrelated donors was comparable.
Collapse
Affiliation(s)
- Franca Fagioli
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Torino.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Campana D, Coustan-Smith E. Measurements of treatment response in childhood acute leukemia. THE KOREAN JOURNAL OF HEMATOLOGY 2012; 47:245-54. [PMID: 23320002 PMCID: PMC3538795 DOI: 10.5045/kjh.2012.47.4.245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 11/14/2012] [Indexed: 12/31/2022]
Abstract
Measuring response to chemotherapy is a backbone of the clinical management of patients with acute leukemia. This task has historically relied on the ability to identify leukemic cells among normal bone marrow cells by their morphology. However, more accurate ways to identify leukemic cells have been developed, which allow their detection even when they are present in small numbers that would be impossible to be recognized by microscopic inspection. The levels of such minimal residual disease (MRD) are now widely used as parameters for risk assignment in acute lymphoblastic leukemia (ALL) and increasingly so in acute myeloid leukemia (AML). However, different MRD monitoring methods may produce discrepant results. Moreover, results of morphologic examination may be in stark contradiction to MRD measurements, thus creating confusion and complicating treatment decisions. This review focusses on the relation between results of different approaches to measure response to treatment and define relapse in childhood acute leukemia.
Collapse
Affiliation(s)
- Dario Campana
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
141
|
Abstract
Abstract
After approximately 20 years of development and after several prospective clinical trials, the detection of minimal residual disease (MRD) has emerged as part of state-of-the-art diagnostics to guide the majority of contemporary treatment programs both in pediatric and adult acute lymphoblastic leukemia (ALL). For ALL, several methods of MRD analysis are available, but 2 are widely applicable. One is based on the detection of aberrant expression of leukemia specific antigens by flow cytometry and the other one uses the specific rearrangements of the TCR or Ig genes, which can be detected by quantitative PCR in the DNA of leukemic cells. In some cases with known fusion genes such as BCR/ABL, RT-PCR can be used as a third method of identifying leukemic cells by analyzing RNA in patient samples. Clinical application of such sophisticated tools in the stratification and treatment of ALL requires reliable, reproducible, and quality-assured methods to ensure patient safety.
Collapse
|
142
|
Abstract
Abstract
The achievement of complete hematologic remission (CR) is a prerequisite for cure in acute myeloid leukemia (AML). The conventional definition of CR, based on the morphologic recognition of ≤ 5% of leukemic blasts in the BM, does not provide sufficient insight into the quality of the response. Despite CR rates of 50%-80% (depending on age), the majority of patients with AML relapse within 3-5 years from diagnosis. Therefore, there is great need of more sensitive prognostic factors that can predict relapse. Minimal residual disease (MRD), defined as any measurable disease or leukemia detectable above a certain threshold (defined by the methodology applied), predicts failure to maintain a morphologic CR and affects survival negatively. AML is lagging behind acute lymphoblastic leukemia with respect to the implementation of MRD criteria for guidance during therapy. AML is particularly disadvantaged compared with acute lymphoblastic leukemia in that approximately half of AML patients lack a molecular target suitable for MRD monitoring. The detection of altered antigen (Ag) expression by leukemic myeloblasts is a valid alternative to DNA- or RNA-based MRD assays. Although associated with presenting prognostic factors (eg, cytogenetics and genotype), MRD represents the collective end result of all of the cellular mechanisms that determine a patient's response to a given therapy. Therefore, MRD has 2 potential roles in AML treatment: (1) as a posttherapy prognosticator used to assign patients to optimal postinduction/consolidation therapy, and (2) as an early surrogate end point for the evaluation of therapy efficacy.
Collapse
|
143
|
Abstract
Only the most specific main institutional affiliation is listed for Inside Blood authors, without indications of subdivisions, departments, parent institutions, or postal/geographic information. Please confirm or correct affiliation. Minimal residual disease (MRD) detection is standard of care in acute lymphoblastic leukemia (ALL),(1) but not acute myeloid leukemia (AML). In this issue of Blood, an AML trial by Loken and colleagues from the Children's Oncology Group (COG) retrospectively demonstrates clinical significance of MRD.(2) These data reaffirm that AML-MRD is unlike ALL-MRD.
Collapse
|
144
|
Abstract
In the study reported in this issue of Blood by Leung et al, detectable minimal residual disease (MRD) before hematopoietic stem cell transplantation (HSCT) was found to be prognostic for outcome, but did not prevent cure for children with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML).
Collapse
|
145
|
Abstract
Although acute myeloid leukaemia (AML) has long been recognized for its morphological and cytogenetic heterogeneity, recent high-resolution genomic profiling has demonstrated a complexity even greater than previously imagined. This complexity can be seen in the number and diversity of genetic alterations, epigenetic modifications, and characteristics of the leukaemic stem cells. The broad range of abnormalities across different AML subtypes suggests that improvements in clinical outcome will require the development of targeted therapies for each subtype of disease and the design of novel clinical trials to test these strategies. It is highly unlikely that further gains in long-term survival rates will be possible by mere intensification of conventional chemotherapy. In this review, we summarize recent studies that provide new insight into the genetics and biology of AML, discuss risk stratification and therapy for this disease, and profile some of the therapeutic agents currently under investigation.
Collapse
Affiliation(s)
- Jeffrey E Rubnitz
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | |
Collapse
|
146
|
Abstract
The most common cause of treatment failure in childhood acute lymphoblastic leukemia (ALL) remains relapse, occurring in ~ 15%-20% of patients. Survival of relapsed patients can be predicted by site of relapse, length of first complete remission, and immunophenotype of relapsed ALL. BM and early relapse (< 30 months from diagnosis), as well as T-ALL, are associated with worse prognosis than isolated extramedullary or late relapse (> 30 months from diagnosis). In addition, persistence of minimal residual disease (MRD) at the end of induction or consolidation therapy predicts poor outcome because children with detectable MRD are more likely to relapse than those in molecular remission, even after allogeneic hematopoietic stem cell transplantation. We offer hematopoietic stem cell transplantation to any child with high-risk features because these patients are virtually incurable with chemotherapy alone. By contrast, we treat children with first late BM relapse of B-cell precursor ALL and good clearance of MRD with a chemotherapy approach. We use both systemic and local treatment for extramedullary relapse, mainly represented by radiotherapy and, in case of testicular involvement, by orchiectomy. Innovative approaches, including new agents or strategies of immunotherapy, are under investigation in trials enrolling patients with resistant or more advanced disease.
Collapse
|
147
|
Prognostic and therapeutic implications of minimal residual disease at the time of transplantation in acute leukemia. Bone Marrow Transplant 2012; 48:630-41. [PMID: 22825427 DOI: 10.1038/bmt.2012.139] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Relapse remains the major cause of treatment failure after hematopoietic cell transplantation (HCT) in acute leukemia, even in patients transplanted in morphologic CR. Various techniques now enable the sensitive quantification of 'minimal' amounts of residual disease (MRD) in patients with acute leukemia in remission. Numerous studies convincingly demonstrate that MRD at the time of transplantation is a powerful, independent predictor of subsequent relapse, with current detection levels of one leukemic cell in 10(5)-10(6) normal cells being prognostically relevant. This recognition provides the rationale to assign patients with detectable MRD (that is, 'MRD(+)' patients) to intensified therapies before, during, or after transplantation, although data supporting these strategies are still sparse. Limited evidence from observational studies suggests that outcomes with autologous HCT are so poor that MRD(+) patients should preferentially be assigned to allogeneic HCT, which can cure a subgroup of these patients, particularly if unmanipulated (T-cell replete) grafts and/or minimized immunosuppression are used to optimize the graft-vs-leukemia effect. Emerging data suggest that additional therapy with non-cross-resistant agents to decrease residual tumor burden before transplantation in MRD(+) patients might be beneficial. Further, other studies hint at immunotherapy (for example, rapid withdrawal of immunosuppression and/or donor lymphocyte infusions) as a means to prevent overt relapse if patients remain, or become, MRD(+) after HCT. Ultimately, controlled clinical studies are needed to define the value of MRD-directed therapies, and patients should be encouraged to enter such trials.
Collapse
|