1
|
Zhao Y, Siddiqi I, Wildes TJ, Charles D, Deak K, Wang E. Blast Phase of Myeloproliferative Neoplasm Resembles Acute Myeloid Leukemia, Myelodysplasia-Related, in Clinical Presentation, Cytogenetic Pattern, and Genomic Profile, and Often Undergoes Reversion to Second Chronic Phase Status After Induction Chemotherapy. Arch Pathol Lab Med 2024; 148:1310-1319. [PMID: 38426696 DOI: 10.5858/arpa.2023-0363-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2023] [Indexed: 03/02/2024]
Abstract
CONTEXT.— BCR::ABL-negative myeloproliferative neoplasm (MPN) has a prolonged clinical course, and some cases eventually undergo transformation to blast phase; its pathogenesis remains to be elucidated. OBJECTIVE.— To evaluate the clinicopathologic characteristics of MPN in blast phase. DESIGN.— The study aimed to retrospectively analyze the clinical and laboratory data of 24 MPN cases. RESULTS.— Median latency to blast phase was 48 months (range, 7-384 months). Complex karyotypes were seen in 12 of the 24 cases (50%). Overall, 16 cases (66.7%) exhibited high allele burdens of MPN driver mutations along with increased blasts, consistent with linear clonal evolution, whereas the remainder (8; 33.3%) showed loss or partial loss of the driver mutation, suggestive of a parallel evolution. Additional mutations were noted in 23 cases (100%), including TP53 mutations in 10 of 24 cases (41.7%). Following chemotherapy, 15 of the 24 patients (62.5%) reverted to a second chronic phase while retaining or regaining MPN driver mutations and losing blast-related mutations, although 9 of the 15 patients (60%) later died of disease progression. Median overall survival was 10 months (CI, 4.6-15.4), with those harboring complex karyotypes demonstrating decreased survival (6 versus 29 months; P = .004). CONCLUSIONS.— MPN blast phase resembles acute myeloid leukemia, myelodysplasia-related, in cytogenetic pattern, mutation profile, and clinical outcome. Two patterns of clonal evolution are inferred by dynamic analysis of mutation profiles: linear and parallel evolutions. Although overall survival was dismal, 62.5% of our cases achieved second chronic phase, and they showed better survival than those without second chronic phase.
Collapse
Affiliation(s)
- Yue Zhao
- From the Department of Pathology, College of Basic Medical Sciences and the First Hospital, China Medical University, Shenyang, China (Zhao)
- the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Zhao, Wildes, Charles, Deak)
| | - Imran Siddiqi
- the Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles (Siddiqi, Wang)
| | - Tyler J Wildes
- the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Zhao, Wildes, Charles, Deak)
| | - Derald Charles
- the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Zhao, Wildes, Charles, Deak)
| | - Kristen Deak
- the Department of Pathology, Duke University School of Medicine, Durham, North Carolina (Zhao, Wildes, Charles, Deak)
| | - Endi Wang
- the Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles (Siddiqi, Wang)
| |
Collapse
|
2
|
Prognosis of older patients with newly diagnosed AML undergoing antileukemic therapy: A systematic review. PLoS One 2022; 17:e0278578. [PMID: 36469519 PMCID: PMC9721486 DOI: 10.1371/journal.pone.0278578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVE The prognostic value of age and other non-hematological factors in predicting outcomes in older patients with newly diagnosed acute myeloid leukemia (AML) undergoing antileukemic therapy is not well understood. We performed a systematic review to determine the association between these factors and mortality and health-related quality of life or fatigue among these patients. METHODS We searched Medline and Embase through October 2021 for studies in which researchers quantified the relationship between age, comorbidities, frailty, performance status, or functional status; and mortality and health-related quality of life or fatigue in older patients with AML receiving antileukemic therapy. We assessed the risk of bias of the included studies using the Quality in Prognostic Studies tool, conducted random-effects meta-analyses, and assessed the quality of the evidence using the Grading of Recommendations, Assessment, Development and Evaluation approach. RESULTS We included 90 studies. Meta-analysis showed that age (per 5-year increase, HR 1.16 95% CI 1.11-1.21, high-quality evidence), comorbidities (Hematopoietic Cell Transplantation-specific Comorbidity Index: 3+ VS less than 3, HR 1.60 95% CI 1.31-1.95, high-quality evidence), and performance status (Eastern Cooperative Oncology Group/ World Health Organization (ECOG/WHO): 2+ VS less than 2, HR 1.63 95% CI 1.43-1.86, high-quality evidence; ECOG/WHO: 3+ VS less than 3, HR 2.00 95% CI 1.52-2.63, moderate-quality evidence) were associated with long-term mortality. These studies provided inconsistent and non-informative results on short-term mortality (within 90 days) and quality of life. CONCLUSION High-quality or moderate-quality evidence support that age, comorbidities, performance status predicts the long-term prognosis of older patients with AML undergoing antileukemic treatment.
Collapse
|
3
|
Rezaei M, Tan J, Zeng C, Li Y, Ganjalikhani-Hakemi M. TIM-3 in Leukemia; Immune Response and Beyond. Front Oncol 2021; 11:753677. [PMID: 34660319 PMCID: PMC8514831 DOI: 10.3389/fonc.2021.753677] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 02/05/2023] Open
Abstract
T cell immunoglobulin and mucin domain 3 (TIM-3) expression on malignant cells has been reported in some leukemias. In myelodysplastic syndrome (MDS), increased TIM-3 expression on TH1 cells, regulatory T cells, CD8+ T cells, and hematopoietic stem cells (HSCs), which play a role in the proliferation of blasts and induction of immune escape, has been reported. In AML, several studies have reported overexpression of TIM-3 on leukemia stem cells (LSCs) but not on healthy HSCs. Overexpression of TIM-3 on exhausted CD4+ and CD8+ T cells and leukemic cells in CML, ALL, and CLL patients could be a prognostic risk factor for poor therapeutic response and relapse in patients. Currently, several TIM-3 inhibitors are used in clinical trials for leukemias, and some have shown encouraging response rates for MDS and AML treatment. For AML immunotherapy, blockade TIM-3 may have dual effects: directly inhibiting AML cell proliferation and restoring T cell function. However, blockade of PD-1 and TIM-3 fails to restore the function of exhausted CD8+ T cells in the early clinical stages of CLL, indicating that the effects of TIM-3 blockade may be different in AML and other leukemias. Thus, further studies are required to evaluate the efficacy of TIM-3 inhibitors in different types and stages of leukemia. In this review, we summarize the biological functions of TIM-3 and its contribution as it relates to leukemias. We also discuss the effects of TIM-3 blockade in hematological malignancies and clinical trials of TIM-3 for leukemia therapy.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chengwu Zeng
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
4
|
Zhou X, Kuang Y, Liang S, Wang L. Metformin inhibits cell proliferation in SKM-1 cells via AMPK-mediated cell cycle arrest. J Pharmacol Sci 2019; 141:146-152. [PMID: 31744691 DOI: 10.1016/j.jphs.2019.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/11/2019] [Accepted: 10/24/2019] [Indexed: 02/08/2023] Open
Abstract
Metformin, a widely used antidiabetic drug, has previously been demonstrated to exert anti-cancer effects in certain hematological malignancies, but its effects on the transformation of myelodysplastic syndromes to acute myeloid leukemia (AML-MDS) remain unclear. The present study aimed to investigate the effects of metformin on SKM-1 cells (an AML-MDS cell line) and its underlying mechanisms. SKM-1 cells were treated with different concentrations of metformin. Cell proliferation was assayed by CCK-8. Apoptosis and cell cycle phases were detected by flow cytometry, while cell cycle related proteins and AMPK were tested by Western blot. SKM-1 cells were transfected with LV-AMPKα1-RNAi to reduce the expression of AMPK. Metformin inhibited cell proliferation in a dose and time dependent manner by inducing G0/G1 phase arrest rather than apoptosis induction. Metformin promoted the expression of p-AMPK, P53, P21CIP1 and P27KIP1, while inhibited the expression of CDK4 and CyclinD1. AMPK knockdown attenuated the effects of metformin on SKM-1 cells. These findings suggested that metformin inhibited proliferation of SKM-1 cells, potentially through an AMPK-mediated cell cycle arrest.
Collapse
Affiliation(s)
- Xiaojia Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunchun Kuang
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Simin Liang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
5
|
Pourmoussa A, Kwan K. An Unlikely Rapid Transformation of Myelodysplastic Syndrome to Acute Leukemia: A Case Report. Perm J 2018; 21:16-091. [PMID: 28488979 DOI: 10.7812/tpp/16-091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Myelodysplastic syndrome is characterized by stem-cell-derived clonal myelopoiesis with an alteration in proliferation and differentiation. This condition carries a potential for transformation to acute leukemia, primarily in cases that are accompanied by high-risk features at diagnosis. CASE PRESENTATION A 68-year-old man with recently diagnosed myelodysplastic syndrome and Sweet syndrome (acute febrile neutrophilic dermatosis) presented to our Emergency Department with shortness of breath. During his hospital course, he developed signs and symptoms, predominantly consisting of respiratory difficulties, that were not typically characteristic of transformation to acute leukemia. Several days into his hospitalization, it was determined that the patient's underlying hematologic process seemed to have rapidly evolved into an acute myeloid leukemia, which accounted for the progression of symptoms. This patient ultimately opted for comfort measures only and died shortly thereafter. DISCUSSION Two important factors stood out as representing an atypical presentation. First, this patient lacked any of the high-risk features of myelodysplastic syndrome that typically portend transformation. In addition, his progression to acute leukemia in 28 days from the time of diagnosis was far more rapid than the 274-day median previously described in the literature. We theorize that the presence of Sweet syndrome may have served as a predisposing factor to transformation. This finding may offer benefit to physicians to potentially better predict this outcome and pursue more aggressive treatment measures earlier in the course of the disease in such a setting.
Collapse
Affiliation(s)
| | - Karen Kwan
- Physician in the Department of Hematology and Oncology at the Los Angeles Medical Center in CA.
| |
Collapse
|
6
|
Dong W, Ding T, Wu L, Ren X, Epling-Burnette PK, Yang L. Effect of IL-7 and IL-15 on T cell phenotype in myelodysplastic syndromes. Oncotarget 2018; 7:27479-88. [PMID: 27036031 PMCID: PMC5053665 DOI: 10.18632/oncotarget.8459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/16/2016] [Indexed: 11/25/2022] Open
Abstract
Aberrant T cell phenotype is one of the characteristics of myelodysplastic syndromes (MDS). In this study, we detected an increased concentration of IL-15 in the plasma of MDS patients (n = 20) compared with that in the plasma of healthy controls (n = 20). In MDS patients, reduced naïve CD4+ and CD8+ T cells [16.11 ± 6.56 vs. 24.11 ± 7.18 for CD4+ T cells (p < 0.001) and 13.15 ± 5.67 vs. 23.51 ± 6.25 for CD8+ T cells (p < 0.001)] were observed. The reduced naïve and increased effector memory T cells were significantly correlated with IL-15 plasma level. Then, the effect of IL-15 and IL-7 was tested in vitro. Peripheral blood mononuclear cells from MDS were treated for 15 days with IL-15. This treatment significantly decreased naïve CD4+ (p < 0.001) and CD8+ (p < 0.001) T cells and correspondingly increased terminal memory CD4+ and CD8+ T cells (p < 0.001). Treatment with IL-7 increased naïve CD4+ (p < 0.05) and CD8+ (p < 0.001) T cells. Our results indicated that exposure to high levels of IL-15 may be involved in the T cell phenotype conversion observed in MDS. IL-7 may be one of the promising therapeutic candidates for recovering the effector immune compartment in MDS patients.
Collapse
Affiliation(s)
- Wen Dong
- Department of Orthopaedic Surgery, Tianjin Hongqiao Hospital, Tianjin, P.R. China
| | - Tingting Ding
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Lei Wu
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| | | | - Lili Yang
- Department of Immunology, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, P.R. China.,National Clinical Research Center of Cancer, P.R. China.,Tianjin Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P.R. China
| |
Collapse
|
7
|
Asayama T, Tamura H, Ishibashi M, Kuribayashi-Hamada Y, Onodera-Kondo A, Okuyama N, Yamada A, Shimizu M, Moriya K, Takahashi H, Inokuchi K. Functional expression of Tim-3 on blasts and clinical impact of its ligand galectin-9 in myelodysplastic syndromes. Oncotarget 2017; 8:88904-88917. [PMID: 29179486 PMCID: PMC5687656 DOI: 10.18632/oncotarget.21492] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
T-cell immunoglobulin mucin-3 (Tim-3), an inhibitory immune checkpoint receptor, is highly expressed on acute myeloid leukemia cells and its ligand galectin-9 is reported to drive leukemic progression by binding with Tim-3. However, it remains unclear whether the Tim-3–galectin-9 pathway is associated with the pathophysiology of myelodysplastic syndromes (MDS). Thus, we investigated the expression and function of Tim-3 and the clinical impact of its ligand galectin-9 in MDS. Tim-3 expression levels on MDS blasts by CD45/side-scatter or CD34/CD45 gating were increased as MDS progressed to the advanced stage. Tim-3 expression in the MDS blasts was upregulated in the presence of the cell culture supernatant of human stromal cells or the MDS-related cytokine transforming growth factor-β1. The proliferation of Tim-3+ MDS blasts was inhibited by the blockade of anti-Tim-3 antibody. Furthermore, plasma levels of galectin-9 were elevated as MDS progressed to the advanced stage in 70 MDS/acute leukemia transformed from MDS patients and was a prognostic factor in 40 MDS patients. Our data demonstrated that the Tim-3-galectin-9 pathway is associated with the pathogenesis and disease progression of MDS. These findings provide new insight into potential immunotherapy targeting the galectin-9–Tim-3 pathway in MDS.
Collapse
Affiliation(s)
- Toshio Asayama
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Hideto Tamura
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | | | | | | - Namiko Okuyama
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Akiko Yamada
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Keiichi Moriya
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Hidemi Takahashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Koiti Inokuchi
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
8
|
Enrico A, Bestach Y, Flores MG, Arbelbide J, Serale C, Novoa V, Crisp R, Rivas MM, Larripa I, Belli C. Influence of Acute Myeloid Leukemia Progression on the Prognosis of 831 Patients With Myelodysplastic Syndromes From the Argentine Database. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:743-752.e5. [PMID: 28797621 DOI: 10.1016/j.clml.2017.06.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 06/15/2017] [Accepted: 06/19/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND A large group of patients with myelodysplastic syndromes (MDS) will die of causes intrinsic to bone marrow failure. One third of patients will develop acute myeloid leukemia (AML), which is associated with an extremely poor outcome and a short survival. Our objectives were to analyze the prognostic variables and scoring systems in the attempt to determine the influence of progression on the overall survival of MDS patients. PATIENTS AND METHODS We performed a retrospective analysis of 831 MDS patients, including those from the Argentine Registry. RESULTS Of the 831 MDS patients, 158 (19.0%) experienced transformation, with a median overall survival of 17.9 months from diagnosis and 3.5 months after progression. The survival of patients with adverse karyotypes or greater risk, according to the International Prognostic Scoring System-revised (IPSS-R) or World Health Organization-based Prognostic Scoring System (WPSS) was not affected when stratified by patients with and without evolution to AML (P > .05). In contrast, the survival of lower risk patients was significantly reduced for those patients with progression to AML (P < .001) and those younger (P = .024) than those who died of non-AML-related causes. The intermediate-risk patients were heterogeneously distributed; however, an upgrade from a lower IPSS-R to a higher WPSS-hemoglobin risk category was associated with a worse outcome, not affected by progression (P = .420), with a median event-free survival of 16 months. CONCLUSION The use of the IPSS-R and WPSS systems simultaneously might help in identifying those patients who require more aggressive treatment. Nevertheless, more efforts are needed to improve the identification of those lower risk patients whose survival is significantly reduced by progression to AML.
Collapse
Affiliation(s)
- Alicia Enrico
- Area de Hematología, Hospital Italiano de La Plata, La Plata, Argentina
| | - Yesica Bestach
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas/Academia Nacional de Medicina, Buenos Aires City, Argentina
| | - Maria Gabriela Flores
- Servicio de Hematología, Hospital General de Agudos C Durand, Buenos Aires City, Argentina
| | - Jorge Arbelbide
- Servicio de Hematología, Hospital Italiano de Buenos Aires, Buenos Aires City, Argentina
| | - Camila Serale
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas/Academia Nacional de Medicina, Buenos Aires City, Argentina
| | - Viviana Novoa
- Servicio de Hematología, Hospital General de Agudos C Durand, Buenos Aires City, Argentina
| | - Renée Crisp
- Servicio de Hematología, Hospital Nacional A Posadas, El Palomar, Argentina
| | - María Marta Rivas
- Servicio de Hematología, Hospital Universitario Austral, Pilar Centro, Argentina
| | - Irene Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas/Academia Nacional de Medicina, Buenos Aires City, Argentina
| | - Carolina Belli
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas/Academia Nacional de Medicina, Buenos Aires City, Argentina.
| |
Collapse
|
9
|
Tan SF, Pearson JM, Feith DJ, Loughran TP. The emergence of acid ceramidase as a therapeutic target for acute myeloid leukemia. Expert Opin Ther Targets 2017; 21:583-590. [PMID: 28434262 DOI: 10.1080/14728222.2017.1322065] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is the most common adult leukemia. Only a fraction of AML patients will survive with existing chemotherapy regimens. Hence, there is an urgent and unmet need to identify novel targets and develop better therapeutics in AML. In the past decade, the field of sphingolipid metabolism has emerged into the forefront of cancer biology due to its importance in cancer cell proliferation and survival. In particular, acid ceramidase (AC) has emerged as a promising therapeutic target due to its role in neutralizing the pro-death effects of ceramide. Areas covered: This review highlights key information about AML biology as well as current knowledge on dysregulated sphingolipid metabolism in cancer and AML. We describe AC function and dysregulation in cancer, followed by a review of studies that report elevated AC in AML and compounds known to inhibit the enzyme. Expert opinion: AML has a great need for new drug targets and better therapeutic agents. The finding of elevated AC in AML supports the concept that this enzyme represents a novel and realistic therapeutic target for this common leukemia. More effort is needed towards developing better AC inhibitors for clinical use and combination treatment with existing AML therapies.
Collapse
Affiliation(s)
- Su-Fern Tan
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA
| | - Jennifer M Pearson
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA
| | - David J Feith
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA.,b University of Virginia Cancer Center , Charlottesville , VA , USA
| | - Thomas P Loughran
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA.,b University of Virginia Cancer Center , Charlottesville , VA , USA
| |
Collapse
|
10
|
Leinonen MK, Rantanen M, Pitkäniemi J, Malila N. Coverage and accuracy of myeloproliferative and myelodysplastic neoplasms in the Finnish Cancer Registry. Acta Oncol 2016; 55:782-6. [PMID: 26767306 DOI: 10.3109/0284186x.2015.1127416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Registration of haematological malignancies presents specific challenges, and a wide range of data is required to ensure case ascertainment and proper classification of these diseases. We studied the data quality of myeloproliferative and myelodysplastic neoplasms in the Finnish Cancer Registry (FCR), comparing information with hospital discharges. Material and methods Hospital discharges (HILMO) in 2007-2013 including diagnostic codes of myeloproliferative and myelodysplastic neoplasms were extracted. Patients were individually linked to the FCR database for all haematological malignancies registered in 1953-2013. Coverage and accuracy of the FCR and agreement between registers was estimated. Results In total 5289 individuals were retrieved from two registers. Of these, 1406 were common, 1080 only found in the FCR and 2803 only in the HILMO. Coverage of myeloproliferative and myelodysplastic neoplasms in the FCR was 47.0% (95% CI 45.7-48.4%). Almost one quarter of the registrations in the FCR was based on a death certificate only. The accuracy of diagnosis was 51.4% (95% CI 49.4-53.3%), but it varied substantially by disease category. Kappa statistic for agreement between registers was excellent (0.83, 95% CI 0.80-0.85) for common cases. 7.6% of cases in the HILMO was registered as leukaemias in the FCR. Conclusions More than half of the patients found in the HILMO were entirely missing from the FCR. However, some of the diagnoses in HILMO may be preliminary and this represents the maximal number of missing cases. Cancer registers benefit from supplementary data sources, such as hospital discharges, to increase coverage and accuracy of register data on haematological malignancies.
Collapse
Affiliation(s)
- Maarit K Leinonen
- Cancer Society of Finland, Finnish Cancer Registry, Helsinki, Finland
| | - Matti Rantanen
- Cancer Society of Finland, Finnish Cancer Registry, Helsinki, Finland
| | - Janne Pitkäniemi
- Cancer Society of Finland, Finnish Cancer Registry, Helsinki, Finland
| | - Nea Malila
- Cancer Society of Finland, Finnish Cancer Registry, Helsinki, Finland
| |
Collapse
|
11
|
Hu KX, Sun QY, Guo M, Qiao JX, Yu CL, Qiao JH, Dong Z, Sun WJ, Zuo HL, Huang YJ, Cai B, Ai HS. A Study of Human Leukocyte Antigen Mismatched Cellular Therapy (Stem Cell Microtransplantation) in High-Risk Myelodysplastic Syndrome or Transformed Acute Myelogenous Leukemia. Stem Cells Transl Med 2016; 5:524-9. [PMID: 26838271 DOI: 10.5966/sctm.2015-0196] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED The treatment outcomes of myelodysplastic syndrome (MDS) and transformed acute myelogenous leukemia (tAML) remain very unsatisfactory. We designed a combination of human leukocyte antigen (HLA)-mismatched hematopoietic stem cell microtransplantation (MST) with chemotherapy for patients with MDS and tAML and evaluated its effects and toxicity. Patients were between 13 and 79 years old. Patients with MDS (n=21) were given HLA-mismatched MST combined with decitabine and cytarabine; patients with tAML (n=22) were given HLA-mismatched MST combined with decitabine and cytarabine, and also mitoxantrone. Patients in complete remission (CR) also received MST plus decitabine and medium-dose cytarabine chemotherapy without graft-versus-host disease (GVHD) prophylaxis. The overall response rate of the patients with MDS was significantly higher than that of those with tAML (81% vs. 50%; p=.03). The CR rates were 52.4% and 36.4% in the two groups, respectively. There was no difference in the cytogenetic CR rate between the MDS and tAML groups (85.7% vs. 70%, respectively; p=.7). The 24-month overall survival of the patients with MDS was significantly higher than that of the patients with tAML (84.7% and 34.1%, respectively; p=.003). The median recovery times of neutrophils and platelets were, respectively, 14 and 17 days in the patients with MDS, and 16 and 19 days in those with tAML. The treatment-related mortality rates were 4.8% and 18.2%, respectively, in the MDS and tAML groups (p=.34). No GVHD was observed in any patient. Microtransplantation combined with decitabine and chemotherapy may provide a novel, effective, and safe treatment for high-risk MDS and tAML. SIGNIFICANCE Microtransplantation (MST) refers to regular chemotherapy combined with granulocyte colony-stimulating factor-mobilized peripheral blood stem cell infusion of human leukocyte antigen-mismatched donor cells without using immunosuppressive agents. It aims to support hematopoietic recovery and perform graft-versus-leukemia (GVL) effects but differs from traditional allogeneic stem cell transplantation because the rate of donor cell chimerism is low and there is and no graft-versus-host disease (GVHD) risk. Thus, a trial was designed to evaluate the safety and efficacy of MST in patients with myelodysplastic syndrome and those with transformed acute myelogenous leukemia. Higher complete remission and cytogenetic complete response rates were observed, and the treatment improved disease progress-free survival, sped hematopoietic recovery, and avoided GVHD.
Collapse
Affiliation(s)
- Kai-Xun Hu
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Qi-Yun Sun
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Mei Guo
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Jun-Xiao Qiao
- Department of Hematology, Second Artillery General Hospital, Beijing, People's Republic of China
| | - Chang-Lin Yu
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Jian-Hui Qiao
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Zheng Dong
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Wan-Jun Sun
- Department of Hematology, Second Artillery General Hospital, Beijing, People's Republic of China
| | - Hong-Li Zuo
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Ya-Jing Huang
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Bo Cai
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Hui-Sheng Ai
- Department of Hematology and Transplantation, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
12
|
Hasserjian RP, Campigotto F, Klepeis V, Fu B, Wang SA, Bueso-Ramos C, Cascio MJ, Rogers HJ, Hsi ED, Soderquist C, Bagg A, Yan J, Ochs R, Orazi A, Moore F, Mahmoud A, George TI, Foucar K, Odem J, Booth C, Morice W, DeAngelo DJ, Steensma D, Stone RM, Neuberg D, Arber DA. De novo acute myeloid leukemia with 20-29% blasts is less aggressive than acute myeloid leukemia with ≥30% blasts in older adults: a Bone Marrow Pathology Group study. Am J Hematol 2014; 89:E193-9. [PMID: 25042343 DOI: 10.1002/ajh.23808] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 11/11/2022]
Abstract
It is controversial whether acute myeloid leukemia (AML) patients with 20-29% bone marrow (BM) blasts, formerly referred to as refractory anemia with excess blasts in transformation (RAEBT), should be considered AML or myelodysplastic syndrome (MDS) for the purposes of treatment and prognostication. We retrospectively studied 571 de novo AML in patients aged >50 years, including 142 RAEBT and 429 with ≥30% blasts (AML30), as well as 151 patients with 10-19% BM blasts (RAEB2). RAEBT patients were older and had lower white blood count, but higher hemoglobin, platelet count, and karyotype risk scores compared to AML30, while these features were similar to RAEB2. FLT3 and NPM1 mutations and monocytic morphology occurred more commonly in AML30 than in RAEBT. RAEBT patients were treated less often with induction therapy than AML30, whereas allogeneic stem cell transplant frequency was similar. The median and 4-year OS of RAEBT patients were longer than those of AML30 patients (20.5 vs 12.0 months and 28.6% vs 20.4%, respectively, P = 0.003); this difference in OS was manifested in patients in the intermediate UKMRC karyotype risk group, whereas OS of RAEBT patients and AML30 patients in the adverse karyotype risk group were not significantly different. Multivariable analysis showed that RAEBT (P < 0.0001), hemoglobin (P = 0.005), UKMRC karyotype risk group (P = 0.002), normal BM karyotype (P = 0.004), treatment with induction therapy (P < 0.0001), and stem cell transplant (P < 0.0001) were associated with longer OS. Our findings favor considering de novo RAEBT as a favorable prognostic subgroup of AML.
Collapse
Affiliation(s)
| | - Federico Campigotto
- Department of Biostatistics and Computational Biology; Dana Farber Cancer Institute; Boston Massachusetts
| | - Veronica Klepeis
- Department of Pathology; Massachusetts General Hospital; Boston Massachusetts
| | - Bin Fu
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sa A. Wang
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Carlos Bueso-Ramos
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | | | | | - Craig Soderquist
- Department of Pathology; University of Pennsylvania; Philadelphia Pennsylvania
| | - Adam Bagg
- Department of Pathology; University of Pennsylvania; Philadelphia Pennsylvania
| | - Jiong Yan
- Department of Pathology; Weill Cornell Medical College; New York New Yort
| | - Rachel Ochs
- Department of Pathology; Weill Cornell Medical College; New York New Yort
| | - Attilio Orazi
- Department of Pathology; Weill Cornell Medical College; New York New Yort
| | - Frank Moore
- Department of Pathology; University of New Mexico Health Sciences Center; Albuquerque New Mexico
| | - Amer Mahmoud
- Department of Pathology; University of New Mexico Health Sciences Center; Albuquerque New Mexico
| | - Tracy Irene George
- Department of Pathology; University of New Mexico Health Sciences Center; Albuquerque New Mexico
| | - Kathryn Foucar
- Department of Pathology; University of New Mexico Health Sciences Center; Albuquerque New Mexico
| | - Jamie Odem
- Department of Pathology; Mayo Clinic; Rochester Minnesota
| | - Cassie Booth
- Department of Pathology; Mayo Clinic; Rochester Minnesota
| | - William Morice
- Department of Pathology; Mayo Clinic; Rochester Minnesota
| | - Daniel J. DeAngelo
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston Massachusetts
| | - David Steensma
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston Massachusetts
| | - Richard Maury Stone
- Department of Medical Oncology; Dana Farber Cancer Institute; Boston Massachusetts
| | - Donna Neuberg
- Department of Biostatistics and Computational Biology; Dana Farber Cancer Institute; Boston Massachusetts
| | | |
Collapse
|
13
|
Prognosis of secondary acute myeloid leukemia. Leuk Res 2013; 37:857-8. [DOI: 10.1016/j.leukres.2013.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 04/09/2013] [Accepted: 04/12/2013] [Indexed: 11/22/2022]
|