Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Blood Adv ; 8(10): 2592-2599, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38531057

ABSTRACT

ABSTRACT: Chimeric antigen receptor (CAR) T-cell (CAR-T) immunotherapy is an effective therapy for relapsed/refractory B-cell non-Hodgkin lymphoma (r/r B-NHL). However, data are limited on the impact of the convergence of race and social determinants of health on outcomes for patients treated with CAR-T therapy. We examined the impact of interactions between race and insurance type on health care use and outcomes in patients treated with CAR-T therapy for aggressive B-NHL. Adult patients with r/r B-NHL treated with CD19 CAR-Ts were identified between 2015 and 2021 across 13 US academic centers. Insurance type, demographic, and clinical data were collected and analyzed. In total, 466 adult patients were included in our analysis. Median follow-up after CAR-T therapy was 12.7 months. Median progression-free survival (mPFS) was longer for Caucasians (11.5 months) than for African Americans (3.5 months; hazard ratio [HR], 1.56 [1.03-2.4]; P = .04) or Asians (2.7 months; HR, 1.7 [1.02-2.67]; P = .04). Differences in median overall survival (mOS) were not significant. For Medicare (n = 206) vs Medicaid (n = 33) vs private insurance (n = 219) vs self-pay (n = 7): mPFS was 15.9 vs 4.2 vs 6.0 vs 0.9 months (P < .001), respectively; and mOS was 31.2 vs 12.8 vs 21.5 vs 3.2 months (P < .001), respectively. Our multicenter retrospective analysis showed that race and insurance status can affect outcomes for patients treated with CAR-T therapy.


Subject(s)
Immunotherapy, Adoptive , Social Determinants of Health , Humans , Male , Female , Middle Aged , Adult , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/mortality , Treatment Outcome , Aged , United States , Retrospective Studies , Racial Groups
4.
Clin Cancer Res ; 29(13): 2375-2384, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37036505

ABSTRACT

PURPOSE: Treatment options are limited beyond JAK inhibitors for patients with primary myelofibrosis (MF) or secondary MF. Preclinical studies have revealed that PI3Kδ inhibition cooperates with ruxolitinib, a JAK1/2 inhibitor, to reduce proliferation and induce apoptosis of JAK2V617F-mutant cell lines. PATIENTS AND METHODS: In a phase I dose-escalation and -expansion study, we evaluated the safety and efficacy of a selective PI3Kδ inhibitor, umbralisib, in combination with ruxolitinib in patients with MF who had a suboptimal response or lost response to ruxolitinib. Enrolled subjects were required to be on a stable dose of ruxolitinib for ≥8 weeks and continue that MTD at study enrollment. The recommended dose of umbralisib in combination with ruxolitinib was determined using a modified 3+3 dose-escalation design. Safety, pharmacokinetics, and efficacy outcomes were evaluated, and spleen size was measured with a novel automated digital atlas. RESULTS: Thirty-seven patients with MF (median age, 67 years) with prior exposure to ruxolitinib were enrolled. A total of 2 patients treated with 800 mg umbralisib experienced reversible grade 3 asymptomatic pancreatic enzyme elevation, but no dose-limiting toxicities were seen at lower umbralisib doses. Two patients (5%) achieved a durable complete response, and 12 patients (32%) met the International Working Group-Myeloproliferative Neoplasms Research and Treatment response criteria of clinical improvement. With a median follow-up of 50.3 months for censored patients, overall survival was greater than 70% after 3 years of follow-up. CONCLUSIONS: Adding umbralisib to ruxolitinib in patients was well tolerated and may resensitize patients with MF to ruxolitinib without unacceptable rates of adverse events seen with earlier generation PI3Kδ inhibitors. Randomized trials testing umbralisib in the treatment of MF should be pursued.


Subject(s)
Janus Kinase Inhibitors , Primary Myelofibrosis , Humans , Aged , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/metabolism , Phosphatidylinositol 3-Kinases , Pyrimidines/therapeutic use , Nitriles/therapeutic use , Janus Kinase Inhibitors/therapeutic use
5.
Blood Adv ; 7(12): 2657-2669, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-36094847

ABSTRACT

Most patients receiving chimeric antigen receptor T-cell therapy (CAR-T) for aggressive B-cell non-Hodgkin lymphoma (B-NHL) do not experience a durable remission. Several novel agents are approved to treat relapsed, refractory aggressive B-NHL; however, it remains unclear how to sequence these therapies pre- and post-CAR-T. We conducted a multicenter retrospective analysis to describe peri-CAR-T practice patterns and survival predictors for patients receiving CD19-directed CAR-T. Patients (n = 514) from 13 centers treated with CAR-T for B-NHL between 2015-2021 were included in the study. Survival curves were constructed using Kaplan-Meier method. Multivariate Cox regression analysis was used to determine the impact of the variables on survival outcomes. For all patients receiving CAR-T, a greater number of lines of therapy pre-CAR-T apheresis and bridging therapy were predictive of inferior progression-free survival (PFS) and overall survival (OS). The median PFS and OS from the time of CAR-T cell infusion were 7.6 and 25.6 months, respectively. From the time of progression post-CAR-T, the median OS was 5.5 months. The median PFS of treatments administered in the first-line post-CAR-T failure was 2.8 months. Patients with refractory disease on day 30 had inferior OS and were less likely to receive subsequent treatment(s) than other patients with CAR-T failure. Allogeneic hematopoietic cell transplantation for selected patients at any time following CAR-T failure led to durable responses in over half of patients at 1 year. These data provide a benchmark for future clinical trials in patients with post-CAR-T cell progression, which remains an unmet clinical need.


Subject(s)
Lymphoma, B-Cell , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Retrospective Studies , Immunotherapy, Adoptive/methods , Progression-Free Survival
7.
BMC Cancer ; 22(1): 1013, 2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36153475

ABSTRACT

BACKGROUND: Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) comprise several rare hematologic malignancies with shared concomitant dysplastic and proliferative clinicopathologic features of bone marrow failure and propensity of acute leukemic transformation, and have significant impact on patient quality of life. The only approved disease-modifying therapies for any of the MDS/MPN are DNA methyltransferase inhibitors (DNMTi) for patients with dysplastic CMML, and still, outcomes are generally poor, making this an important area of unmet clinical need. Due to both the rarity and the heterogeneous nature of MDS/MPN, they have been challenging to study in dedicated prospective studies. Thus, refining first-line treatment strategies has been difficult, and optimal salvage treatments following DNMTi failure have also not been rigorously studied. ABNL-MARRO (A Basket study of Novel therapy for untreated MDS/MPN and Relapsed/Refractory Overlap Syndromes) is an international cooperation that leverages the expertise of the MDS/MPN International Working Group (IWG) and provides the framework for collaborative studies to advance treatment of MDS/MPN and to explore clinical and pathologic markers of disease severity, prognosis, and treatment response. METHODS: ABNL MARRO 001 (AM-001) is an open label, randomly allocated phase 1/2 study that will test novel treatment combinations in MDS/MPNs, beginning with the novel targeted agent itacitinib, a selective JAK1 inhibitor, combined with ASTX727, a fixed dose oral combination of the DNMTi decitabine and the cytidine deaminase inhibitor cedazuridine to improve decitabine bioavailability. DISCUSSION: Beyond the primary objectives of the study to evaluate the safety and efficacy of novel treatment combinations in MDS/MPN, the study will (i) Establish the ABNL MARRO infrastructure for future prospective studies, (ii) Forge innovative scientific research that will improve our understanding of pathogenetic mechanisms of disease, and (iii) Inform the clinical application of diagnostic criteria, risk stratification and prognostication tools, as well as response assessments in this heterogeneous patient population. TRIAL REGISTRATION: This trial was registered with ClinicalTrials.gov on August 19, 2019 (Registration No. NCT04061421).


Subject(s)
Myelodysplastic-Myeloproliferative Diseases , Quality of Life , Acetonitriles , Cytidine Deaminase , DNA/therapeutic use , Decitabine/therapeutic use , Humans , Methyltransferases , Prospective Studies , Pyrazoles , Pyrimidines , Pyrroles , Syndrome
8.
J Hematol Oncol ; 15(1): 96, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842643

ABSTRACT

Ibrutinib is effective in the treatment of relapsed/refractory (R/R) marginal zone lymphoma (MZL) with an overall response rate (ORR) of 48%. However, factors associated with response (or lack thereof) to ibrutinib in R/R MZL in clinical practice are largely unknown. To answer this question, we performed a multicenter (25 US centers) cohort study and divided the study population into three groups: "ibrutinib responders"-patients who achieved complete or partial response (CR/PR) to ibrutinib; "stable disease (SD)"; and "primary progressors (PP)"-patients with progression of disease as their best response to ibrutinib. One hundred and nineteen patients met the eligibility criteria with 58%/17% ORR/CR, 29% with SD, and 13% with PP. The median PFS and OS were 29 and 71.4 months, respectively, with no difference in PFS or OS based on the ibrutinib line of therapy or type of therapy before ibrutinib. Patients with complex cytogenetics had an inferior PFS (HR = 3.08, 95% CI 1.23-7.67, p = 0.02), while those with both complex cytogenetics (HR = 3.00, 95% CI 1.03-8.68, p = 0.04) and PP (HR = 13.94, 95% CI 5.17-37.62, p < 0.001) had inferior OS. Only primary refractory disease to first-line therapy predicted a higher probability of PP to ibrutinib (RR = 3.77, 95% CI 1.15-12.33, p = 0.03). In this largest study to date evaluating outcomes of R/R MZL treated with ibrutinib, we show that patients with primary refractory disease and those with PP on ibrutinib are very high-risk subsets and need to be prioritized for experimental therapies.


Subject(s)
Lymphoma, B-Cell, Marginal Zone , Adenine/analogs & derivatives , Cohort Studies , Humans , Lymphoma, B-Cell, Marginal Zone/drug therapy , Lymphoma, B-Cell, Marginal Zone/pathology , Neoplasm Recurrence, Local/drug therapy , Piperidines , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Treatment Outcome
11.
Cancer ; 127(21): 3991-3997, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34289094

ABSTRACT

BACKGROUND: Aggressive large B-cell lymphomas (LBCLs) are curable, but previous studies have shown inferior outcomes in minorities. Nurse navigation programs can improve patient outcomes by providing patient support. This study presents the outcomes of White and minority patients with aggressive LBCL at an institution with an active nurse navigation program. METHODS: The authors prospectively collected baseline characteristics, treatment regimens, and outcome data for patients with aggressive LBCL. Navigation encounters were characterized as low or high intensity. Overall survival (OS) and progression-free survival (PFS) were calculated with Kaplan-Meier methods. Baseline characteristics were compared with Fisher exact tests. RESULTS: Two hundred four consecutive patients (47 minority patients and 157 White patients) were included. Results were presented as minorities versus Whites. There were no differences in prognostic scores (Revised International Prognostic Index score of 3-5, 43% vs 47%; P = .50), frontline chemotherapy (98% vs 96%; P = .68), or the incidence of relapsed/refractory disease (40% vs 38%; P = .74). For relapsed/refractory LBCL, similar proportions of patients underwent hematopoietic stem cell transplantation (32% vs 29%; P > .99) or chimeric antigen receptor T-cell therapy (16% vs 19%; P > .99). Enrollment in clinical trials was comparable (17% vs 14%; P = .64). More than 85% received nurse navigation, but minorities had higher intensity navigation encounters (42% vs 21%; P = .01). The 2-year OS rates were 81% and 76% for minorities and Whites, respectively (P = .27); the 2-year PFS rates were 62% and 65%, respectively (P = .78). CONCLUSIONS: This study shows similar survival between Whites and minorities with aggressive LBCL, which was likely due to equal access to guideline-concordant therapy. Minorities received higher intensity navigation encounters, which may have helped them to overcome socioeconomic disadvantages.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lymphoma, Large B-Cell, Diffuse , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Health Services Accessibility , Humans , Immunotherapy, Adoptive , Lymphoma, Large B-Cell, Diffuse/therapy , Progression-Free Survival , Retrospective Studies
12.
Onco Targets Ther ; 14: 2185-2201, 2021.
Article in English | MEDLINE | ID: mdl-33814917

ABSTRACT

Collectively, hematological malignancies account for the fourth most common malignancy. Myeloma and lymphoma are the most common types of hematological malignancies. Unfortunately, the management of refractory myeloma and lymphoma remains challenging. The discovery of new immunological therapies, namely chimeric antigen receptors T cells (CAR-T), outlined unprecedented B cell malignancies results. In this context, the CAR-T-based approach has led to the proliferation of many clinical studies. In this review, we will deal with the CAR-T structure, and we will summarize the primary clinical studies assessing the risks and benefits of CAR-T cell therapy. We will also deal with the adverse events and management of cytokine release syndromes/immune effector cell-associated neurotoxicity syndrome (ICANS). Subsequently, we will review potential future improvements to overcome refractoriness and improve expansion while decreasing CAR-T's off-target effects. The advances in the CAR-T platform represent a step forward with promising unlimited future possibilities that made it a paradigm-shifting for the management of B cell malignancies.

13.
Exp Hematol ; 97: 57-65.e5, 2021 05.
Article in English | MEDLINE | ID: mdl-33617893

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a myelodysplastic syndrome/myeloproliferative neoplasm overlap syndrome characterized by monocytic proliferation in the presence of dysplastic bone marrow changes, inflammatory symptoms, and propensity for transformation to acute myeloid leukemia (AML), with a poor prognosis and limited treatment options. Unlike the α and ß isoforms, the phosphatidylinositol-3-kinase (PI3K)-δ signaling protein is predominantly expressed by hematopoietic cells and therefore has garnered interest as a potential target for the treatment of lymphomas and leukemias. We revealed a pattern of increased PIK3CD:PIK3CA ratio in monocytic M5 AML patients and cell lines, and this ratio correlated with responsiveness to pharmacological PI3K-δ inhibition in vitro. Because CMML is a disease defined by monocytic clonal proliferation, we tested the PI3K-δ inhibitor umbralisib as a single agent and in combination with the JAK1/2 inhibitor ruxolitinib, in CMML. Our ex vivo experiments with primary CMML patient samples revealed synergistic inhibition of viability and clonogenicity with this combination. Phospho-specific flow cytometry revealed that dual inhibition had the unique ability to decrease STAT5, ERK, AKT, and S6 phosphorylation simultaneously, which offers a mechanistic hypothesis for the enhanced efficacy of the combination treatment. These preclinical data indicate promising activity by co-inhibition of PI3K-δ and JAK1/2 and support the use of ruxolitinib + umbralisib combination therapy in CMML under active clinical investigation.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Leukemia, Myelomonocytic, Chronic/drug therapy , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Synergism , Humans , Leukemia, Myelomonocytic, Chronic/enzymology , Molecular Targeted Therapy , Nitriles , Pyrimidines
15.
Article in English | MEDLINE | ID: mdl-31762532

ABSTRACT

Splenomegaly segmentation on computed tomography (CT) abdomen anatomical scans is essential for identifying spleen biomarkers and has applications for quantitative assessment in patients with liver and spleen disease. Deep convolutional neural network automated segmentation has shown promising performance for splenomegaly segmentation. However, manual labeling of abdominal structures is resource intensive, so the labeled abdominal imaging data are rare resources despite their essential role in algorithm training. Hence, the number of annotated labels (e.g., spleen only) are typically limited with a single study. However, with the development of data sharing techniques, more and more publicly available labeled cohorts are available from different resources. A key new challenging is to co-learn from the multi-source data, even with different numbers of labeled abdominal organs in each study. Thus, it is appealing to design a co-learning strategy to train a deep network from heterogeneously labeled scans. In this paper, we propose a new deep convolutional neural network (DCNN) based method that integrates heterogeneous multi-resource labeled cohorts for splenomegaly segmentation. To enable the proposed approach, a novel loss function is introduced based on the Dice similarity coefficient to adaptively learn multi-organ information from different resources. Three cohorts were employed in our experiments, the first cohort (98 CT scans) has only splenomegaly labels, while the second training cohort (100 CT scans) has 15 distinct anatomical labels with normal spleens. A separate, independent cohort consisting of 19 splenomegaly CT scans with labeled spleen was used as testing cohort. The proposed method achieved the highest median Dice similarity coefficient value (0.94), which is superior (p-value<0.01 against each other method) to the baselines of multi-atlas segmentation (0.86), SS-Net segmentation with only spleen labels (0.90) and U-Net segmentation with multi-organ training (0.91). Our approach for adapting the loss function and training structure is not specific to the abdominal context and may be beneficial in other situations where datasets with varied label sets are available.

16.
Comput Biol Med ; 107: 109-117, 2019 04.
Article in English | MEDLINE | ID: mdl-30798219

ABSTRACT

Delineation of Computed Tomography (CT) abdominal anatomical structure, specifically spleen segmentation, is useful for not only measuring tissue volume and biomarkers but also for monitoring interventions. Recently, segmentation algorithms using deep learning have been widely used to reduce time humans spend to label CT data. However, the computerized segmentation has two major difficulties: managing intermediate results (e.g., resampled scans, 2D sliced image for deep learning), and setting up the system environments and packages for autonomous execution. To overcome these issues, we propose an automated pipeline for the abdominal spleen segmentation. This pipeline provides an end-to-end synthesized process that allows users to avoid installing any packages and to deal with the intermediate results locally. The pipeline has three major stages: pre-processing of input data, segmentation of spleen using deep learning, 3D reconstruction with the generated labels by matching the segmentation results with the original image dimensions, which can then be used later and for display or demonstration. Given the same volume scan, the approach described here takes about 50 s on average whereas the manual segmentation takes about 30 min on the average. Even if it includes all subsidiary processes such as preprocessing and necessary setups, the whole pipeline process requires on the average 20 min from beginning to end.


Subject(s)
Deep Learning , Imaging, Three-Dimensional/methods , Spleen/diagnostic imaging , Tomography, X-Ray Computed/methods , Algorithms , Humans , Time Factors
17.
IEEE Trans Med Imaging ; 38(5): 1185-1196, 2019 05.
Article in English | MEDLINE | ID: mdl-30442602

ABSTRACT

The findings of splenomegaly, abnormal enlargement of the spleen, is a non-invasive clinical biomarker for liver and spleen diseases. Automated segmentation methods are essential to efficiently quantify splenomegaly from clinically acquired abdominal magnetic resonance imaging (MRI) scans. However, the task is challenging due to: 1) large anatomical and spatial variations of splenomegaly; 2) large inter- and intra-scan intensity variations on multi-modal MRI; and 3) limited numbers of labeled splenomegaly scans. In this paper, we propose the Splenomegaly Segmentation Network (SS-Net) to introduce the deep convolutional neural network (DCNN) approaches in multi-modal MRI splenomegaly segmentation. Large convolutional kernel layers were used to address the spatial and anatomical variations, while the conditional generative adversarial networks were employed to leverage the segmentation performance of SS-Net in an end-to-end manner. A clinically acquired cohort containing both T1-weighted (T1w) and T2-weighted (T2w) MRI splenomegaly scans was used to train and evaluate the performance of multi-atlas segmentation (MAS), 2D DCNN networks, and a 3-D DCNN network. From the experimental results, the DCNN methods achieved superior performance to the state-of-the-art MAS method. The proposed SS-Net method has achieved the highest median and mean Dice scores among the investigated baseline DCNN methods.


Subject(s)
Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Neural Networks, Computer , Splenomegaly/diagnostic imaging , Humans , Imaging, Three-Dimensional/methods , Spleen/diagnostic imaging
18.
Article in English | MEDLINE | ID: mdl-30334788

ABSTRACT

A key limitation of deep convolutional neural networks (DCNN) based image segmentation methods is the lack of generalizability. Manually traced training images are typically required when segmenting organs in a new imaging modality or from distinct disease cohort. The manual efforts can be alleviated if the manually traced images in one imaging modality (e.g., MRI) are able to train a segmentation network for another imaging modality (e.g., CT). In this paper, we propose an end-to-end synthetic segmentation network (SynSeg-Net) to train a segmentation network for a target imaging modality without having manual labels. SynSeg-Net is trained by using (1) unpaired intensity images from source and target modalities, and (2) manual labels only from source modality. SynSeg-Net is enabled by the recent advances of cycle generative adversarial networks (CycleGAN) and DCNN. We evaluate the performance of the SynSeg-Net on two experiments: (1) MRI to CT splenomegaly synthetic segmentation for abdominal images, and (2) CT to MRI total intracranial volume synthetic segmentation (TICV) for brain images. The proposed end-to-end approach achieved superior performance to two stage methods. Moreover, the SynSeg-Net achieved comparable performance to the traditional segmentation network using target modality labels in certain scenarios. The source code of SynSeg-Net is publicly available 2.

19.
Leuk Res ; 65: 67-73, 2018 02.
Article in English | MEDLINE | ID: mdl-29310020

ABSTRACT

The National Comprehensive Cancer Network (NCCN) defines the following types of acute myeloid leukemia (AML) as favorable-risk: acute promyelocytic leukemia with t(15;17) (APL); AML with core-binding factor (CBF) rearrangements, including t(8;21) and inv(16) or t(16;16) without mutations in KIT (CBF-KITwt); and AML with normal cytogenetics and mutations in NPM1 (NPM1mut); or biallelic mutations in CEBPA (CEBPAmut/mut), without FLT3-ITD. Although these AMLs are categorized as favorable risk by NCCN, clinical experience suggests that there are differences in clinical outcome amongst these cytogenetically and molecularly distinct leukemias. This study compared clinical and genotypic characteristics of 60 patients with favorable-risk AML, excluding APL, and demonstrated significant differences between them. Patients with NPM1mut AML were significantly older than those in the other groups. Targeted next-generation sequencing on DNA from peripheral blood or bone marrow revealed significantly more mutations in NPM1mut AML than the other favorable-risk diseases, especially in genes related to DNA splicing and methylation. CEBPAmut/mut AMLs exhibited more mutations in transcription-related genes. Patients with NPM1mut AML and CEBPAmut/mut AML show significantly reduced overall survival in comparison with CBF-KITwt AML. These findings emphasize that favorable-risk AML patients have divergent outcomes and that differences in clinical and genotypic characteristics should be considered in their evaluation and management.


Subject(s)
Genotype , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Adult , Age Factors , Aged , CCAAT-Enhancer-Binding Proteins/genetics , Core Binding Factors/genetics , Female , Gene Order , Humans , Kaplan-Meier Estimate , Karyotyping , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/drug therapy , Male , Middle Aged , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Proto-Oncogene Proteins c-kit/genetics , Retrospective Studies , Risk Assessment , Treatment Outcome
20.
Curr Hematol Malig Rep ; 12(5): 468-477, 2017 10.
Article in English | MEDLINE | ID: mdl-28983777

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a myeloid malignancy which shares clinical and morphologic features of myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPNs) and is classified by the WHO as an MDS/MPN. The defining feature of CMML is clonal hematopoiesis that results in peripheral monocytosis. The benefit of early treatment is currently unclear, and treatment may be held until the disease exhibits accelerated blast counts or the patient becomes symptomatic. Optimal treatments for CMML are not well defined. Conventional treatments include hydroxyurea, cytarabine, and hypomethylating agents. However, all treatment options are limited and, with the exception of allogeneic stem cell transplantation, are considered palliative. As we continue to learn about the genomics of CMML and about arising therapeutic targets and those under active clinical investigation, the future therapy of CMML will likely improve considerably. Here, we review the data available for conventional therapies and highlight emerging therapeutic strategies.


Subject(s)
Cytarabine/therapeutic use , Hydroxyurea/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Stem Cell Transplantation , Allografts , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , World Health Organization
SELECTION OF CITATIONS
SEARCH DETAIL
...