Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hematol Oncol ; 40(4): 491-504, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35368098

ABSTRACT

Despite much of the past 2 years being engulfed by the devastating consequences of the SAR-CoV-2 pandemic, significant progress, even breathtaking, occurred in the field of chronic myeloid malignancies. Some of this was show-cased at the 15th Post-American Society of Hematology (ASH) and the 25th John Goldman workshops on myeloproliferative neoplasms (MPN) held on 9th-10th December 2020 and 7th-10th October 2021, respectively. The inaugural Post-ASH MPN workshop was set out in 2006 by John Goldman (deceased) and Tariq Mughal to answer emerging translational hematology and therapeutics of patients with these malignancies. Rather than present a resume of the discussions, this perspective focuses on some of the pivotal translational hematology and therapeutic insights in these diseases.


Subject(s)
COVID-19 , Graft vs Host Disease , Hematology , Myeloproliferative Disorders , Hematopoietic Stem Cells , Humans , Myeloproliferative Disorders/drug therapy
2.
Hematol Oncol ; 38(5): 654-664, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32592408

ABSTRACT

This review reflects the presentations and discussion at the 14th post-American Society of Hematology (ASH) International Workshop on Chronic Myeloproliferative Malignancies, which took place on the December 10 and 11, 2019, immediately after the 61st ASH Annual Meeting in Orlando, Florida. Rather than present a resume of the proceedings, we address some of the topical translational science research and clinically relevant topics in detail. We consider how recent studies using single-cell genomics and other molecular methods reveal novel aspects of hematopoiesis which in turn raise the possibility of new therapeutic approaches for patients with myeloproliferative neoplasms (MPNs). We discuss how alternative therapies could benefit patients with chronic myeloid leukemia who develop BCR-ABL1 mutant subclones following ABL1-tyrosine kinase inhibitor therapy. In MPNs, we focus on efforts beyond JAK-STAT and the merits of integrating activin receptor ligand traps, interferon-α, and allografting in the current treatment algorithm for patients with myelofibrosis.


Subject(s)
Disease Susceptibility , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/therapy , Anemia/diagnosis , Anemia/etiology , Anemia/therapy , Biomarkers , Biomarkers, Tumor , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Disease Management , Drug Development , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Molecular Diagnostic Techniques , Molecular Targeted Therapy , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/diagnosis , Prognosis , Single-Cell Analysis/methods , Translational Research, Biomedical , Treatment Outcome
3.
Hematol Oncol ; 37(3): 240-252, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31013548

ABSTRACT

The 60th American Society of Hematology (ASH) held in San Diego in December 2018 was followed by the 13th Post-ASH chronic myeloproliferative neoplasms (MPNs) workshop on December 4 and 5, 2018. This closed annual workshop, first introduced in 2006 by Goldman and Mughal, was organized in collaboration with Alpine Oncology Foundation and allowed experts in preclinical and clinical research in the chronic MPNs to discuss the current scenario, including relevant presentations at ASH, and address pivotal open questions that impact translational research and clinical management. This review is based on the presentations and deliberations at this workshop, and rather than provide a resume of the proceedings, we have selected some of the important translational science and treatment issues that require clarity. We discuss the experimental and observational evidence to support the intimate interaction between aging, inflammation, and clonal evolution of MPNs, the clinical impact of the unfolding mutational landscape on the emerging targets and treatment of MPNs, new methods to detect clonal heterogeneity, the challenges in managing childhood and adolescent MPN, and reflect on the treatment of systemic mastocytosis (SM) following the licensing of midostaurin.


Subject(s)
Myeloproliferative Disorders/therapy , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends , Aging , Animals , Congresses as Topic , DNA Mutational Analysis , Humans , Inflammation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Mastocytosis/therapy , Medical Oncology/methods , Medical Oncology/trends , Mice , Mutation , Prognosis , Societies, Medical , Staurosporine/analogs & derivatives , Staurosporine/therapeutic use , United States
4.
Blood ; 127(24): 3004-14, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26966091

ABSTRACT

The spectrum of somatic alterations in hematologic malignancies includes substitutions, insertions/deletions (indels), copy number alterations (CNAs), and a wide range of gene fusions; no current clinically available single assay captures the different types of alterations. We developed a novel next-generation sequencing-based assay to identify all classes of genomic alterations using archived formalin-fixed paraffin-embedded blood and bone marrow samples with high accuracy in a clinically relevant time frame, which is performed in our Clinical Laboratory Improvement Amendments-certified College of American Pathologists-accredited laboratory. Targeted capture of DNA/RNA and next-generation sequencing reliably identifies substitutions, indels, CNAs, and gene fusions, with similar accuracy to lower-throughput assays that focus on specific genes and types of genomic alterations. Profiling of 3696 samples identified recurrent somatic alterations that impact diagnosis, prognosis, and therapy selection. This comprehensive genomic profiling approach has proved effective in detecting all types of genomic alterations, including fusion transcripts, which increases the ability to identify clinically relevant genomic alterations with therapeutic relevance.


Subject(s)
DNA Fingerprinting/methods , Gene Expression Profiling/methods , Genomics/methods , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Chromosome Aberrations , Clinical Laboratory Techniques/methods , DNA Mutational Analysis/methods , DNA, Neoplasm/analysis , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Polymorphism, Genetic , RNA, Neoplasm/analysis , Sensitivity and Specificity , Systems Integration
5.
Hematol Oncol ; 36(5): 740-748, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30074634

ABSTRACT

Following the 47th American Society of Hematology Meeting in 2005, the late John Goldman and Tariq Mughal commenced a conference, the 1st Post-ASH Workshop, which brought together clinicians and scientists, to accelerate the adoption of new therapies for patients with myeloproliferative neoplasms (MPNs). The concept began with recognition of the CML success story following imatinib therapy, the discovery of JAK2V617F , and the demonstration that BCR-ABL1-negative MPNs are driven by abnormal JAK2 activation. This review is based on the presentations and deliberations at the XIIth Post-ASH Workshop on BCR-ABL1 positive and negative MPNs that took place on December 12 to 13, 2017, in Atlanta, Georgia, immediately following the 59th American Society of Hematology Meeting. We have selected some of the translational research and clinical topics, rather than an account of the proceedings. We discuss the role of immunotherapy in MPNs and the impact of the mutational landscape on TKI treatment in CML. We also consider how we might reduce TKI cardiovascular side effects, the potential role of nutrition as adjunctive nonpharmacologic intervention to reduce chronic inflammation in MPNs, and novel investigational therapies for MPNs.


Subject(s)
Hematologic Neoplasms , Immunotherapy/methods , Myeloproliferative Disorders , Precision Medicine/methods , Amino Acid Substitution , Fusion Proteins, bcr-abl , Hematologic Neoplasms/genetics , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Humans , Janus Kinase 2 , Mutation, Missense , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/therapy , Translational Research, Biomedical
6.
Blood ; 125(12): 1857-65, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25624319

ABSTRACT

Myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN) are hematologically diverse stem cell malignancies sharing phenotypic features of both myelodysplastic syndromes and myeloproliferative neoplasms. There are currently no standard treatment recommendations for most adult patients with MDS/MPN. To optimize efforts to improve the management and disease outcomes, it is essential to identify meaningful clinical and biologic end points and standardized response criteria for clinical trials. The dual dysplastic and proliferative features in these stem cell malignancies define their uniqueness and challenges. We propose response assessment guidelines to harmonize future clinical trials with the principal objective of establishing suitable treatment algorithms. An international panel comprising laboratory and clinical experts in MDS/MPN was established involving 3 independent academic MDS/MPN workshops (March 2013, December 2013, and June 2014). These recommendations are the result of this collaborative project sponsored by the MDS Foundation.


Subject(s)
Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/therapy , Hematology/standards , Medical Oncology/standards , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/therapy , Algorithms , Cell Proliferation , Clinical Trials as Topic , Disease Progression , Humans , International Cooperation , Mutation , Phenotype , Practice Guidelines as Topic , Surveys and Questionnaires , Treatment Outcome
7.
Am J Hematol ; 92(10): 1062-1067, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28685840

ABSTRACT

The purpose of the study was to assess consensus and interobserver agreement among an international panel of six hematopathologists regarding characterization and reproducibility of bone marrow (BM) histologic features used to diagnose early stage myeloproliferative neoplasms, in particular differentiation of so-called masked/prodromal polycythemia vera (mPV) from JAK2-mutated essential thrombocythemia (ET). The six members of the hematopathology panel evaluated 98 BM specimens independently and in a blinded fashion without knowledge of clinical data. The specimens included 48 cases of mPV according to the originally published hemoglobin threshold values for this entity (male: 16.0-18.4 g/dL, female: 15.0-16.4 g/dL), 31 cases with overt PV according to the updated 2016 WHO criteria, and 19 control cases. The latter group included cases of JAK2-mutated ET, primary myelofibrosis, myelodysplastic syndrome, and various reactive conditions. Inter-rater agreement between the panelists was very high (overall agreement 92.6%, kappa 0.812), particularly with respect to separating mPV from ET. Virtually all cases of mPV were correctly classified as PV according to their BM morphology. In conclusion, a central blinded review of histology slides by six hematopathologists demonstrated that highly reproducible specific histological pattern characterize PV and confirmed the notion that there are no significant differences between mPV and overt PV in relation to BM morphology.


Subject(s)
Bone Marrow/pathology , Polycythemia Vera/pathology , Thrombocythemia, Essential/pathology , Diagnosis, Differential , Europe , Female , Hemoglobins/analysis , Humans , Janus Kinase 2/genetics , Male , Mutation , Polycythemia Vera/blood , Polycythemia Vera/genetics , Reproducibility of Results , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/genetics , World Health Organization
9.
Haematologica ; 101(5): 541-58, 2016 05.
Article in English | MEDLINE | ID: mdl-27132280

ABSTRACT

With the deaths of Janet Rowley and John Goldman in December 2013, the world lost two pioneers in the field of chronic myeloid leukemia. In 1973, Janet Rowley, unraveled the cytogenetic anatomy of the Philadelphia chromosome, which subsequently led to the identification of the BCR-ABL1 fusion gene and its principal pathogenetic role in the development of chronic myeloid leukemia. This work was also of major importance to support the idea that cytogenetic changes were drivers of leukemogenesis. John Goldman originally made seminal contributions to the use of autologous and allogeneic stem cell transplantation from the late 1970s onwards. Then, in collaboration with Brian Druker, he led efforts to develop ABL1 tyrosine kinase inhibitors for the treatment of patients with chronic myeloid leukemia in the late 1990s. He also led the global efforts to develop and harmonize methodology for molecular monitoring, and was an indefatigable organizer of international conferences. These conferences brought together clinicians and scientists, and accelerated the adoption of new therapies. The abundance of praise, tributes and testimonies expressed by many serve to illustrate the indelible impressions these two passionate and affable scholars made on so many people's lives. This tribute provides an outline of the remarkable story of chronic myeloid leukemia, and in writing it, it is clear that the historical triumph of biomedical science over this leukemia cannot be considered without appreciating the work of both Janet Rowley and John Goldman.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cytogenetic Analysis/history , Cytogenetic Analysis/methods , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , History, 20th Century , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Molecular Targeted Therapy/history , Molecular Targeted Therapy/methods , Mutation , Philadelphia Chromosome , Prognosis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Research/history
10.
Haematologica ; 100(9): 1117-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26341525

ABSTRACT

In the 2008 WHO classification, chronic myeloid malignancies that share both myelodysplastic and myeloproliferative features define the myelodysplastic/myeloproliferative group, which includes chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, atypical chronic myeloid leukemia, refractory anemia with ring sideroblasts and thrombocytosis, and myelodysplastic/myeloproliferative unclassified. With the notable exception of refractory anemia with ring sideroblasts and thrombocytosis, there is much overlap among the various subtypes at the molecular and clinical levels, and a better definition of these entities, an understanding of their biology and an identification of subtype-specific molecular or cellular markers are needed. To address some of these challenges, a panel comprised of laboratory and clinical experts in myelodysplastic/myeloproliferative was established, and four independent academic MDS/MPN workshops were held on: 9(th) March 2013, in Miami, Florida, USA; 6(th) December 2013, in New Orleans, Louisiana, USA; 13(th) June 2014 in Milan, Italy; and 5(th) December 2014 in San Francisco, USA. During these meetings, the current understanding of these malignancies and matters of biology, diagnosis and management were discussed. This perspective and the recommendations on molecular pathogenesis, diagnosis and clinical characterization for adult onset myelodysplastic/myeloproliferative is the result of a collaborative project endorsed and supported by the MDS Foundation.


Subject(s)
Hematologic Neoplasms , Myelodysplastic-Myeloproliferative Diseases , Adult , Female , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Male , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/pathology , Myelodysplastic-Myeloproliferative Diseases/therapy , Practice Guidelines as Topic
11.
EJHaem ; 5(1): 61-69, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38406504

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) predominantly occurs in adults ≥60 years old; 10-20% of cases are pediatric or adolescent/young adult (AYA) patients. Tagraxofusp (TAG, Elzonris®) is the only approved treatment for BPDCN; in the United States it is approved for patients aged ≥2 years. Data on treating pediatric and AYA BPDCN patients are limited. We present a case series of pediatric and AYA patients with BPDCN treated with TAG. Eight patients (five newly diagnosed; three relapsed/refractory [R/R]), aged 2-21 years, received 12 mcg/kg TAG. Seven patients were female; most had skin (n = 6) and/or bone marrow (n = 4) involvement. No new safety signals were identified. Grade 3 adverse events were headache (n = 1) and transaminitis (n = 2). Three patients with newly diagnosed BPDCN achieved complete response, one achieved partial response, and one had stable disease (SD). One patient with R/R BPDCN achieved a minor response; one had SD. Seven patients (88%) were bridged to stem cell transplant: 80% of newly diagnosed patients and 100% of R/R patients. Five patients remained alive at last follow-up. These cases highlight the efficacy and safety of TAG in pediatric and AYA patients for whom there is no other approved BPDCN therapy.

12.
Clin Lymphoma Myeloma Leuk ; 24(4): e130-e137, 2024 04.
Article in English | MEDLINE | ID: mdl-38267355

ABSTRACT

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive myeloid malignancy of the dendritic cell lineage that affects patients of all ages, though the incidence appears to be highest in patients over the age of 60 years. Diagnosis is based on the presence of plasmacytoid dendritic cell precursors expressing CD123, the interleukin-3 (IL-3) receptor alpha, and a distinct histologic appearance. Timely diagnosis remains a challenge, due to lack of disease awareness and overlapping biologic and clinical features with other hematologic malignancies. Prognosis is poor with a median overall survival of 8 to 14 months, irrespective of disease presentation pattern. Historically, the principal treatment was remission induction therapy followed by a stem cell transplant (SCT) in eligible patients. However, bridging to SCT is often not achieved with induction chemotherapy regimens. The discovery that CD123 is universally expressed in BPDCN and is considered to have a pathogenetic role in its development paved the way for the successful introduction of tagraxofusp, a recombinant human IL-3 fused to a truncated diphtheria toxin payload, as an initial treatment for BPDCN. Tagraxofusp was approved in 2018 by the United States Food and Drug Administration for the treatment of patients aged 2 years and older with newly diagnosed and relapsed/refractory BPDCN, and by the European Medicines Agency in 2021 for first-line treatment of adults. The advent of tagraxofusp has opened a new era of precision oncology in the treatment of BPDCN. Herein, we present an overview of BPDCN biology, its diagnosis, and treatment options, illustrated by clinical cases.


Subject(s)
Hematologic Neoplasms , Myeloproliferative Disorders , Skin Neoplasms , Adult , Humans , Middle Aged , Interleukin-3 Receptor alpha Subunit , Interleukin-3/therapeutic use , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Precision Medicine , Acute Disease , Myeloproliferative Disorders/pathology , Skin Neoplasms/pathology , Dendritic Cells/pathology , Biology
13.
Blood Adv ; 8(3): 591-602, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38052038

ABSTRACT

ABSTRACT: CD123, a subunit of the interleukin-3 receptor, is expressed on ∼80% of acute myeloid leukemias (AMLs). Tagraxofusp (TAG), recombinant interleukin-3 fused to a truncated diphtheria toxin payload, is a first-in-class drug targeting CD123 approved for treatment of blastic plasmacytoid dendritic cell neoplasm. We previously found that AMLs with acquired resistance to TAG were re-sensitized by the DNA hypomethylating agent azacitidine (AZA) and that TAG-exposed cells became more dependent on the antiapoptotic molecule BCL-2. Here, we report a phase 1b study in 56 adults with CD123-positive AML or high-risk myelodysplastic syndrome (MDS), first combining TAG with AZA in AML/MDS, and subsequently TAG, AZA, and the BCL-2 inhibitor venetoclax (VEN) in AML. Adverse events with 3-day TAG dosing were as expected, without indication of increased toxicity of TAG or AZA+/-VEN in combination. The recommended phase 2 dose of TAG was 12 µg/kg/day for 3 days, with 7-day AZA +/- 21-day VEN. In an expansion cohort of 26 patients (median age 71) with previously untreated European LeukemiaNet adverse-risk AML (50% TP53 mutated), triplet TAG-AZA-VEN induced response in 69% (n=18/26; 39% complete remission [CR], 19% complete remission with incomplete count recovery [CRi], 12% morphologic leukemia-free state [MLFS]). Among 13 patients with TP53 mutations, 7/13 (54%) achieved CR/CRi/MLFS (CR = 4, CRi = 2, MLFS = 1). Twelve of 17 (71%) tested responders had no flow measurable residual disease. Median overall survival and progression-free survival were 14 months (95% CI, 9.5-NA) and 8.5 months (95% CI, 5.1-NA), respectively. In summary, TAG-AZA-VEN shows encouraging safety and activity in high-risk AML, including TP53-mutated disease, supporting further clinical development of TAG combinations. The study was registered on ClinicalTrials.gov as #NCT03113643.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Recombinant Fusion Proteins , Sulfonamides , Adult , Aged , Humans , Azacitidine/therapeutic use , Interleukin-3 Receptor alpha Subunit , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Proto-Oncogene Proteins c-bcl-2
14.
Am J Hematol ; 87(5): 562-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22460584

ABSTRACT

Immediately after the annual scientific meeting of the American Society of Hematology (ASH), a select group of clinical and laboratory investigators in myeloproliferative neoplasms (MPN) is summoned to a post-ASH conference on chronic myeloid leukemia and the BCR-ABL1-negative MPN. The 6th such meeting occurred on December 13­14,2011, in La Jolla, California, USA, under the direction of its founder,Dr. Tariq Mughal. The current document is the first of two reports on this post-ASH event and summarizes the most recent preclinical and clinical advances in polycythemia vera, essential thrombocythemia,and primary myelofibrosis.


Subject(s)
Genes, Neoplasm , Myeloproliferative Disorders/genetics , Clinical Trials as Topic , DNA Methylation , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Disease Progression , Drugs, Investigational/therapeutic use , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Inflammation , Interferon alpha-2 , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Multicenter Studies as Topic , Myeloproliferative Disorders/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Polyethylene Glycols/therapeutic use , Prognosis , Recombinant Proteins/therapeutic use , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Treatment Outcome
15.
J Clin Oncol ; 40(26): 3032-3036, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35820082

ABSTRACT

Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive myeloid malignancy. We report long-term results, including data from the continued access phase, of the largest prospective BPDCN trial evaluating the CD123-targeted therapy tagraxofusp (TAG) in adults with treatment-naive and relapsed/refractory BPDCN. The primary outcome was complete response (CR) + clinical CR (CRc: CR with residual skin abnormality not indicative of active disease). Eighty-four (65 treatment-naive and 19 relapsed/refractory) of 89 patients received TAG 12 µg/kg once daily; the median follow-up was 34.0 months. For treatment-naive patients, the overall response rate was 75%; 57% achieved CR + CRc. The median time to remission was 39 (range, 14-131) days, and the median CR + CRc duration was 24.9 (95% CI, 3.8 to not reached) months. Nineteen patients (51%) with CR + CRc were bridged to stem-cell transplant, with a median CR + CRc duration of 22.2 (range, 1.5-57.4) months. Most common adverse events were increased alanine (64%) or aspartate (60%) aminotransferase and hypoalbuminemia (51%); most occurred in cycle 1 and were transient. Capillary leak syndrome occurred in 21% of patients (grade ≥ 3: 7%). In first-line patients with BPDCN, TAG monotherapy resulted in high and durable responses, allowing many to bridge to stem-cell transplant. TAG was generally well-tolerated with a predictable and manageable safety profile.


Subject(s)
Hematologic Neoplasms , Myeloproliferative Disorders , Skin Neoplasms , Acute Disease , Adult , Clinical Trials as Topic , Dendritic Cells/metabolism , Dendritic Cells/pathology , Hematologic Neoplasms/therapy , Humans , Interleukin-3 Receptor alpha Subunit/metabolism , Interleukin-3 Receptor alpha Subunit/therapeutic use , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Prospective Studies , Recombinant Fusion Proteins , Skin Neoplasms/pathology
16.
Haematologica ; 96(4): 590-601, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21242185

ABSTRACT

This review focuses on topical issues in the biology and treatment of the myeloproliferative neoplasms (MPNs). Studies in transgenic mice suggest that BCR-ABL1 reduces the fraction of self-renewing 'leukemic' stem cells in the bone marrow but that some of these cells survive treatment with imatinib. This also seems to operate in humans. Data from models also strongly support the notion that JAK2(V617F) can initiate and sustain MPNs in mice; relevance to disease in humans is less clear. These data also support the hypothesis that level of JAK2(V617F) expression influences the MPN phenotype: higher levels favor erythrocytosis whereas lower levels favor thrombocytosis. Although TET2-mutations were thought to precede JAK2(V617F) in some persons with MPNs, it now appears that TET2 mutations may occur after JAK2(V617F). Further understanding of signal-transduction pathways activated in chronic myeloid leukemia suggests various possible targets for new therapies including the WNT/beta catenin, notch and hedgehog pathways. Finally, the clinical role of the new JAK2- and BCR-ABL1-inhibitors is considered. Much further progress is likely in several of these areas soon.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Animals , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Humans , Immunotherapy , Janus Kinase 2/genetics , Mice , Mice, Transgenic , Molecular Targeted Therapy , Myeloproliferative Disorders/therapy , Neoplastic Stem Cells/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors
17.
Leuk Lymphoma ; 62(11): 2568-2586, 2021 11.
Article in English | MEDLINE | ID: mdl-33999767

ABSTRACT

Following the observation of interleukin 3 receptor α chain (IL-3Rα; CD123) upregulation on leukemia stem cells (LSCs) almost two decades ago, targeted treatment via CD123-diptheria toxin conjugates has now been tested in patients with diverse myeloid malignancies. Targeted eradication of LSCs could result in effective treatments for many challenging diseases initiated by these cells. Consequently, considerable effort has been directed toward targeting CD123 as a potential strategy for treating patients with hematologic malignancies in which CD123 is overexpressed. However, these therapies have had limited success so far, highlighting the need for suitable criteria to identify patients who could benefit from them. Given the diversity in CD123 expression across different hematologic malignancies, understanding CD123 expression patterns and the functional pathogenetic significance is crucial. Here, we review the methodologies available for CD123 assessment and discuss the biological and clinical characteristics of patients for whom CD123-targeting therapies may have a clinical impact.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Humans , Interleukin-3 Receptor alpha Subunit
18.
Med ; 2(12): 1292-1313, 2021 12 10.
Article in English | MEDLINE | ID: mdl-35590147

ABSTRACT

Detection of minimal residual disease in patients with cancer, who are in complete remission with no cancer cells detectable, has the potential to improve recurrence-free survival through treatment selection. Studies analyzing circulating tumor DNA (ctDNA) in patients with solid tumors suggest the potential to accurately predict and detect relapse, enabling treatment strategies that may improve clinical outcomes. Over the past decade, assays for ctDNA detection in plasma samples have steadily increased in sensitivity and specificity. These are applied for the detection of residual disease after treatment and for earlier detection of recurrence. Novel clinical trials are now assessing how assays for "residual disease and recurrence" (RDR) may influence current treatment paradigms and potentially change the landscape of risk classification for cancer recurrence. In this review, we appraise the progress of RDR detection using ctDNA and consider the emerging role of liquid biopsy in the monitoring and management of solid tumors.


Subject(s)
Circulating Tumor DNA , Neoplasm Recurrence, Local , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Humans , Liquid Biopsy , Neoplasm Recurrence, Local/diagnosis , Neoplasm, Residual
19.
Hemasphere ; 5(3): e536, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33623882

ABSTRACT

During the past few years, our understanding of molecular mechanisms and cellular interactions relevant to malignant blood cell disorders has improved substantially. New insights include a detailed knowledge about disease-initiating exogenous factors, endogenous (genetic, somatic, epigenetic) elicitors or facilitators of disease evolution, and drug actions and interactions that underlie efficacy and adverse event profiles in defined cohorts of patients. As a result, precision medicine and personalized medicine are rapidly growing new disciplines that support the clinician in making the correct diagnosis, in predicting outcomes, and in optimally selecting patients for interventional therapies. In addition, precision medicine tools are greatly facilitating the development of new drugs, therapeutic approaches, and new multiparametric prognostic scoring models. However, although the emerging roles of precision medicine and personalized medicine in hematology and oncology are clearly visible, several questions remain. For example, it remains unknown how precision medicine tools can be implemented in healthcare systems and whether all possible approaches are also affordable. In addition, there is a need to define terminologies and to relate these to specific and context-related tools and strategies in basic and applied science. To discuss these issues, a working conference was organized in September 2019. The outcomes of this conference are summarized herein and include a proposal for definitions, terminologies, and applications of precision and personalized medicine concepts and tools in hematologic neoplasms. We also provide proposals aimed at reducing costs, thereby making these applications affordable in daily practice.

SELECTION OF CITATIONS
SEARCH DETAIL