Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.459
Filter
1.
Ann Hematol ; 101(1): 177-189, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34591162

ABSTRACT

An intensified myeloablative conditioning regimen, involving the addition of granulocyte colony-stimulating factor (G-CSF)-combined high-dose cytarabine (12 g/m2) to standard total body irradiation and cyclophosphamide, has been performed for adult patients with myeloid malignancies in single-unit cord blood transplantation (CBT) since 1998 in our institute. We update the results of CBT, as the first allogeneic hematopoietic cell transplantation after this conditioning regimen, in 169 patients with a median long-term follow-up of 10.4 years. The median age was 43 years (range, 16 to 59 years). Ninety-four patients (56%) were in non-remission at the time of CBT, and 124 patients (73%) were acute myeloid leukemia. The median cryopreserved cord blood total nucleated cell dose and CD34+ cell dose was 2.40 × 107/kg and 0.93 × 105/kg, respectively. The cumulative incidence of neutrophil recovery at 42 days was 94.4% (95% confidence interval [CI]: 88.6-97.3%). Among the whole cohort, 105 patients were still alive at the end of the study period. The cumulative incidences of relapse and non-relapse mortality at 10 years were 26.0% (95% CI: 19.5-33.0%) and 16.9% (95% CI: 11.4-23.4%), respectively. There was an overall survival probability of 62.5% (95% CI: 54.3-69.7%) at 10 years. Higher disease risk index alone significantly affected higher overall mortality (hazard ratio 2.21, P = 0.003) in multivariate analysis. These outcomes demonstrate that G-CSF-combined myeloablative conditioning could have favorable long-term remission rates for adult patients with myeloid malignancies undergoing single-unit CBT.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Cyclophosphamide/therapeutic use , Cytarabine/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Immunosuppressive Agents/therapeutic use , Leukemia, Myeloid/therapy , Adolescent , Adult , Female , Humans , Leukemia, Myeloid/epidemiology , Male , Middle Aged , Retrospective Studies , Survival Analysis , Transplantation Conditioning/methods , Treatment Outcome , Whole-Body Irradiation , Young Adult
2.
J Immunol ; 208(2): 492-500, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34937746

ABSTRACT

The interaction of inhibitory receptors with self-MHC class I (MHC-I) molecules is responsible for NK cell education. The intensity of DNAM-1 expression correlates with NK cell education. However, whether DNAM-1 expression directly influences the functional competence of NK cells via the KIR/MHC-I interaction remains unclear. Based on allogeneic haploidentical hematopoietic stem cell transplantation, we investigated the intensity of DNAM-1 expression on reconstituted NK cells via the interaction of KIR with both donor HLA and recipient HLA at days 30, 90, and 180 after hematopoietic stem cell transplantation. The reconstituted NK cells educated by donor and recipient HLA molecules showed the highest DNAM-1 expression, whereas DNAM-1 expression on educated NK cells with only recipient HLA molecules was higher than that on educated NK cells with only donor HLA molecules, indicating that NK cells with donor or recipient HLA molecules regulate DNAM-1 expression and thereby affect NK cell education. Additionally, the effects of recipient cells on NK cell education were greater than those of donor cells. However, only when the DNAM-1, NKP30, and NKG2D receptors were blocked simultaneously was the function of educated and uneducated NK cells similar. Therefore, activating receptors may collaborate with DNAM-1 to induce educated NK cell hyperresponsiveness. Our data, based on in vitro and in vivo studies, demonstrate that the functional competence of NK cells via the KIR/MHC-I interaction correlates with DNAM-1 expression in human NK cells.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Receptors, KIR/immunology , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Case-Control Studies , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Lymphoid/therapy , Leukemia, Myeloid/therapy , Myelodysplastic Syndromes/therapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Cytotoxicity Triggering Receptor 3/metabolism , Prospective Studies
3.
Leuk Res ; 112: 106757, 2022 01.
Article in English | MEDLINE | ID: mdl-34864369

ABSTRACT

We studied 79 patients with AML-MRC or RAEB-T, who were later reclassified according to the WHO classification. Marrow slides were examined cytomorphologically with regard to dysplasia. Patients were followed up until March 2020. Thirty-one patients underwent allogeneic stem cell transplantation (median survival (ms) 16 months), 14 were treated with induction chemotherapy (ms 8.4 months), 18 received hypomethylating agents (ms 9.2 months), 16 received low dose chemotherapy or best supportive care (ms 2.4 months). Only 30.4 % fulfilled the morphologic WHO criteria. 46.8 % were classified as AML-MRC by an antecedent MDS, 54.4 % of the pts were classified by MDS-related chromosomal abnormalities. 5 % did not fulfill any of the criteria and were entered based on 20-29 % medullary blasts. There was no difference in ms between pts presenting with > 50 % dysplasia as compared to pts with dysplasia between 10 % and 50 % (ms 9.1 vs 9.9 months, p = n.s.) or for pts with antecedent MDS (ms 9.1 vs 8.9 months, p = n.s.). Myelodysplasia-related cytogenetic abnormalities were associated with a worse outcome (ms 8.1 vs 13.5 months, p = 0.026). AML-MRC in its current definition is a heterogenous entity. Dysplasia of ≥ 50 % in ≥ two lineages is not helpful for diagnostics and prognostication and therefore should be deleted in future classifications. We recommend utilizing the WHO guidelines for defining dysplasia (10 % or greater in ≥ 1 of the three myeloid cell lines) assisting in establishing the diagnosis of MDS.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid/genetics , Myelodysplastic Syndromes/genetics , Acute Disease , Aged , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/therapy , Chromosome Aberrations/statistics & numerical data , Female , Humans , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/therapy , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Outcome Assessment, Health Care/methods , Retrospective Studies , Survival Analysis , World Health Organization
4.
Leuk Res ; 112: 106732, 2022 01.
Article in English | MEDLINE | ID: mdl-34864447

ABSTRACT

Acute Myeloid Leukemia (AML) represents 1 % of all new cancer diagnosis made annually in the US and has a five-year survival of 30 %. Traditional treatment includes aggressive induction therapy followed by consolidation therapy that may include a hematopoietic stem cell transplant (HSCT). Thus far, HSCT remains the only potentially curative therapy for many patients with AML owing to the graft-versus-leukemia effect elicited by this treatment. The use of novel therapies, specifically immunotherapy, in the treatment of AML has been limited by the lack of appropriate target antigens, therapy associated toxicities and variable success with treatment. Antigenic variability on leukemia cells and the sharing of antigens by malignant and non-malignant cells makes the identification of appropriate antigens problematic. While studies with immunotherapeutic agents are underway, prior investigations have demonstrated a mixed response with some studies prematurely discontinued due to associated toxicities. This review presents a discussion of the envisioned role of immunotherapy in the treatment of AML in the setting of mixed therapeutic success and potentially lethal toxicities.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , Leukemia, Myeloid/therapy , Acute Disease , Animals , Graft vs Host Disease/etiology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/immunology , Immunotherapy/adverse effects , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Prognosis , Recurrence , Transplantation, Homologous , Treatment Outcome
6.
Transplant Cell Ther ; 28(1): 20.e1-20.e10, 2022 01.
Article in English | MEDLINE | ID: mdl-34607074

ABSTRACT

NKG2D-mediated cytotoxicity is regulated by the single nucleotide polymorphism rs1049174, and its antitumor effect has been observed in various clinical settings. There are no previously published data on the influence of donor rs1049174 polymorphism on HLA-haploidentical allogeneic hematopoietic cell transplantation using post-transplantation cyclophosphamide (PTCy-haplo). We aimed to investigate the effect of donor NKG2D gene polymorphism on PTCy-haplo recipients. We retrospectively reviewed 91 consecutive PTCy-haplo recipients at our institution, and genotyped rs1049174 of the NKG2D gene in both donors and patients. In the patients who received PTCy without antithymocyte globulin (ATG) as graft-versus-host disease prophylaxis, the 2-year cumulative incidence of relapse/progression (RI) of PTCy-haplo from rs1049174 CC donors was lower than that from rs1049174 CG/GG donors (25.0% versus 52.4%; P = .041), and rs1049174 CC donors were associated with a decreased risk of relapse/progression (adjusted hazard ratio, 0.2; 95% confidence interval, 0.0 to 0.6; P = .007). Furthermore, a beneficial effect of rs1049174 CC donor on OS and RI was observed in non-acute myelogenous leukemia patients. This study demonstrates that receipt of PTCy-haplo from rs1049174 CC donors was associated with a decreased risk of relapse/progression in the patients who underwent PTCy-haplo without ATG. Future large-scale validation studies are needed to test the significance of donor NKG2D polymorphism in the development of a new donor selection algorithm for PTCy-haplo.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid/therapy , NK Cell Lectin-Like Receptor Subfamily K/genetics , Tissue Donors , Adolescent , Adult , Aged , Cyclophosphamide/therapeutic use , Female , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Recurrence , Retrospective Studies , Transplantation Conditioning , Transplantation, Haploidentical , Young Adult
7.
Cell Mol Biol (Noisy-le-grand) ; 67(3): 92-98, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34933728

ABSTRACT

Despite the great advance in treatment, cytogenetically normal Acute myeloid leukemia (CN-AML) is still a challenging entity. The discovery of IDH1 mutation in AML together with the frequent co-mutations; NPM1 and FLT3-ITD throughs a new insight into the pathogenesis and outcome of CN-AML. Recently, there has been an increasing number of recurring mutations in other genes for which the forecasting effect is still required. Despite the large number of risk variables established, there are relatively few prognostic indicators that can help in treatment decisions in AML patients. This study aimed at recording the frequency of IDH1 and NPM1 mutations in newly diagnosed AML and, dual clinicopathological significance. IDH1 and NPM1 mutations were analyzed using High-Resolution Melting curve analysis PCR in 78 newly diagnosed AML patients; 30 pediatric and 48 adult AML patients. IDH1 mutation was detected in 6 out of the 48 adult AML cases (12.5%) and all of them had intermediate cytogenetic prognostic stratification. 5/6 mutant IDH1 patients showed NPM1 co-mutation (P-value= 0.008). Mutant IDH1 patients showed significant resistance to induction therapy (P-value <0.001) and even those who achieved complete remission were relapsed later. Within the intermediate cytogenetic group, the IDH1 mutated patients had short overall survival (HR 12.9, 95% CI (3.1- 53.45) and event-free survival (HR 15.7, 95% CI (2.99-82.72) and P-value <0.001). IDH1 mutation is closely linked to the intermediate cytogenetic stratified group and in particular old age patients and has a great impact on their survival.


Subject(s)
Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid/genetics , Mutation , Nucleophosmin/genetics , Acute Disease , Adolescent , Adult , Aged , Child , Child, Preschool , Cytogenetic Analysis/methods , Female , Humans , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Male , Middle Aged , Prognosis , Survival Analysis , Young Adult
8.
Nat Commun ; 12(1): 6436, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750374

ABSTRACT

Successful treatment of acute myeloid leukemia (AML) with chimeric antigen receptor (CAR) T cells is hampered by toxicity on normal hematopoietic progenitor cells and low CAR T cell persistence. Here, we develop third-generation anti-CD123 CAR T cells with a humanized CSL362-based ScFv and a CD28-OX40-CD3ζ intracellular signaling domain. This CAR demonstrates anti-AML activity without affecting the healthy hematopoietic system, or causing epithelial tissue damage in a xenograft model. CD123 expression on leukemia cells increases upon 5'-Azacitidine (AZA) treatment. AZA treatment of leukemia-bearing mice causes an increase in CTLA-4negative anti-CD123 CAR T cell numbers following infusion. Functionally, the CTLA-4negative anti-CD123 CAR T cells exhibit superior cytotoxicity against AML cells, accompanied by higher TNFα production and enhanced downstream phosphorylation of key T cell activation molecules. Our findings indicate that AZA increases the immunogenicity of AML cells, enhancing recognition and elimination of malignant cells by highly efficient CTLA-4negative anti-CD123 CAR T cells.


Subject(s)
Azacitidine/administration & dosage , Immunotherapy, Adoptive/methods , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid/therapy , Single-Chain Antibodies/immunology , Xenograft Model Antitumor Assays/methods , Acute Disease , Animals , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic , DNA Methylation/drug effects , Enzyme Inhibitors/administration & dosage , HEK293 Cells , HL-60 Cells , Humans , Interleukin-3 Receptor alpha Subunit/metabolism , Kaplan-Meier Estimate , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism
9.
Curr Oncol Rep ; 23(9): 109, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34272619

ABSTRACT

PURPOSE OF REVIEW: Until recently, improvement in terms of survival for patients with acute myeloid leukemia (AML) was achieved mostly in younger patients with dose intensification of conventional chemotherapy and a broadening use of allogeneic hematopoietic cell transplantation (allo-HCT) whereas the results remained dismal and very stable in patients older than 60 years. The current review highlights the recent developments in standard intensive post-remission chemotherapy, evidence for the use of recently approved agents, and discusses the relevance of measurable residual disease (MRD) measurement in treatment adaptation. RECENT FINDINGS: Current approvals of midostaurin, venetoclax, gemtuzumab ozogamicin, VYXEOS, ivosidenib, enasidenib, glasdegib, and CC-486 have changed the structure, aim, and schedule of consolidation therapy, and new, well-tolerated agents are being evaluated as maintenance therapies. Furthermore, MRD assessment has been implemented to guide the duration and type of consolidation and maintenance therapy as well as indicate the optimal timing of allo-HCT. Novel therapies have changed the structure and perspective of post-remission therapy in AML for both young and elderly patients. In addition, MRD assessment could guide the type, duration, and intensity of consolidation and maintenance therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid/therapy , Neoplasm, Residual/drug therapy , Acute Disease , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Female , Humans , Male , Middle Aged , Neoplasm, Residual/diagnosis , Remission Induction , Survival Analysis , Transplantation, Homologous
10.
Front Immunol ; 12: 683381, 2021.
Article in English | MEDLINE | ID: mdl-34220833

ABSTRACT

Natural killer (NK) cells are prominent cytotoxic and cytokine-producing components of the innate immune system representing crucial effector cells in cancer immunotherapy. Presently, various NK cell-based immunotherapies have contributed to the substantial improvement in the reconstitution of NK cells against advanced-staged and high-risk AML. Various NK cell sources, including haploidentical NK cells, adaptive NK cells, umbilical cord blood NK cells, stem cell-derived NK cells, chimeric antigen receptor NK cells, cytokine-induced memory-like NK cells, and NK cell lines have been identified. Devising innovative approaches to improve the generation of therapeutic NK cells from the aforementioned sources is likely to enhance NK cell expansion and activation, stimulate ex vivo and in vivo persistence of NK cells and improve conventional treatment response of myeloid leukemia. The tumor-promoting properties of the tumor microenvironment and downmodulation of NK cellular metabolic activity in solid tumors and hematological malignancies constitute a significant impediment in enhancing the anti-tumor effects of NK cells. In this review, we discuss the current NK cell sources, highlight ongoing interventions in enhancing NK cell function, and outline novel strategies to circumvent immunosuppressive factors in the tumor microenvironment to improve the efficacy of NK cell-based immunotherapy and expand their future success in treating myeloid leukemia.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural/immunology , Leukemia, Myeloid/therapy , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Combined Modality Therapy , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Management , Humans , Immunologic Factors/metabolism , Immunologic Memory , Immunotherapy, Adoptive/methods , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/etiology , Tumor Microenvironment/immunology
11.
Int J Mol Sci ; 22(13)2021 Jul 02.
Article in English | MEDLINE | ID: mdl-34281210

ABSTRACT

Micro RNAs (miRNAs) are a class of small non-coding RNAs that have a crucial role in cellular processes such as differentiation, proliferation, migration, and apoptosis. miRNAs may act as oncogenes or tumor suppressors; therefore, they prevent or promote tumorigenesis, and abnormal expression has been reported in many malignancies. The role of miRNA in leukemia pathogenesis is still emerging, but several studies have suggested using miRNA expression profiles as biomarkers for diagnosis, prognosis, and response to therapy in leukemia. In this review, the role of miRNAs most frequently involved in leukemia pathogenesis is discussed, focusing on the class of circulating miRNAs, consisting of cell-free RNA molecules detected in several body fluids. Circulating miRNAs could represent new potential non-invasive diagnostic and prognostic biomarkers of leukemia that are easy to isolate and characterize. The dysregulation of some miRNAs involved in both myeloid and lymphoid leukemia, such as miR-155, miR-29, let-7, and miR-15a/miR-16-1 clusters is discussed, showing their possible employment as therapeutic targets.


Subject(s)
Biomarkers/blood , Leukemia, Lymphoid/blood , Leukemia, Myeloid/blood , MicroRNAs/blood , Epigenesis, Genetic , Humans , Leukemia, Lymphoid/therapy , Leukemia, Myeloid/therapy , Molecular Targeted Therapy , RNA, Circular/metabolism , RNA, Long Noncoding/metabolism
12.
Blood ; 138(8): 649-661, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34157074

ABSTRACT

Cohesin is a multisubunit protein complex that forms a ring-like structure around DNA. It is essential for sister chromatid cohesion, chromatin organization, transcriptional regulation, and DNA damage repair and plays a major role in dynamically shaping the genome architecture and maintaining DNA integrity. The core complex subunits STAG2, RAD21, SMC1, and SMC3, as well as its modulators PDS5A/B, WAPL, and NIPBL, have been found to be recurrently mutated in hematologic and solid malignancies. These mutations are found across the full spectrum of myeloid neoplasia, including pediatric Down syndrome-associated acute megakaryoblastic leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, and de novo and secondary acute myeloid leukemias. The mechanisms by which cohesin mutations act as drivers of clonal expansion and disease progression are still poorly understood. Recent studies have described the impact of cohesin alterations on self-renewal and differentiation of hematopoietic stem and progenitor cells, which are associated with changes in chromatin and epigenetic state directing lineage commitment, as well as genomic integrity. Herein, we review the role of the cohesin complex in healthy and malignant hematopoiesis. We discuss clinical implications of cohesin mutations in myeloid malignancies and discuss opportunities for therapeutic targeting.


Subject(s)
Cell Cycle Proteins , Chromosomal Proteins, Non-Histone , Hematologic Neoplasms , Leukemia, Myeloid , Mutation , Myeloproliferative Disorders , Neoplasm Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Leukemic , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Myeloproliferative Disorders/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Cohesins
13.
Chest ; 160(5): 1783-1788, 2021 11.
Article in English | MEDLINE | ID: mdl-34102143
14.
Front Immunol ; 12: 659595, 2021.
Article in English | MEDLINE | ID: mdl-34012445

ABSTRACT

Although the majority of patients with acute myeloid leukemia (AML) treated with intensive chemotherapy achieve a complete remission (CR), many are destined to relapse if treated with intensive chemotherapy alone. Allogeneic stem cell transplant (allo-SCT) represents a pivotally important treatment strategy in fit adults with AML because of its augmented anti-leukemic activity consequent upon dose intensification and the genesis of a potent graft-versus-leukemia effect. Increased donor availability coupled with the advent of reduced intensity conditioning (RIC) regimens has dramatically increased transplant access and consequently allo-SCT is now a key component of the treatment algorithm in both patients with AML in first CR (CR1) and advanced disease. Although transplant related mortality has fallen steadily over recent decades there has been no real progress in reducing the risk of disease relapse which remains the major cause of transplant failure and represents a major area of unmet need. A number of therapeutic approaches with the potential to reduce disease relapse, including advances in induction chemotherapy, the development of novel conditioning regimens and the emergence of the concept of post-transplant maintenance, are currently under development. Furthermore, the use of genetics and measurable residual disease technology in disease assessment has improved the identification of patients who are likely to benefit from an allo-SCT which now represents an increasingly personalized therapy. Future progress in optimizing transplant outcome will be dependent on the successful delivery by the international transplant community of randomized prospective clinical trials which permit examination of current and future transplant therapies with the same degree of rigor as is routinely adopted for non-transplant therapies.


Subject(s)
Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid/therapy , Transplantation Conditioning/methods , Acute Disease , Adult , Disease-Free Survival , Humans , Leukemia, Myeloid/pathology , Remission Induction , Transplantation, Homologous
15.
Curr Hematol Malig Rep ; 16(3): 276-285, 2021 06.
Article in English | MEDLINE | ID: mdl-33890194

ABSTRACT

PURPOSE OF REVIEW: In this review, we provide a comprehensive and contemporary understanding of malignant monocytosis and provide a framework by which the appropriate diagnosis with malignant monocytosis can be rendered. RECENT FINDINGS: Increasing data support the use of molecular data to refine the diagnostic approach to persistent monocytosis. The absence of a TET2, SRSF2, or ASXL1 mutation has ≥ 90% negative predictive value for a diagnosis of CMML. These data may also reliably differentiate chronic myelomonocytic leukemia, the malignancy that is most associated with mature monocytosis, from several other diseases that can be associated with typically a lesser degree of monocytosis. These include acute myelomonocytic leukemia, acute myeloid leukemia with monocytic differentiation, myelodysplastic syndromes, and myeloproliferative neoplasms driven by BCR-ABL1, PDGFRA, PDGFRB, or FGFR1 rearrangements or PCM1-JAK2 fusions among other rarer aberrations. The combination of monocyte partitioning with molecular data in patients with persistent monocytosis may increase the predictive power for the ultimate development of CMM but has not been prospectively validated. Many conditions, both benign and malignant, can be associated with an increase in mature circulating monocytes. After reasonably excluding a secondary or reactive monocytosis, there should be a concern for and investigation of malignant monocytosis, which includes hematopathologic review of blood and marrow tissues, flow cytometric analysis, and cytogenetic and molecular studies to arrive at an appropriate diagnosis.


Subject(s)
Disease Susceptibility , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/etiology , Monocytes/metabolism , Monocytes/pathology , Age of Onset , Algorithms , Animals , Biomarkers, Tumor , Biopsy , Bone Marrow/pathology , Clinical Decision-Making , Disease Management , Gene Expression Regulation, Leukemic , Humans , Immunohistochemistry , Leukemia, Myeloid/mortality , Leukemia, Myeloid/therapy , Neoplasms, Second Primary/etiology
16.
Curr Hematol Malig Rep ; 16(3): 267-275, 2021 06.
Article in English | MEDLINE | ID: mdl-33880680

ABSTRACT

PURPOSE OF REVIEW: Monocytosis is a frequently encountered clinical condition that needs appropriate investigation due to a broad range of differential diagnoses. This review is meant to summarize the latest literature in the diagnostic testing and interpretation and offer a stepwise diagnostic approach for a patient presenting with monocytosis. RECENT FINDINGS: Basic studies have highlighted the phenotypic and functional heterogeneity in the monocyte compartment. Studies, both translational and clinical, have provided insights into why monocytosis occurs and how to distinguish the different etiologies. Flow cytometry studies have illustrated that monocyte repartitioning can distinguish chronic myelomonocytic leukemia, a prototypical neoplasm with monocytosis from other reactive or neoplastic causes. In summary, we provide an algorithmic approach to the diagnosis of a patient presenting with monocytosis and expect this document to serve as a reference guide for clinicians.


Subject(s)
Leukemia, Myeloid/diagnosis , Biomarkers, Tumor , Bone Marrow/pathology , Clonal Evolution/genetics , Clonal Evolution/immunology , Diagnosis, Differential , Disease Management , Disease Susceptibility , Flow Cytometry , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/therapy , Leukemia, Myelomonocytic, Chronic
17.
JCO Glob Oncol ; 7: 577-584, 2021 04.
Article in English | MEDLINE | ID: mdl-33891480

ABSTRACT

PURPOSE: The COVID-19 pandemic is a colossal challenge for global health; nonetheless, specific subgroups face considerably higher risks for infection and mortality. Among patients with malignant diseases, those with hematologic neoplasms are at a higher risk for poor outcomes. The objective of this study was to register treatment modifications associated with the COVID-19 pandemic and their short-term consequences in Latin America. METHODS: Multicenter, prospective, observational, cohort study including patients older than 14 years from 14 centers in four countries (Mexico, Peru, Guatemala, and Panama) who had a confirmed diagnosis of acute leukemia, and who were undergoing active treatment since the first COVID-19 case in each country until the cutoff on July 15, 2020. RESULTS: We recruited 635 patients. Treatment modifications because of the COVID-19 pandemic were reported in 40.8% of cases. The main reason for such modifications was logistic issues (55.0%) and the most frequent modification was chemotherapy delay (42.0%). A total of 13.1% patients developed COVID-19 disease, with a mortality of 37.7%. Several factors were identified as independently associated with mortality, including a diagnosis of acute myeloid leukemia (odds ratio 2.38 [95% CI, 1.47 to 3.84]; P < .001), while the use of telemedicine was identified as a protective factor (odds ratio 0.36 [95% CI, 0.18 to 0.82]; P = .014). CONCLUSION: These results highlight the collateral damage of COVID-19 in oncology patients.


Subject(s)
COVID-19/prevention & control , Leukemia, Myeloid/therapy , Medical Oncology/methods , SARS-CoV-2/isolation & purification , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/epidemiology , COVID-19/virology , Comorbidity , Epidemics , Female , Guatemala/epidemiology , Humans , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/epidemiology , Male , Mexico/epidemiology , Middle Aged , Panama/epidemiology , Peru/epidemiology , Prospective Studies , SARS-CoV-2/physiology , Young Adult
18.
Blood Cancer J ; 11(3): 64, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753715

ABSTRACT

Ecotropic viral integration site 1 (Evi1) was discovered in 1988 as a common site of ecotropic viral integration resulting in myeloid malignancies in mice. EVI1 is an oncogenic zinc-finger transcription factor whose overexpression contributes to disease progression and an aggressive phenotype, correlating with poor clinical outcome in myeloid malignancies. Despite progress in understanding the biology of EVI1 dysregulation, significant improvements in therapeutic outcome remain elusive. Here, we highlight advances in understanding EVI1 biology and discuss how this new knowledge informs development of novel therapeutic interventions. EVI1 is overexpression is correlated with poor outcome in some epithelial cancers. However, the focus of this review is the genetic lesions, biology, and current therapeutics of myeloid malignancies overexpressing EVI1.


Subject(s)
Leukemia, Myeloid/genetics , MDS1 and EVI1 Complex Locus Protein/genetics , Animals , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genomic Instability , Humans , Leukemia, Myeloid/pathology , Leukemia, Myeloid/therapy , Mutation , Protein Processing, Post-Translational , Transcriptional Activation
20.
Blood ; 137(23): 3212-3217, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33720354

ABSTRACT

Relapsed myeloid disease after allogeneic stem cell transplantation (HSCT) remains largely incurable. We previously demonstrated the potent activity of immune checkpoint blockade in this clinical setting with ipilimumab or nivolumab. To define the molecular and cellular pathways by which CTLA-4 blockade with ipilimumab can reinvigorate an effective graft-versus-leukemia (GVL) response, we integrated transcriptomic analysis of leukemic biopsies with immunophenotypic profiling of matched peripheral blood samples collected from patients treated with ipilimumab following HSCT on the Experimental Therapeutics Clinical Trials Network 9204 trial. Response to ipilimumab was associated with transcriptomic evidence of increased local CD8+ T-cell infiltration and activation. Systemically, ipilimumab decreased naïve and increased memory T-cell populations and increased expression of markers of T-cell activation and costimulation such as PD-1, HLA-DR, and ICOS, irrespective of response. However, responding patients were characterized by higher turnover of T-cell receptor sequences in peripheral blood and showed increased expression of proinflammatory chemokines in plasma that was further amplified by ipilimumab. Altogether, these data highlight the compositional T-cell shifts and inflammatory pathways induced by ipilimumab both locally and systemically that associate with successful GVL outcomes. This trial was registered at www.clinicaltrials.gov as #NCT01822509.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cell Transplantation , Ipilimumab/administration & dosage , Neoplasm Proteins , Allogeneic Cells , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Female , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/therapy , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...