Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
JCO Oncol Pract ; 18(4): e564-e573, 2022 04.
Article in English | MEDLINE | ID: mdl-34914541

ABSTRACT

INTRODUCTION: Electronic consultations (e-consults) may be a valuable tool in the current era of increased demand for hematologists. Despite the increasing use of e-consults in hematology, their optimal utilization and impact on patient outcomes and workload are largely unknown. METHODS: In this retrospective cohort study, we studied the hematology consult experience at Veterans Affairs Connecticut from 2006 to 2018. We included 7,664 hematology consults (3,240 e-consults and 4,424 face-to-face [FTF] consults) requested by 1,089 unique clinicians. RESULTS: We found that e-consults were rapidly adopted and used equally among physicians of different degrees of experience. The number of FTF consults did not decrease after the introduction of e-consult services. E-consults were preferentially used for milder laboratory abnormalities that had been less likely to result in a consult before their availability. Referring clinicians used e-consults preferentially for periprocedural management, anemia, leukopenia, and anticoagulation questions. Eighty-three percent of e-consults were resolved without needing an FTF visit in the year after the consult. Consults for pancytopenia, gammopathy, leukocytosis, and for patients with known malignancy were less likely to be resolved by e-consult. Among patients who were diagnosed with a new hematologic malignancy after their consult, having an e-consult before an FTF visit did not adversely affect survival. CONCLUSION: In summary, e-consults safely expanded delivery of hematology services in our health care system but increased total consult volume. We report novel data on what types of consults may be best suited to the electronic modality, the impact of e-consults on workload, and their optimal use and implementation.


Subject(s)
Delivery of Health Care, Integrated , Hematology , Remote Consultation , Electronics , Humans , Retrospective Studies
3.
Nat Commun ; 10(1): 366, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30664659

ABSTRACT

Comprehensive preclinical studies of Myelodysplastic Syndromes (MDS) have been elusive due to limited ability of MDS stem cells to engraft current immunodeficient murine hosts. Here we report a MDS patient-derived xenotransplantation model in cytokine-humanized immunodeficient "MISTRG" mice that provides efficient and faithful disease representation across all MDS subtypes. MISTRG MDS patient-derived xenografts (PDX) reproduce patients' dysplastic morphology with multi-lineage representation, including erythro- and megakaryopoiesis. MISTRG MDS-PDX replicate the original sample's genetic complexity and can be propagated via serial transplantation. MISTRG MDS-PDX demonstrate the cytotoxic and differentiation potential of targeted therapeutics providing superior readouts of drug mechanism of action and therapeutic efficacy. Physiologic humanization of the hematopoietic stem cell niche proves critical to MDS stem cell propagation and function in vivo. The MISTRG MDS-PDX model opens novel avenues of research and long-awaited opportunities in MDS research.


Subject(s)
Disease Models, Animal , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Myelodysplastic Syndromes/immunology , Stem Cell Niche/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers/metabolism , Cytokines/genetics , Cytokines/immunology , Gene Expression , Gene Knock-In Techniques , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Transgenic , Myelodysplastic Syndromes/pathology , Transplantation, Heterologous
4.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30652973

ABSTRACT

BACKGROUND: Protein disulfide isomerase (PDI) is a thiol isomerase secreted by vascular cells that is required for thrombus formation. Quercetin flavonoids inhibit PDI activity and block platelet accumulation and fibrin generation at the site of a vascular injury in mouse models, but the clinical effect of targeting extracellular PDI in humans has not been studied. METHODS: We conducted a multicenter phase II trial of sequential dosing cohorts to evaluate the efficacy of targeting PDI with isoquercetin to reduce hypercoagulability in cancer patients at high risk for thrombosis. Patients received isoquercetin at 500 mg (cohort A, n = 28) or 1000 mg (cohort B, n = 29) daily for 56 days, with laboratory assays performed at baseline and the end of the study, along with bilateral lower extremity compression ultrasound. The primary efficacy endpoint was a reduction in D-dimer, and the primary clinical endpoint included pulmonary embolism or proximal deep vein thrombosis. RESULTS: The administration of 1000 mg isoquercetin decreased D-dimer plasma concentrations by a median of -21.9% (P = 0.0002). There were no primary VTE events or major hemorrhages observed in either cohort. Isoquercetin increased PDI inhibitory activity in plasma (37.0% in cohort A, n = 25, P < 0.001; 73.3% in cohort B, n = 22, P < 0.001, respectively). Corroborating the antithrombotic efficacy, we also observed a significant decrease in platelet-dependent thrombin generation (cohort A median decrease -31.1%, P = 0.007; cohort B median decrease -57.2%, P = 0.004) and circulating soluble P selectin at the 1000 mg isoquercetin dose (median decrease -57.9%, P < 0.0001). CONCLUSIONS: Isoquercetin targets extracellular PDI and improves markers of coagulation in advanced cancer patients. TRIAL REGISTRATION: Clinicaltrials.gov NCT02195232. FUNDING: Quercegen Pharmaceuticals; National Heart, Lung, and Blood Institute (NHLBI; U54HL112302, R35HL135775, and T32HL007917); and NHLBI Consortium Linking Oncology and Thrombosis (U01HL143365).


Subject(s)
Neoplasms/complications , Protein Disulfide-Isomerases/antagonists & inhibitors , Quercetin/analogs & derivatives , Venous Thromboembolism/prevention & control , Aged , Blood Coagulation/drug effects , Dose-Response Relationship, Drug , Feasibility Studies , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Neoplasms/blood , Quercetin/administration & dosage , Treatment Outcome , Venous Thromboembolism/blood , Venous Thromboembolism/epidemiology , Venous Thromboembolism/etiology
5.
Exp Hematol Oncol ; 7: 15, 2018.
Article in English | MEDLINE | ID: mdl-29989046

ABSTRACT

Bullous hemorrhagic dermatosis (BHD) is a systemic side-effect of low molecular weight heparin, characterized by multiple intra-epidermal hemorrhages distant from the site of injection. There have been several small case series and literature reviews on BHD, but none have captured a complete set of reported patients. We sought to describe a case of BHD with late diagnosis and completely summarize the existing English and Spanish literature with searches of Pubmed, Scopus, Ovid Embase and Ovid Medline. After narrowing to 33 relevant reports, we describe 90 reported cases worldwide from 2004 to 2017, in addition to a new case from our institution as a means of comparison. We found that BHD was common in elderly men (mean age 72 ± 12; male:female, 1.9:1) and typically occurred within 7 days of administration of anticoagulation (median 7 days ± 6.4) usually with enoxaparin use (66% of cases). Lesions occurred primarily on the extremities only (67.9% of cases). Coagulation testing was most often normal before administration, and the majority of patients had coagulation testing in therapeutic range during treatment. Most practitioners stopped anticoagulation if continued therapeutic intervention was no longer required (57% of cases), or changed therapy to another anticoagulation if continued treatment was required (14.3% of cases). Therapy was continued outright in 23% of patients. The lesions usually resolved within 2 weeks (mean days, 13.0 ± 7.4). There was no difference in time to resolution between patients who continued the culprit anticoagulant or changed to a different anticoagulant, and those who discontinued anticoagulation altogether (13.9 days vs. 12.1, p = 0.49). Four deaths have been reported in this clinical context, two specified as intracranial hemorrhage. These deaths were unrelated to the occurrence of BHD. Continuation of low-molecular weight heparins appeared to be safe in patients with BHD.

6.
Br J Haematol ; 182(4): 495-503, 2018 08.
Article in English | MEDLINE | ID: mdl-29808907

ABSTRACT

Smouldering multiple myeloma (SMM) is associated with increased risk of progression to multiple myeloma within 2 years, with no approved treatments. Elotuzumab has been shown to promote natural killer (NK) cell stimulation and antibody-dependent cellular cytotoxicity (ADCC) in vitro. CD56dim (CD56dim /CD16+ /CD3- /CD45+ ) NK cells represent the primary subset responsible for elotuzumab-induced ADCC. In this phase II, non-randomized study (NCT01441973), patients with SMM received elotuzumab 20 mg/kg intravenously (cycle 1: days 1, 8; monthly thereafter) or 10 mg/kg (cycles 1, 2: weekly; every 2 weeks thereafter). The primary endpoint was the relationship between baseline proportion of bone marrow-derived CD56dim NK cells and maximal M protein reduction; secondary endpoints included overall response rate (ORR) and progression-free survival (PFS). Fifteen patients received 20 mg/kg and 16 received 10 mg/kg; combined data arepresented. At database lock (DBL, September 2014), no association was found between baseline CD56dim NK cell proportion and maximal M protein reduction. With minimum 28 months' follow-up (DBL: January 2016), ORR (90% CI) was 10% (2·7-23·2) and 2-year PFS rate was 69% (52-81%). Upper respiratory tract infections occurred in 18/31 (58%) patients. Four (13%) patients experienced infusion reactions, all grade 1-2. Elotuzumab plus lenalidomide/dexamethasone is under investigation for SMM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Smoldering Multiple Myeloma , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD/blood , Antigens, CD/immunology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease-Free Survival , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lenalidomide/administration & dosage , Lenalidomide/adverse effects , Male , Middle Aged , Neoplasm Proteins/blood , Neoplasm Proteins/immunology , Smoldering Multiple Myeloma/blood , Smoldering Multiple Myeloma/drug therapy , Smoldering Multiple Myeloma/immunology , Smoldering Multiple Myeloma/mortality , Survival Rate
8.
Clin Case Rep ; 4(12): 1120-1121, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27980745

ABSTRACT

Ibrutinib is a first-in-class inhibitor of Bruton's tyrosine kinase, which is approved for use in chronic lymphocytic leukemia, mantle cell lymphoma, and Waldenstrom's macroglobulinemia. Although ibrutinib has been linked to an increased incidence of atrial fibrillation, this is the first report of an association with nonischemic cardiomyopathy and ventricular arrhythmia.

10.
Hematol Oncol Clin North Am ; 26(2): 231-52, vii, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22463825

ABSTRACT

Thrombocytopenia, usually defined as a platelet count of less than 150,000/µL, is a common reason for a hematology consult in both the inpatient and outpatient setting. In most patients, the cause of the thrombocytopenia can be identified and treated. This article reviews the clinical approach to the patient with thrombocytopenia, the mechanisms that underlie it, and the laboratory tests available to investigate it. A practical approach to the investigation and management of thrombocytopenia in the clinical settings commonly encountered by the hematology consultant is then described.


Subject(s)
Thrombocytopenia/etiology , Drug-Related Side Effects and Adverse Reactions , Humans , Infections/complications , Neoplasms/complications , Thrombocytopenia/diagnosis , Transfusion Reaction , Transplantation/adverse effects
11.
Blood ; 119(10): 2302-9, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22234692

ABSTRACT

Tumor microenvironment (TME) is commonly implicated in regulating the growth of tumors, but whether it can directly alter the genetics of tumors is not known. Genomic instability and dendritic cell (DC) infiltration are common features of several cancers, including multiple myeloma (MM). Mechanisms underlying genomic instability in MM are largely unknown. Here, we show that interaction between myeloma and DCs, but not monocytes, leads to rapid induction of the genomic mutator activation-induced cytidine deaminase (AID) and AID-dependent DNA double-strand breaks (DSBs) in myeloma cell lines as well as primary MM cells. Both myeloid as well as plasmacytoid DCs have the capacity to induce AID in tumor cells. The induction of AID and DSBs in tumor cells by DCs requires DC-tumor contact and is inhibited by blockade of receptor activator of NF-κB/receptor activator of NF-κB ligand (RANKL) interactions. AID-mediated genomic damage led to altered tumorigenicity and indolent behavior of tumor cells in vivo. These data show a novel pathway for the capacity of DCs in the TME to regulate genomic integrity. DC-mediated induction of AID and resultant genomic damage may therefore serve as a double-edged sword and be targeted by approaches such as RANKL inhibition already in the clinic.


Subject(s)
Cytidine Deaminase/genetics , Dendritic Cells/metabolism , Genomic Instability , Multiple Myeloma/genetics , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival/genetics , Cells, Cultured , Coculture Techniques , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , NF-kappa B/metabolism , RANK Ligand/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Cells, Cultured
12.
J Biol Chem ; 280(47): 39016-23, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16186125

ABSTRACT

Alpha-hemoglobin-stabilizing protein (AHSP) is an erythroid protein that binds and stabilizes alpha-hemoglobin during normal erythropoiesis and in pathological states of alpha-hemoglobin excess. AHSP has been proposed as a candidate gene in some Heinz body hemolytic anemias and as a modifier gene in the beta-thalassemia syndromes. To gain additional insight into the molecular mechanisms controlling the erythroid-specific expression of the AHSP gene and provide the necessary tools for further genetic studies of these disorders, we have initiated identification and characterization of the regulatory elements controlling the human AHSP gene. We mapped the 5'-end of the AHSP erythroid cDNA and cloned the 5'-flanking genomic DNA containing the putative AHSP gene promoter. In vitro studies using transfection of promoter/reporter plasmids in human tissue culture cell lines, DNase I footprinting analyses and gel mobility shift assays, identified an AHSP gene erythroid promoter with functionally important binding sites for GATA-1- and Oct-1-related proteins. In transgenic mice, a reporter gene directed by a minimal human AHSP promoter was expressed in bone marrow, spleen, and reticulocytes, but not in nonerythroid tissues. In vivo studies using chromatin immunoprecipitation assays demonstrated hyperacetylation of the promoter region and occupancy by GATA-1. The AHSP promoter is an excellent candidate region for mutations associated with decreased AHSP gene expression.


Subject(s)
Blood Proteins/genetics , GATA1 Transcription Factor/metabolism , Molecular Chaperones/genetics , Octamer Transcription Factor-1/metabolism , Animals , Base Sequence , Binding Sites/genetics , Blood Proteins/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Erythropoiesis/genetics , Gene Expression , Globins/genetics , HeLa Cells , Humans , Mice , Mice, Transgenic , Molecular Chaperones/metabolism , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
13.
J Biol Chem ; 279(53): 55024-33, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15456760

ABSTRACT

Alpha-spectrin is a membrane protein critical for the flexibility and stability of the erythrocyte. We are attempting to identify and characterize the molecular mechanisms controlling the erythroid-specific expression of the alpha-spectrin gene. Previously, we demonstrated that the core promoter of the human alpha-spectrin gene directed low levels of erythroid-specific expression only in the early stages of erythroid differentiation. We have now identified a region 3' of the core promoter that contains a DNase I hypersensitive site and directs high level, erythroid-specific expression in reporter gene/transfection assays. In vitro DNase I footprinting and electrophoretic mobility shift assays identified two functional GATA-1 sites in this region. Both GATA-1 sites were required for full activity, suggesting that elements binding to each site interact in a combinatorial manner. This region did not demonstrate enhancer activity in any orientation or position relative to either the alpha-spectrin core promoter or the thymidine kinase promoter in reporter gene assays. In vivo studies using chromatin immunoprecipitation assays demonstrated hyperacetylation of this region and occupancy by GATA-1 and CBP (cAMP-response element-binding protein (CREB)-binding protein). These results demonstrate that a region 3' of the alpha-spectrin core promoter contains a GATA-1-dependent positive regulatory element that is required in its proper genomic orientation. This is an excellent candidate region for mutations associated with decreased alpha-spectrin gene expression in patients with hereditary spherocytosis and hereditary pyropoikilocytosis.


Subject(s)
Erythrocytes/metabolism , Promoter Regions, Genetic , Spectrin/biosynthesis , Spectrin/genetics , Base Sequence , Binding Sites , CREB-Binding Protein , Cell Differentiation , Cell Membrane/metabolism , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , DNA/chemistry , DNA Primers/chemistry , DNA, Complementary/metabolism , DNA-Binding Proteins/chemistry , Deoxyribonuclease I/chemistry , Erythroid-Specific DNA-Binding Factors , Ethidium/pharmacology , Exons , GATA1 Transcription Factor , Genes, Reporter , HeLa Cells , Humans , Immunoprecipitation , Introns , K562 Cells , Luciferases/metabolism , Models, Genetic , Molecular Sequence Data , Mutation , Nuclear Proteins/chemistry , Plasmids/metabolism , Temperature , Trans-Activators/chemistry , Transcription Factors/chemistry , Transfection
14.
J Biol Chem ; 277(44): 41563-70, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12196550

ABSTRACT

alpha-Spectrin is a highly expressed membrane protein critical for the flexibility and stability of the erythrocyte. Qualitative and quantitative defects of alpha-spectrin are present in the erythrocytes of many patients with abnormalities of red blood cell shape including hereditary spherocytosis and elliptocytosis. We wished to determine the regulatory elements that determine the erythroid-specific expression of the alpha-spectrin gene. We mapped the 5' end of the alpha-spectrin erythroid cDNA and cloned the 5' flanking genomic DNA containing the putative alpha-spectrin gene promoter. Using transfection of promoter/reporter plasmids in human tissue culture cell lines, in vitro DNase I footprinting analyses, and gel mobility shift assays, an alpha-spectrin gene erythroid promoter with binding sites for GATA-1- and NF-E2-related proteins was identified. Both binding sites were required for full promoter activity. In transgenic mice, a reporter gene directed by the alpha-spectrin promoter was expressed in yolk sac, fetal liver, and erythroid cells of bone marrow but not adult reticulocytes. No expression of the reporter gene was detected in nonerythroid tissues. We conclude that this alpha-spectrin gene promoter contains the sequences necessary for low level expression in erythroid progenitor cells.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , Promoter Regions, Genetic , Spectrin/genetics , Transcription Factors/metabolism , 5' Flanking Region , Animals , Base Sequence , Binding Sites , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Erythroid-Specific DNA-Binding Factors , Erythropoiesis , GATA1 Transcription Factor , HeLa Cells , Humans , K562 Cells , Mice , Mice, Transgenic , Molecular Sequence Data , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit
SELECTION OF CITATIONS
SEARCH DETAIL
...