Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
1.
J Med Econ ; 27(1): 109-125, 2024.
Article in English | MEDLINE | ID: mdl-38085684

ABSTRACT

AIM: To evaluate the real-world comparative effectiveness and the cost-effectiveness, from a UK National Health Service perspective, of natalizumab versus fingolimod in patients with rapidly evolving severe relapsing-remitting multiple sclerosis (RES-RRMS). METHODS: Real-world data from the MSBase Registry were obtained for patients with RES-RRMS who were previously either naive to disease-modifying therapies or had been treated with interferon-based therapies, glatiramer acetate, dimethyl fumarate, or teriflunomide (collectively known as BRACETD). Matched cohorts were selected by 3-way multinomial propensity score matching, and the annualized relapse rate (ARR) and 6-month-confirmed disability worsening (CDW6M) and improvement (CDI6M) were compared between treatment groups. Comparative effectiveness results were used in a cost-effectiveness model comparing natalizumab and fingolimod, using an established Markov structure over a lifetime horizon with health states based on the Expanded Disability Status Scale. Additional model data sources included the UK MS Survey 2015, published literature, and publicly available sources. RESULTS: In the comparative effectiveness analysis, we found a significantly lower ARR for patients starting natalizumab compared with fingolimod (rate ratio [RR] = 0.65; 95% confidence interval [CI], 0.57-0.73) or BRACETD (RR = 0.46; 95% CI, 0.42-0.53). Similarly, CDI6M was higher for patients starting natalizumab compared with fingolimod (hazard ratio [HR] = 1.25; 95% CI, 1.01-1.55) and BRACETD (HR = 1.46; 95% CI, 1.16-1.85). In patients starting fingolimod, we found a lower ARR (RR = 0.72; 95% CI, 0.65-0.80) compared with starting BRACETD, but no difference in CDI6M (HR = 1.17; 95% CI, 0.91-1.50). Differences in CDW6M were not found between the treatment groups. In the base-case cost-effectiveness analysis, natalizumab dominated fingolimod (0.302 higher quality-adjusted life-years [QALYs] and £17,141 lower predicted lifetime costs). Similar cost-effectiveness results were observed across sensitivity analyses. CONCLUSIONS: This MSBase Registry analysis suggests that natalizumab improves clinical outcomes when compared with fingolimod, which translates to higher QALYs and lower costs in UK patients with RES-RRMS.


There are several medications used to treat people with relapsing remitting multiple sclerosis, such as interferon-based therapies (Betaferon/Betaseron (US), Rebif, Avonex, Extavia), glatiramer acetate (Copaxone), teriflunomide (Aubagio), and dimethyl fumarate (Tecfidera), collectively named BRACETD. Other treatments for multiple sclerosis (MS) have a narrower use, such as natalizumab (Tysabri) or fingolimod (Gilenya), among others.This study objective was to assess how well natalizumab and fingolimod helped treating MS (clinical effectiveness) and subsequently estimate what the cost of these treatments is in comparison to the benefit they bring to people with rapidly evolving severe MS that use them in the United Kingdom (UK) (cost-effectiveness).We used an international disease registry (MSBase), which collects clinical data from people with MS in various centers around the world to compare the effectiveness of natalizumab, fingolimod and BRACETD treatments. We used a technique called propensity score matching to obtain results from comparable patient groups. People treated with natalizumab had better disease control, namely with fewer relapses and higher improvement on their disability level, than patients on fingolimod or BRACETD. Conversely, there were no differences between each group of people on a measure called disability worsening.Based on these clinical results, we built an economic model that simulates the lifetime costs and consequences of treating people with MS with natalizumab in comparison with fingolimod. We found that using natalizumab was less costly and was more effective compared to using fingolimod in UK patients.


Subject(s)
Multiple Sclerosis, Relapsing-Remitting , Multiple Sclerosis , Humans , Natalizumab/therapeutic use , Fingolimod Hydrochloride/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Immunosuppressive Agents/therapeutic use , Multiple Sclerosis/drug therapy , Cost-Effectiveness Analysis , Cost-Benefit Analysis , State Medicine , United Kingdom
2.
Vet J ; 300-302: 106031, 2023.
Article in English | MEDLINE | ID: mdl-37778652

ABSTRACT

Bovine respiratory disease (BRD) is one of the leading causes of mortality and morbidity in calves across diverse management systems. Despite expert opinion often citing the influence of housing environment on the level of respiratory disease in calf groups, there have been few reviews of environmental factors that predispose to BRD. This systematic review was undertaken to identify the measurable environmental variables associated with respiratory disease in housed preweaned calves. To achieve this Pubmed, CAB Direct and Scopus databases were searched. To be considered for inclusion, publications had to be fully published in English, published before 24 November, 2022 and include at least one measurable/ manipulated environmental variable and a standardized method of BRD detection. Twelve publications were included in this review. These examined a wide range of risk factors including air microbial count (four publications), air particulate matter (one publication); air endotoxins (one publication) and air ammonia (four publications). From the included publications, a statistically significant relationship to BRD was identified in 2/4 examining air microbial count, 1/1 examining air particulate matter, 1/1 examining air endotoxins and 2/4 examining air ammonia. This review indicated a paucity of evidence from the peer-review literature demonstrating a significant association between the many investigated exposure factors and BRD occurrence. An optimal environment for housed calves could not be clearly identified in this review.


Subject(s)
Bovine Respiratory Disease Complex , Cattle Diseases , Respiration Disorders , Respiratory Tract Diseases , Animals , Cattle , Ammonia , Housing , Cattle Diseases/epidemiology , Respiratory Tract Diseases/veterinary , Risk Factors , Respiration Disorders/veterinary , Bovine Respiratory Disease Complex/epidemiology , Bovine Respiratory Disease Complex/etiology
3.
Leukemia ; 37(11): 2197-2208, 2023 11.
Article in English | MEDLINE | ID: mdl-37689825

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous disease characterized by clonal expansion of myeloid blasts in the bone marrow (BM). Despite advances in therapy, the prognosis for AML patients remains poor, and there is a need to identify novel molecular pathways regulating tumor cell survival and proliferation. F-box ubiquitin E3 ligase, FBXO21, has low expression in AML, but expression correlates with survival in AML patients and patients with higher expression have poorer outcomes. Silencing FBXO21 in human-derived AML cell lines and primary patient samples leads to differentiation, inhibition of tumor progression, and sensitization to chemotherapy agents. Additionally, knockdown of FBXO21 leads to up-regulation of cytokine signaling pathways. Through a mass spectrometry-based proteomic analysis of FBXO21 in AML, we identified that FBXO21 ubiquitylates p85α, a regulatory subunit of the phosphoinositide 3-kinase (PI3K) pathway, for degradation resulting in decreased PI3K signaling, dimerization of free p85α and ERK activation. These findings reveal the ubiquitin E3 ligase, FBXO21, plays a critical role in regulating AML pathogenesis, specifically through alterations in PI3K via regulation of p85α protein stability.


Subject(s)
F-Box Proteins , Leukemia, Myeloid, Acute , Humans , Cell Proliferation/physiology , F-Box Proteins/genetics , Leukemia, Myeloid, Acute/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proteomics , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism
4.
Vet J ; 300-302: 106032, 2023.
Article in English | MEDLINE | ID: mdl-37757972

ABSTRACT

Bovine respiratory disease (BRD) is a challenge in all housed farming systems that raise calves. Farm to farm variation in BRD prevalence can be partially attributed to variation in host immunity, pathogens and housing environment. Unlike host immunity and BRD pathogens, housing environment has not been well investigated. The objective of this systematic review was to identify the measurable environmental variables associated with BRD in housed preweaned calves. Pubmed, CAB Direct and Scopus databases were searched. To be considered for inclusion publications had to be published in English, before 24 November, 2022 and include at least one measurable/ manipulated environmental variable and a standardized method of BRD detection. In total 12 publications were included in this review. In this second part of the systematic review the environmental variables identified were; temperature (9 publications); relative humidity (8 publications); bedding (5 publications); ventilation (1 publication); air CO2 concentration (1 publication) and air velocity (4 publications). Of the publications that were examined a statistically significant relationship to BRD was identified in 4/9 publications examining temperature, 3/8 examining relative humidity, 2/4 examining air velocity, 2/5 examining bedding, 0/1 examining ventilation rates and 0/1 examining CO2 concentration. From this review it is clear high airspeed at calf level should be avoided as should deep, wet pack bedding. The relationship between BRD prevalence and both high and low temperature requires more investigation to identify temperature thresholds associated with increased risk of BRD as well as the most influential modifiers. An optimal environment for housed calves could not be clearly identified in this review.


Subject(s)
Bovine Respiratory Disease Complex , Cattle Diseases , Respiration Disorders , Respiratory Tract Diseases , Animals , Cattle , Humidity , Housing , Temperature , Carbon Dioxide , Cattle Diseases/epidemiology , Cattle Diseases/etiology , Respiratory Tract Diseases/veterinary , Respiration Disorders/veterinary , Bovine Respiratory Disease Complex/epidemiology , Bovine Respiratory Disease Complex/etiology
5.
Transplant Cell Ther ; 28(9): 560-571, 2022 09.
Article in English | MEDLINE | ID: mdl-35691572

ABSTRACT

The development of chimeric antigen receptor (CAR) T cells began as a means toward specific yet modular therapies against cancer. Recent advancements in several CAR T cell therapies show the promise of cellular immunotherapy in cancer treatment. CAR T cell therapy is still immature, however, and improvements are needed to fully realize its curative potential. The approved CAR T cells are designed with simple logic capabilities; an antigen sensor that, when bound to the target antigen, triggers costimulation domains and native T cell activation. This single-type sensor and native activation design, although capable, also has severe limitations. Reliance on a single-type sensor leads to unwanted toxicity toward antigen-expressing normal tissues, and unmodulated activation leads to unwanted cytokine toxicity. Synthetic biology (SB) offers a powerful solution to these limitations: modular receptors with customizable sensors and output behaviors that enable higher Boolean logic. SB T cells already have shown incredible capabilities, such as multiple-antigen discrimination and improved persistence. In light of these results, cellular immunotherapy may already be branching into a new subfield that we term here as "synthetic immunotherapy." Here we review the current logic capabilities of CAR T cells, the resulting limitations, and the engineering undertaken to address these issues. We then discuss several tools of SB and show how SB CAR T cells pave the way for synthetic immunotherapy.


Subject(s)
Immunotherapy , Receptors, Antigen, T-Cell , Immunologic Factors , Immunotherapy, Adoptive , Logic , T-Lymphocytes
6.
J Geriatr Oncol ; 13(6): 871-874, 2022 07.
Article in English | MEDLINE | ID: mdl-35450817

ABSTRACT

INTRODUCTION: Survival benefit associated with intensive over low-intensity chemotherapy in older adults with acute myeloid leukemia (AML) is controversial. Geriatric assessment and genetic risk categories correlate with survival following intensive chemotherapy in older adults with AML and can guide treatment selection. MATERIALS AND METHODS: In a single-center trial, we integrated both geriatric assessment, and genetic risk categories to personalize selection of intensive versus low-intensity chemotherapy in older adults ≥60 years with AML (NCT03226418). In the present report, we demonstrate feasibility of this approach. RESULTS: Broad eligibility criteria and co-management of patients with community oncologists allowed enrollment of 45% of all patients with AML treated at our center during the study period. The median time from enrollment to therapy initiation was two days (range 0-9). Over half of the trial patients had a score of ≥3 on hematopoietic cell transplantation comorbidity index, impairment in physical function (Short Physical Performance Battery), and Montreal Cognitive Assessment. Three fit patients received intensive chemotherapy, whereas other patients received low-intensity chemotherapy. Mortality at 30 days from diagnosis was 3.7% (95% confidence interval [CI] 0.7-18.3%) and at 90 days was 29.6% (95% CI 15.9-48.5%). One-year overall survival was 66% (95% CI 60-87%). DISCUSSION: Our data demonstrate the feasibility of integrating geriatric assessment in precision oncology trials to define fitness for intensive chemotherapy. Broad eligibility criteria and academic-community collaboration can expand access to clinical trials.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Aged , Geriatric Assessment , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Precision Medicine , Risk Factors , Treatment Outcome
7.
Leukemia ; 36(5): 1296-1305, 2022 05.
Article in English | MEDLINE | ID: mdl-35177813

ABSTRACT

Acute myeloid leukemia (AML) is a devastating cancer affecting the hematopoietic system. Previous research has relied on RNA sequencing and microarray techniques to study the downstream effects of genomic alterations. While these studies have proven efficacious, they fail to capture the changes that occur at the proteomic level. To interrogate the effect of protein expression alterations in AML, we performed a quantitative mass spectrometry in parallel with RNAseq analysis using AML mouse models. These combined results identified 34 proteins whose expression was upregulated in AML tumors, but strikingly, were unaltered at the transcriptional level. Here we focus on mitochondrial electron transfer proteins ETFA and ETFB. Silencing of ETFA and ETFB led to increased mitochondrial activity, mitochondrial stress, and apoptosis in AML cells, but had little to no effect on normal human CD34+ cells. These studies identify a set of proteins that have not previously been associated with leukemia and may ultimately serve as potential targets for therapeutic manipulation to hinder AML progression and help contribute to our understanding of the disease.


Subject(s)
Leukemia, Myeloid, Acute , Mitochondrial Proteins , Animals , Apoptosis , Drug Discovery , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/pharmacology , Proteomics
8.
Blood ; 136(21): 2373-2385, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32929473

ABSTRACT

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. It is generally considered that CBFß-SMMHC, the fusion protein encoded by CBFB-MYH11, is a dominant negative repressor of RUNX1. However, recent findings challenge the RUNX1-repression model for CBFß-SMMHC-mediated leukemogenesis. To definitively address the role of Runx1 in CBFB-MYH11-induced leukemia, we crossed conditional Runx1 knockout mice (Runx1f/f) with conditional Cbfb-MYH11 knockin mice (Cbfb+/56M). On Mx1-Cre activation in hematopoietic cells induced by poly (I:C) injection, all Mx1-CreCbfb+/56M mice developed leukemia in 5 months, whereas no leukemia developed in Runx1f/fMx1-CreCbfb+/56M mice, and this effect was cell autonomous. Importantly, the abnormal myeloid progenitors (AMPs), a leukemia-initiating cell population induced by Cbfb-MYH11 in the bone marrow, decreased and disappeared in Runx1f/fMx1-CreCbfb+/56M mice. RNA-seq analysis of AMP cells showed that genes associated with proliferation, differentiation blockage, and leukemia initiation were differentially expressed between Mx1-CreCbfb+/56M and Runx1f/fMx1-CreCbfb+/56M mice. In addition, with the chromatin immunocleavage sequencing assay, we observed a significant enrichment of RUNX1/CBFß-SMMHC target genes in Runx1f/fMx1-CreCbfb+/56M cells, especially among downregulated genes, suggesting that RUNX1 and CBFß-SMMHC mainly function together as activators of gene expression through direct target gene binding. These data indicate that Runx1 is indispensable for Cbfb-MYH11-induced leukemogenesis by working together with CBFß-SMMHC to regulate critical genes associated with the generation of a functional AMP population.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Gene Expression Regulation, Leukemic , Leukemia, Experimental/genetics , Myeloid Cells/metabolism , Neoplasm Proteins/physiology , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/physiology , Transcriptional Activation , Animals , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Knock-In Techniques , Humans , Leukemia, Experimental/etiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Neoplastic Stem Cells/cytology , Oncogene Proteins, Fusion/genetics , Poly I-C/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA-Seq , Single-Cell Analysis
9.
Proc Natl Acad Sci U S A ; 117(38): 23626-23635, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32883883

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) formation and lineage differentiation involve gene expression programs orchestrated by transcription factors and epigenetic regulators. Genetic disruption of the chromatin remodeler chromodomain-helicase-DNA-binding protein 7 (CHD7) expanded phenotypic HSPCs, erythroid, and myeloid lineages in zebrafish and mouse embryos. CHD7 acts to suppress hematopoietic differentiation. Binding motifs for RUNX and other hematopoietic transcription factors are enriched at sites occupied by CHD7, and decreased RUNX1 occupancy correlated with loss of CHD7 localization. CHD7 physically interacts with RUNX1 and suppresses RUNX1-induced expansion of HSPCs during development through modulation of RUNX1 activity. Consequently, the RUNX1:CHD7 axis provides proper timing and function of HSPCs as they emerge during hematopoietic development or mature in adults, representing a distinct and evolutionarily conserved control mechanism to ensure accurate hematopoietic lineage differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins , Hematopoiesis , Animals , Cell Differentiation , Cell Line , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Hematopoietic Stem Cells , Humans , Male , Mice , Spleen/cytology , Zebrafish
10.
Leuk Res ; 96: 106409, 2020 09.
Article in English | MEDLINE | ID: mdl-32652328

ABSTRACT

Acute myeloid leukemia (AML) is often characterized by the presence of specific and recurrent chromosomal abnormalities. Current treatments have greatly increased remission rate, but relapse still occurs. Therefore, novel therapeutic approaches are required. Previously, using a conditional Cbfb-MYH11 knockin mouse model, we showed that Cbfb-MYH11 induces the expression of a cytokine receptor, IL1RL1. Treatment with IL-33, the only known ligand of IL1RL1, promotes leukemia cell survival in vitro. We further found that IL1RL1+ cells survive better with chemotherapy than IL1RL1- population. However, the mechanism is not clear. Here, we show that IL-33 treatment decreased drug sensitivity in the human inv(16) AML cell line ME-1. By RT-PCR, we found that IL-33 increased the expression of IL-4 and IL-6 and led to the activation of both p38 MAPK and NF-κB. We also showed that IL-33 decreased apoptosis with increased phosphorylation of p38 MAPK. Moreover, pre-treatment with MAPK inhibitor attenuated the phosphorylation of p38 enhanced by IL-33 and reversed the anti-apoptotic effect by IL-33. Taken together, our findings give news insights into the potential mechanism of the anti-apoptotic effect by IL-33/IL1RL1 axis in AML which will help in future drug development.


Subject(s)
Apoptosis , Core Binding Factor beta Subunit/physiology , Interleukin-33/pharmacology , Leukemia, Myeloid, Acute/pathology , Myosin Heavy Chains/physiology , NF-kappa B/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Gene Expression Regulation, Leukemic , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , Phosphorylation , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases/genetics
11.
Cancer Gene Ther ; 27(1-2): 45-55, 2020 02.
Article in English | MEDLINE | ID: mdl-31028289

ABSTRACT

Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults and is associated with poor long-term survival often owing to relapse. Current treatments for AML are associated with considerable toxicity and are frequently not effective after relapse. Thus, it is important to develop novel therapeutic strategies. Short interfering RNA (siRNA)-based therapeutics targeting key oncogenes have been proposed as treatments for AML. We recently developed novel siRNA delivery polycations (PCX) based on AMD3100 (plerixafor), an FDA-approved inhibitor of the CXC chemokine receptor 4 (CXCR4). Inhibitors of CXCR4 have been shown to sensitize leukemia cells to chemotherapy. Therefore, PCX has the potential to target leukemia cells via two mechanisms: inhibition of CXCR4 and delivery of siRNAs against critical genes. In this report, we show that PCX exerts a cytotoxic effect on leukemia cells more effectively than other CXCR4 inhibitors, including AMD3100. In addition, we show that PCX can deliver siRNAs against the transcription factor RUNX1 to mouse and human leukemia cells. Overall, our study provides the first evidence that dual-function PCX/siRNA nanoparticles can simultaneously inhibit CXCR4 and deliver siRNAs, targeting key oncogenes in leukemia cells and that PCX/siRNA has clinical potential for the treatment of AML.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Leukemia, Myeloid, Acute/drug therapy , RNA, Small Interfering/administration & dosage , Receptors, CXCR4/antagonists & inhibitors , Animals , Benzylamines/administration & dosage , Cell Line, Tumor , Cholesterol/chemistry , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Cyclams/administration & dosage , Disease Models, Animal , Female , Gene Knock-In Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Transgenic , Nanoparticles/chemistry , Oncogene Proteins, Fusion/genetics , Polyelectrolytes/chemistry , RNA, Small Interfering/genetics , Receptors, CXCR4/genetics
12.
Cancers (Basel) ; 11(11)2019 Nov 03.
Article in English | MEDLINE | ID: mdl-31684170

ABSTRACT

The hematopoietic system is maintained throughout life by stem cells that are capable of differentiating into all hematopoietic lineages. An intimate balance between self-renewal, differentiation, and quiescence is required to maintain hematopoiesis and disruption of this balance can result in malignant transformation. FBXO9, the substrate recognition component from the SCF E3 ubiquitin ligase family, is downregulated in patients with acute myeloid leukemia (AML) compared to healthy bone marrow, and this downregulation is particularly evident in patients with inv(16) AML. To study FBXO9 in malignant hematopoiesis, we generated a conditional knockout mouse model using a novel CRISPR/Cas9 strategy. Deletion of Fbxo9 in the murine hematopoietic system showed no adverse effects on stem and progenitor cell function but in AML lead to markedly accelerated and aggressive leukemia development in mice with inv(16). Not only did Fbxo9 play a role in leukemia initiation but it also functioned to maintain AML activity and promote disease progression. Quantitative mass spectrometry from primary tumors reveals tumors lacking Fbxo9 highly express proteins associated with metastasis and invasion as well as components of the ubiquitin proteasome system. We confirmed that the loss of FBXO9 leads to increased proteasome activity and tumors cells were more sensitive to in vitro proteasome inhibition with bortezomib, suggesting that FBXO9 expression may predict patients' response to bortezomib.

13.
Mol Cancer Res ; 17(6): 1241-1252, 2019 06.
Article in English | MEDLINE | ID: mdl-30814129

ABSTRACT

Acute myeloid leukemia (AML) is a neoplastic disease characterized by the uncontrolled proliferation and accumulation of immature myeloid cells. A common mutation in AML is the inversion of chromosome 16 [inv (16)], which generates a fusion between the genes for core binding factor beta (CBFB) and smooth muscle myosin heavy chain gene (MYH11), forming the oncogene CBFB-MYH11. The expressed protein, CBFß-SMMHC, forms a heterodimer with the key hematopoietic transcription factor RUNX1. Although CBFß-SMMHC was previously thought to dominantly repress RUNX1, recent work suggests that CBFß-SMMHC functions together with RUNX1 to activate transcription of specific target genes. However, the mechanism of this activity or a requirement for additional cofactors is not known. Here, we show that the epigenetic regulator histone deacetylase 1 (HDAC1) forms a complex with CBFß-SMMHC, colocalizes with RUNX1 and CBFß-SMMHC on the promoters of known fusion protein target genes, and that Hdac1 is required for expression of these genes. These results imply that HDAC1 is an important component of the CBFß-SMMHC transcriptional complex, and that leukemia cells expressing the fusion protein may be sensitive to treatment with HDAC1 inhibitors. Using a knock-in mouse model expressing CBFß-SMMHC, we found that in vivo treatment with the HDAC1 inhibitor entinostat decreased leukemic burden, and induced differentiation and apoptosis of leukemia cells. Together, these results demonstrate that HDAC1 is an important cofactor of CBFß-SMMHC and a potential therapeutic target in inv (16) AML. IMPLICATIONS: This report describes a novel role for HDAC1 as a cofactor for the leukemogenic fusion protein CBFß-SMMHC and shows that inhibitors of HDAC1 effectively target leukemia cells expressing the fusion protein in vivo.


Subject(s)
Chromosome Inversion/genetics , Core Binding Factor beta Subunit/genetics , Histone Deacetylase 1/genetics , Leukemia, Myeloid, Acute/genetics , Animals , Apoptosis/genetics , COS Cells , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Chlorocebus aethiops , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL
14.
Sci Rep ; 9(1): 1729, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30742053

ABSTRACT

Acute myeloid leukemia (AML) is often characterized by the presence of specific, recurrent chromosomal abnormalities. One of the most common aberrations, inversion of chromosome 16 [inv(16)], generates the fusion oncogene CBFB-MYH11. Previously, we used a mouse knock-in model to show that Cbfb-MYH11 induces changes in gene expression and results in the accumulation of abnormal myeloid cells, a subset of which are enriched for leukemia stem cell (LSC) activity. One gene upregulated by Cbfb-MYH11 encodes the cytokine receptor IL1RL1 (ST2). IL1RL1 and its ligand IL-33 are known regulators of mature myeloid cells, but their roles in AML are not known. Here, we use Cbfb-MYH11 knock-in mice to show that IL1RL1 is expressed by cell populations with high LSC activity, and that the cell surface expression of IL1RL1 is dynamic, implying that the expression of IL1RL1 is not restricted to a specific stage of differentiation. We also show that treatment with IL-33 increased serial replating ability and expression of pro-survival proteins in vitro. Finally, we show that IL1RL1+ cells can survive chemotherapy better than IL1RL1- cells in vivo. Collectively, our results indicate that IL1RL1 is dynamically expressed in Cbfb-MYH11+ leukemia cells and promotes their survival.


Subject(s)
Gene Expression Regulation, Leukemic , Interleukin-1 Receptor-Like 1 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Signal Transduction/drug effects
15.
Blood ; 130(22): 2431-2442, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29018080

ABSTRACT

Inversion of chromosome 16 is a consistent finding in patients with acute myeloid leukemia subtype M4 with eosinophilia, which generates a CBFB-MYH11 fusion gene. Previous studies showed that the interaction between CBFß-smooth muscle myosin heavy chain (SMMHC; encoded by CBFB-MYH11) and RUNX1 plays a critical role in the pathogenesis of this leukemia. Recently, it was shown that chromodomain helicase DNA-binding protein-7 (CHD7) interacts with RUNX1 and suppresses RUNX1-induced expansion of hematopoietic stem and progenitor cells. These results suggest that CHD7 is also critical for CBFB-MYH11-induced leukemogenesis. To test this hypothesis, we generated Chd7f/fMx1-CreCbfb+/56M mice, which expressed the Cbfb-MYH11 fusion gene and deactivated Chd7 in hematopoietic cells upon inducing Cre with polyinosinic-polycytidylic acid. The Lin-Sca1-c-Kit+ (LK) population was significantly lower in Chd7f/fMx1-CreCbfb+/56M mice than in Mx1-CreCbfb+/56M mice. In addition, there were fewer 5-bromo-2'-deoxyuridine-positive cells in the LK population in Chd7f/fMx1-CreCbfb+/56M mice, and genes associated with cell cycle, cell growth, and proliferation were differentially expressed between Chd7f/fMx1-CreCbfb+/56M and Mx1-CreCbfb+/56M leukemic cells. In vitro studies showed that CHD7 interacted with CBFß-SMMHC through RUNX1 and that CHD7 enhanced transcriptional activity of RUNX1 and CBFß-SMMHC on Csf1r, a RUNX1 target gene. Moreover, RNA sequencing of c-Kit+ cells showed that CHD7 functions mostly through altering the expression of RUNX1 target genes. Most importantly, Chd7 deficiency delayed Cbfb-MYH11-induced leukemia in both primary and transplanted mice. These data indicate that Chd7 is important for Cbfb-MYH11-induced leukemogenesis by facilitating RUNX1 regulation of transcription and cellular proliferation.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia/genetics , Oncogene Proteins, Fusion/genetics , Animals , Cell Cycle , Cell Proliferation , Gene Deletion , Humans , Leukemia/pathology , Mice , Mice, Knockout
16.
BMC Vet Res ; 13(1): 295, 2017 Oct 04.
Article in English | MEDLINE | ID: mdl-28978314

ABSTRACT

BACKGROUND: Randomised controlled trials (RCTs) are a key component of the veterinary evidence base. Sample sizes and defined outcome measures are crucial components of RCTs. To describe the sample size and number of outcome measures of veterinary RCTs either funded by the pharmaceutical industry or not, published in 2011. METHODS: A structured search of PubMed identified RCTs examining the efficacy of pharmaceutical interventions. Number of outcome measures, number of animals enrolled per trial, whether a primary outcome was identified, and the presence of a sample size calculation were extracted from the RCTs. The source of funding was identified for each trial and groups compared on the above parameters. RESULTS: Literature searches returned 972 papers; 86 papers comprising 126 individual trials were analysed. The median number of outcomes per trial was 5.0; there were no significant differences across funding groups (p = 0.133). The median number of animals enrolled per trial was 30.0; this was similar across funding groups (p = 0.302). A primary outcome was identified in 40.5% of trials and was significantly more likely to be stated in trials funded by a pharmaceutical company. A very low percentage of trials reported a sample size calculation (14.3%). CONCLUSIONS: Failure to report primary outcomes, justify sample sizes and the reporting of multiple outcome measures was a common feature in all of the clinical trials examined in this study. It is possible some of these factors may be affected by the source of funding of the studies, but the influence of funding needs to be explored with a larger number of trials. Some veterinary RCTs provide a weak evidence base and targeted strategies are required to improve the quality of veterinary RCTs to ensure there is reliable evidence on which to base clinical decisions.


Subject(s)
Capital Financing , Drug Therapy/veterinary , Outcome Assessment, Health Care , Randomized Controlled Trials as Topic/veterinary , Sample Size , Animals , Cross-Sectional Studies , Drug Therapy/economics , Randomized Controlled Trials as Topic/economics
17.
BMC Vet Res ; 13(1): 234, 2017 Aug 14.
Article in English | MEDLINE | ID: mdl-28807033

ABSTRACT

BACKGROUND: Randomised controlled trials (RCTs) are considered the gold standard form of evidence for assessing treatment efficacy, but many factors can influence their reliability including methodological quality, reporting quality and funding source. The aim of this study was to examine the relationship between funding source and positive outcome reporting in veterinary RCTs published in 2011 and to assess the risk of bias in the RCTs identified. METHODS: A structured search of PubMed was used to identify feline, canine, equine, bovine and ovine clinical trials examining the efficacy of pharmaceutical interventions published in 2011. Funding source and outcomes were extracted from each RCT and an assessment of risk of bias made using the Cochrane risk of bias tool. RESULTS: Literature searches returned 972 papers, with 86 papers (comprising 126 individual RCTs) included in the analysis. There was found to be a significantly higher proportion of positive outcomes reported in the pharmaceutical funding group (P) compared to the non-pharmaceutical (NP) and 'no funding source stated' (NF) groups (P = 56.9%, NP = 34.9%, NF = 29.1%, p < 0.05). A high proportion of trials had an unclear risk of bias across the five criteria examined. CONCLUSIONS: We found evidence that veterinary RCTs were more likely to report positive outcomes if they have pharmaceutical industry funding or involvement. Consistently poor reporting of trials, including non-identification of funding source, was found which hinders the use of the available evidence.


Subject(s)
Bias , Drug Therapy/veterinary , Randomized Controlled Trials as Topic/economics , Randomized Controlled Trials as Topic/veterinary , Treatment Outcome , Animals , Cats , Cattle , Conflict of Interest , Dogs , Drug Industry/economics , Horses , Research Design , Sheep
18.
Adv Exp Med Biol ; 962: 265-282, 2017.
Article in English | MEDLINE | ID: mdl-28299663

ABSTRACT

Mutations in RUNX1 and CBFB have long been recognized as important in hematological malignancies. Point mutations and deletions of RUNX1 are frequently found in myelodysplastic syndrome, myeloproliferative disease, and acute myeloid leukemia. Germline mutations in RUNX1 are associated with familial platelet disorder with predisposition to AML. In addition, as will be discussed in other chapters, both RUNX1 and CBFB are involved in recurrent chromosomal rearrangements in leukemia. More recently, roles for the non-mutated RUNX1 and CBFB genes have been identified in multiple leukemia subtypes. This chapter will discuss the roles of RUNX1 and CBFB, both in diseases caused by their mutations or deletions, as well as in the context of chromosomal rearrangements.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Alleles , Animals , Chromosome Aberrations , Humans
19.
Oncotarget ; 7(40): 66255-66266, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27542261

ABSTRACT

Inversion of chromosome 16 (inv(16)) generates the CBFß-SMMHC fusion protein and is found in nearly all patients with acute myeloid leukemia subtype M4 with Eosinophilia (M4Eo). Expression of CBFß-SMMHC is causative for leukemia development, but the molecular mechanisms underlying its activity are unclear. Recently, there have been important advances in defining the role of CBFß-SMMHC and its binding partners, the transcription factor RUNX1 and the histone deacetylase HDAC8. Importantly, initial trials demonstrate that small molecules targeting these binding partners are effective against CBFß-SMMHC induced leukemia. This review will discuss recent advances in defining the mechanism of CBFß-SMMHC activity, as well as efforts to develop new therapies for inv(16) AML.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 16 , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Protein Interaction Maps/drug effects , Repressor Proteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 2 Subunit/metabolism , Histone Deacetylases/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Binding , Repressor Proteins/metabolism
20.
Leukemia ; 29(8): 1771-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25742748

ABSTRACT

CBFß-SMMHC (core-binding factor ß-smooth muscle myosin heavy chain), the fusion protein generated by the chromosome 16 inversion fusion gene, CBFB-MYH11, is known to initiate leukemogenesis. However, the mechanism through which CBFß-SMMHC contributes to leukemia development is not well understood. Previously, it was proposed that CBFß-SMMHC acts by dominantly repressing the transcription factor RUNX1 (Runt-related protein 1), but we recently showed that CBFß-SMMHC has activities that are independent of RUNX1 repression. In addition, we showed that a modified CBFß-SMMHC with decreased RUNX1-binding activity accelerates leukemogenesis. These results raise questions about the importance of RUNX1 in leukemogenesis by CBFß-SMMHC. To test this, we generated mice expressing Cbfb-MYH11 in a Runx1-deficient background, resulting from either homozygous Runx1-null alleles (Runx1(-/-)) or a single dominant-negative Runx1 allele (Runx1(+/lz)). We found that loss of Runx1 activity rescued the differentiation defects induced by Cbfb-MYH11 during primitive hematopoiesis. During definitive hematopoiesis, RUNX1 loss also significantly reduced the proliferation and differentiation defects induced by Cbfb-MYH11. Importantly, Cbfb-MYH11-induced leukemia had much longer latency in Runx1(+/lz) mice than in Runx1-sufficient mice. These data indicate that Runx1 activity is critical for Cbfb-MYH11-induced hematopoietic defects and leukemogenesis.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Hematopoiesis/physiology , Leukemia/metabolism , Leukemia/pathology , Myosin Heavy Chains/physiology , Animals , Apoptosis , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Flow Cytometry , Gene Knock-In Techniques , Humans , Immunoprecipitation , Leukemia/mortality , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...