Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Heart Rhythm ; 20(2): 190-197, 2023 02.
Article in English | MEDLINE | ID: mdl-36272710

ABSTRACT

BACKGROUND: Antitachycardia pacing (ATP) is an established implantable cardioverter-defibrillator (ICD) therapy that terminates ventricular tachycardias (VTs) without painful ICD shocks. However, factors influencing ATP success are not well understood. OBJECTIVE: The purpose of this study was to examine ATP success rates by patient, device, and programming characteristics. METHODS: This retrospective analysis of the PainFree SmartShock Technology study included spontaneous ATP-treated monomorphic VT episodes. ATP success rates were calculated for various factors. Also, the relationship of ATP programming on shock burden and syncope were investigated. RESULTS: Of the 2770 enrolled patients (2200 [79%] male; mean age 65 years), 1699 (61%) received an ICD and 1071 (39%) a cardiac resynchronization therapy - defibrillator. ATP had >80% rate of success for terminating VTs overall, with similar rates observed between ICD and cardiac resynchronization therapy - defibrillator devices (82.2% vs 80.3%, respectively; P = .81) as well as between primary and secondary prevention patients with ICDs (77.2% vs 83.9% respectively; P = .25). Arrhythmias with a median cycle length of ≥320 ms had a significantly higher ATP success rate (88.0%; 95% confidence interval 84.8%-90.6%). The cumulative percentage of ATP success increased from 71% at 1 ATP sequence delivered to 87% at ≥8 sequences delivered. Programming more ATP sequences was associated with lower shock burden (P = .0005). There was no evidence that more sequences were associated with higher rates of syncope (P = .16). CONCLUSION: Delivering more ATP sequences resulted in a higher overall success of terminating VTs, while programming more ATP was associated with decreased shock burden and no evidence of increased syncope or acceleration. This suggests that more ATP sequences should be programmed when possible, but confirmation in prospective studies will be necessary.


Subject(s)
Cardiac Resynchronization Therapy , Defibrillators, Implantable , Tachycardia, Ventricular , Humans , Male , Aged , Female , Prospective Studies , Retrospective Studies , Syncope/therapy , Adenosine Triphosphate , Treatment Outcome
3.
Int J Cardiol ; 350: 36-40, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34998948

ABSTRACT

BACKGROUND: Patients with inherited arrhythmogenic diseases (IADs) are often prescribed preventative implantable cardioverter-defibrillators (ICDs) to manage their increased sudden cardiac arrest risk. However, it has been suggested that ICDs in IAD patients may come with additional risk. We aimed to leverage the PainFree SmartShock Technology dataset to compare inappropriate therapies, appropriate therapies, mortality, and complications in patients with and without IAD. METHODS: This retrospective analysis included extracted, physician-adjudicated, arrhythmic episodes from ICD devices. The incidence of arrhythmic events was estimated with the Kaplan-Meier method using the log-rank test. Cox proportional hazards regression was used to estimate hazard ratios (HRs) with their 95% confidence intervals (CIs). RESULTS: Of the 1699 ICD patients, 77 patients (4.5%) had IAD. Incidence of inappropriate shock was similar in both patients with (3.2% at 24 months) and without (3.8% at 24 months) IAD (HR: 0.80, CI: 0.19-3.30, p = 0.76). In a multivariable analysis IAD was not significantly associated with reduced mortality (HR: 0.64, CI: 0.08-4.80, p = 0.66). The rates of complications were numerically lower in patients with IAD vs without (8.8% vs 9.6% at 24 months respectively), but not statistically significant (HR: 0.83, CI: 0.20-3.38, p = 0.79). CONCLUSIONS: IAD patients showed a very low annual rate of inappropriate therapy. This suggests that newer algorithms, such as the SST algorithm, are equally good at identifying and treating life-threatening arrhythmias in patients regardless of whether they have IAD.


Subject(s)
Defibrillators, Implantable , Arrhythmias, Cardiac/etiology , Death, Sudden, Cardiac/epidemiology , Defibrillators, Implantable/adverse effects , Humans , Retrospective Studies , Risk Factors , Technology , Treatment Outcome
4.
Heart Rhythm ; 17(8): 1393-1397, 2020 08.
Article in English | MEDLINE | ID: mdl-32247833

ABSTRACT

Pacemaker-related infections remain a constant concern due to increased risk of patient morbidity and mortality. Although transvenous pacemakers are expected to have an infection rate ranging from 0.77% to 2.08%, no cases of leadless pacemaker infection have been reported in clinical trials enrolling more than 3000 patients. Many potential reasons why leadless pacemakers may be resistant to infection include the absence of a subcutaneous pocket and leads, reduced skin and glove contact, size, location, and device material. This review summarizes the current state of evidence regarding the apparent infection resistance of leadless pacemakers.


Subject(s)
Arrhythmias, Cardiac/therapy , Cardiac Pacing, Artificial/methods , Heart Conduction System/physiopathology , Pacemaker, Artificial , Arrhythmias, Cardiac/physiopathology , Equipment Design , Humans
5.
Nat Commun ; 11(1): 790, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034147

ABSTRACT

APOBEC3B, an anti-viral cytidine deaminase which induces DNA mutations, has been implicated as a mediator of cancer evolution and therapeutic resistance. Mutational plasticity also drives generation of neoepitopes, which prime anti-tumor T cells. Here, we show that overexpression of APOBEC3B in tumors increases resistance to chemotherapy, but simultaneously heightens sensitivity to immune checkpoint blockade in a murine model of melanoma. However, in the vaccine setting, APOBEC3B-mediated mutations reproducibly generate heteroclitic neoepitopes in vaccine cells which activate de novo T cell responses. These cross react against parental, unmodified tumors and lead to a high rate of cures in both subcutaneous and intra-cranial tumor models. Heteroclitic Epitope Activated Therapy (HEAT) dispenses with the need to identify patient specific neoepitopes and tumor reactive T cells ex vivo. Thus, actively driving a high mutational load in tumor cell vaccines increases their immunogenicity to drive anti-tumor therapy in combination with immune checkpoint blockade.


Subject(s)
Cancer Vaccines/pharmacology , Cytidine Deaminase/immunology , Immunotherapy/methods , Minor Histocompatibility Antigens/immunology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Drug Resistance, Neoplasm , Epitopes/immunology , Female , Humans , Killer Cells, Natural/immunology , Melanoma/immunology , Melanoma/therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mutation , Tumor Escape/drug effects
6.
DNA Repair (Amst) ; 85: 102738, 2020 01.
Article in English | MEDLINE | ID: mdl-31731258

ABSTRACT

The repair of DNA double-stranded breaks (DSBs) is an essential function performed by the Classical Non-Homologous End-Joining (C-NHEJ) pathway in higher eukaryotes. C-NHEJ, in fact, does double duty as it is also required for the repair of the intermediates formed during lymphoid B- and T-cell recombination. Consequently, the failure to properly repair DSBs leads to both genomic instability and immunodeficiency. A critical DSB protein required for C-NHEJ is the DNA Ligase IV (LIGIV) accessory factor, X-Ray Cross Complementing 4 (XRCC4). XRCC4 is believed to stabilize LIGIV, participate in LIGIV activation, and to help tether the broken DSB ends together. XRCC4's role in these processes has been muddied by the identification of two additional XRCC4 paralogs, XRCC4-Like Factor (XLF), and Paralog of XRCC4 and XLF (PAXX). The roles that these paralogs play in C-NHEJ is partially understood, but, in turn, has itself been obscured by species-specific differences observed in the absence of one or the other paralogs. In order to investigate the role(s) that XRCC4 may play, with or without XLF and/or PAXX, in lymphoid variable(diversity)joining [V(D)J] recombination as well as in DNA DSB repair in human somatic cells, we utilized gene targeting to inactivate the XRCC4 gene in both parental and XLF- HCT116 cells and then inactivated PAXX in those same cell lines. The loss of XRCC4 expression by itself led, as anticipated, to increased sensitivity to DNA damaging agents as well as an increased dependence on microhomology-mediated DNA repair whether in the context of DSB repair or during V(D)J recombination. The additional loss of XLF in these cell lines sensitized the cells even more whereas the presence or absence of PAXX was scarcely negligible. These studies demonstrate that, of the three LIG4 accessory factor paralogs, the absence of XRCC4 influences DNA repair and recombination the most in human cells.


Subject(s)
DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Etoposide/adverse effects , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Ligase ATP/metabolism , Gene Targeting , HCT116 Cells , Humans , V(D)J Recombination
7.
Antibodies (Basel) ; 8(3)2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31544853

ABSTRACT

The DNA cytosine deaminase APOBEC3B (A3B) is normally an antiviral factor in the innate immune response. However, A3B has been implicated in cancer mutagenesis, particularly in solid tumors of the bladder, breast, cervix, head/neck, and lung. Here, we report data on the generation and characterization of a rabbit monoclonal antibody (mAb) for human A3B. One mAb, 5210-87-13, demonstrates utility in multiple applications, including ELISA, immunoblot, immunofluorescence microscopy, and immunohistochemistry. In head-to-head tests with commercial reagents, 5210-87-13 was the only rabbit monoclonal suitable for detecting native A3B and for immunohistochemical quantification of A3B in tumor tissues. This novel mAb has the potential to enable a wide range of fundamental and clinical studies on A3B in human biology and disease.

8.
J Gen Virol ; 100(7): 1140-1152, 2019 07.
Article in English | MEDLINE | ID: mdl-31145054

ABSTRACT

HIV-1 replication in CD4-positive T lymphocytes requires counteraction of multiple different innate antiviral mechanisms. Macrophage cells are also thought to provide a reservoir for HIV-1 replication but less is known in this cell type about virus restriction and counteraction mechanisms. Many studies have combined to demonstrate roles for APOBEC3D, APOBEC3F, APOBEC3G and APOBEC3H in HIV-1 restriction and mutation in CD4-positive T lymphocytes, whereas the APOBEC enzymes involved in HIV-1 restriction in macrophages have yet to be delineated fully. We show that multiple APOBEC3 genes including APOBEC3G are expressed in myeloid cell lines such as THP-1. Vif-deficient HIV-1 produced from THP-1 is less infectious than Vif-proficient virus, and proviral DNA resulting from such Vif-deficient infections shows strong G to A mutation biases in the dinucleotide motif preferred by APOBEC3G. Moreover, Vif mutant viruses with selective sensitivity to APOBEC3G show Vif null-like infectivity levels and similarly strong APOBEC3G-biased mutation spectra. Importantly, APOBEC3G-null THP-1 cells yield Vif-deficient particles with significantly improved infectivities and proviral DNA with background levels of G to A hypermutation. These studies combine to indicate that APOBEC3G is the main HIV-1 restricting APOBEC3 family member in THP-1 cells.


Subject(s)
APOBEC-3G Deaminase/metabolism , HIV Infections/enzymology , HIV-1/physiology , APOBEC-3G Deaminase/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Mutation , Myeloid Cells , THP-1 Cells , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics , vif Gene Products, Human Immunodeficiency Virus/metabolism
9.
Cancer Immunol Res ; 7(5): 828-840, 2019 05.
Article in English | MEDLINE | ID: mdl-30940643

ABSTRACT

Antitumor T-cell responses raised by first-line therapies such as chemotherapy, radiation, tumor cell vaccines, and viroimmunotherapy tend to be weak, both quantitatively (low frequency) and qualitatively (low affinity). We show here that T cells that recognize tumor-associated antigens can directly kill tumor cells if used at high effector-to-target ratios. However, when these tumor-reactive T cells were present at suboptimal ratios, direct T-cell-mediated tumor cell killing was reduced and the ability of tumor cells to evolve away from a coapplied therapy (oncolytic or suicide gene therapy) was promoted. This T-cell-mediated increase in therapeutic resistance was associated with C to T transition mutations that are characteristic of APOBEC3 cytosine deaminase activity and was induced through a TNFα and protein kinase C-dependent pathway. Short hairpin RNA inhibition of endogenous APOBEC3 reduced rates of tumor escape from oncolytic virus or suicide gene therapy to those seen in the absence of antitumor T-cell coculture. Conversely, overexpression of human APOBEC3B in tumor cells enhanced escape from suicide gene therapy and oncolytic virus therapy both in vitro and in vivo Our data suggest that weak affinity or low frequency T-cell responses against tumor antigens may contribute to the ability of tumor cells to evolve away from first-line therapies. We conclude that immunotherapies need to be optimized as early as possible so that, if they do not kill the tumor completely, they do not promote treatment resistance.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Melanoma, Experimental/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line, Tumor , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Female , Ganciclovir/therapeutic use , Mammalian orthoreovirus 3 , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Mutation , Oncolytic Virotherapy , Tumor Escape
10.
Cancer Res ; 79(5): 905-917, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30674530

ABSTRACT

Medulloblastoma and central nervous system primitive neuroectodermal tumors (CNS-PNET) are aggressive, poorly differentiated brain tumors with limited effective therapies. Using Sleeping Beauty (SB) transposon mutagenesis, we identified novel genetic drivers of medulloblastoma and CNS-PNET. Cross-species gene expression analyses classified SB-driven tumors into distinct medulloblastoma and CNS-PNET subgroups, indicating they resemble human Sonic hedgehog and group 3 and 4 medulloblastoma and CNS neuroblastoma with FOXR2 activation. This represents the first genetically induced mouse model of CNS-PNET and a rare model of group 3 and 4 medulloblastoma. We identified several putative proto-oncogenes including Arhgap36, Megf10, and Foxr2. Genetic manipulation of these genes demonstrated a robust impact on tumorigenesis in vitro and in vivo. We also determined that FOXR2 interacts with N-MYC, increases C-MYC protein stability, and activates FAK/SRC signaling. Altogether, our study identified several promising therapeutic targets in medulloblastoma and CNS-PNET. SIGNIFICANCE: A transposon-induced mouse model identifies several novel genetic drivers and potential therapeutic targets in medulloblastoma and CNS-PNET.


Subject(s)
Brain Neoplasms/genetics , Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Neuroectodermal Tumors, Primitive/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , DNA Transposable Elements/genetics , Female , Forkhead Transcription Factors/genetics , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/genetics , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Membrane Proteins/genetics , Mice , Mice, Nude , Mutagenesis, Insertional/methods , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neuroectodermal Tumors, Primitive/metabolism , Neuroectodermal Tumors, Primitive/pathology , Prognosis
11.
Mol Ther Oncolytics ; 11: 1-13, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30294666

ABSTRACT

Tumor cells frequently evade applied therapies through the accumulation of genomic mutations and rapid evolution. In the case of oncolytic virotherapy, understanding the mechanisms by which cancer cells develop resistance to infection and lysis is critical to the development of more effective viral-based platforms. Here, we identify APOBEC3 as an important factor that restricts the potency of oncolytic vesicular stomatitis virus (VSV). We show that VSV infection of B16 murine melanoma cells upregulated APOBEC3 in an IFN-ß-dependent manner, which was responsible for the evolution of virus-resistant cell populations and suggested that APOBEC3 expression promoted the acquisition of a virus-resistant phenotype. Knockdown of APOBEC3 in B16 cells diminished their capacity to develop resistance to VSV infection in vitro and enhanced the therapeutic effect of VSV in vivo. Similarly, overexpression of human APOBEC3B promoted the acquisition of resistance to oncolytic VSV both in vitro and in vivo. Finally, we demonstrate that APOBEC3B expression had a direct effect on the fitness of VSV, an RNA virus that has not previously been identified as restricted by APOBEC3B. This research identifies APOBEC3 enzymes as key players to target in order to improve the efficacy of viral or broader nucleic acid-based therapeutic platforms.

12.
Virology ; 511: 74-81, 2017 11.
Article in English | MEDLINE | ID: mdl-28841445

ABSTRACT

The APOBEC3 DNA cytosine deaminase family comprises a fundamental arm of the innate immune response and is best known for retrovirus restriction. Several APOBEC3 enzymes restrict HIV-1 and related retroviruses by deaminating viral cDNA cytosines to uracils compromising viral genomes. Human APOBEC3B (A3B) shows strong virus restriction activities in a variety of experimental systems, and is subjected to tight post-translational regulation evidenced by cell-specific HIV-1 restriction activity and active nuclear import. Here we ask whether lysines and/or lysine post-translational modifications are required for these A3B activities. A lysine-free derivative of human A3B was constructed and shown to be indistinguishable from the wild-type enzyme in DNA cytosine deamination, HIV-1 restriction, and nuclear localization activities. However, lysine loss did render the protein resistant to degradation by SIV Vif. Taken together, we conclude that lysine side chains and modifications thereof are unlikely to be central to A3B function or regulation in human cells.


Subject(s)
Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Mutational Analysis , Lysine/genetics , Lysine/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Active Transport, Cell Nucleus , Cytosine/metabolism , Deamination , HEK293 Cells , HIV-1/immunology , Humans , Protein Processing, Post-Translational
13.
Sci Adv ; 2(10): e1601737, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27730215

ABSTRACT

Breast tumors often display extreme genetic heterogeneity characterized by hundreds of gross chromosomal aberrations and tens of thousands of somatic mutations. Tumor evolution is thought to be ongoing and driven by multiple mutagenic processes. A major outstanding question is whether primary tumors have preexisting mutations for therapy resistance or whether additional DNA damage and mutagenesis are necessary. Drug resistance is a key measure of tumor evolvability. If a resistance mutation preexists at the time of primary tumor presentation, then the intended therapy is likely to fail. However, if resistance does not preexist, then ongoing mutational processes still have the potential to undermine therapeutic efficacy. The antiviral enzyme APOBEC3B (apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3B) preferentially deaminates DNA C-to-U, which results in signature C-to-T and C-to-G mutations commonly observed in breast tumors. We use clinical data and xenograft experiments to ask whether APOBEC3B contributes to ongoing breast tumor evolution and resistance to the selective estrogen receptor modulator, tamoxifen. First, APOBEC3B levels in primary estrogen receptor-positive (ER+) breast tumors inversely correlate with the clinical benefit of tamoxifen in the treatment of metastatic ER+ disease. Second, APOBEC3B depletion in an ER+ breast cancer cell line results in prolonged tamoxifen responses in murine xenograft experiments. Third, APOBEC3B overexpression accelerates the development of tamoxifen resistance in murine xenograft experiments by a mechanism that requires the enzyme's catalytic activity. These studies combine to indicate that APOBEC3B promotes drug resistance in breast cancer and that inhibiting APOBEC3B-dependent tumor evolvability may be an effective strategy to improve efficacies of targeted cancer therapies.


Subject(s)
Breast Neoplasms/enzymology , Cytidine Deaminase/metabolism , Drug Resistance, Neoplasm , Minor Histocompatibility Antigens/metabolism , Tamoxifen/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cytidine Deaminase/genetics , Female , Gene Deletion , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Nude , Minor Histocompatibility Antigens/genetics , Neoplasm Transplantation
14.
Cancer Res ; 75(21): 4538-47, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26420215

ABSTRACT

Overexpression of the antiviral DNA cytosine deaminase APOBEC3B has been linked to somatic mutagenesis in many cancers. Human papillomavirus infection accounts for APOBEC3B upregulation in cervical and head/neck cancers, but the mechanisms underlying nonviral malignancies are unclear. In this study, we investigated the signal transduction pathways responsible for APOBEC3B upregulation. Activation of protein kinase C (PKC) by the diacylglycerol mimic phorbol-myristic acid resulted in specific and dose-responsive increases in APOBEC3B expression and activity, which could then be strongly suppressed by PKC or NF-κB inhibition. PKC activation caused the recruitment of RELB, but not RELA, to the APOBEC3B promoter, implicating noncanonical NF-κB signaling. Notably, PKC was required for APOBEC3B upregulation in cancer cell lines derived from multiple tumor types. By revealing how APOBEC3B is upregulated in many cancers, our findings suggest that PKC and NF-κB inhibitors may be repositioned to suppress cancer mutagenesis, dampen tumor evolution, and decrease the probability of adverse outcomes, such as drug resistance and metastasis.


Subject(s)
Cytidine Deaminase/biosynthesis , Neoplasms/metabolism , Protein Kinase C/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism , Cell Line, Tumor , Cytidine Deaminase/genetics , Humans , Minor Histocompatibility Antigens , NF-kappa B p50 Subunit/biosynthesis , NF-kappa B p52 Subunit/biosynthesis , Neoplasms/genetics , Papillomavirus Infections/pathology , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Signal Transduction , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelB/antagonists & inhibitors , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...