Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.233
Filter
2.
Biol Pharm Bull ; 47(5): 941-945, 2024.
Article in English | MEDLINE | ID: mdl-38735754

ABSTRACT

Hepatitis B virus reactivation (HBV-R) is a serious complication that can occur in patients with resolved HBV infection during cancer chemotherapy. We examined the levels of HBV surface antibody (HBsAb) and HBV core antibody (HBcAb) to assess the incidence of HBV-R in cancer patients including hematopoietic stem cell transplantation (HSCT) and rituximab administration. This retrospective cohort study included 590 patients with resolved HBV infection. The incidence of HBV-R was evaluated 761.5 (range, 90-3898) days after the inititiation of chemotherapy. Of the patients, 13 (2.2%) developed HBV-R after the start of chemotherapy. All 13 patients exhibited lower HBsAb (<100 mIU/mL) levels at baseline. A higher level of HBcAb (≥100 cut off index (C.O.I.)) was a possible risk factor for HBV-R as well as HSCT and rituximab administration. The simultaneous presence of HBsAb <100 mIU/mL and HBcAb ≥100 C.O.I. increased the risk of HBV-R by 18.5%. Patients treated with rituximab were at a higher risk of HBV-R (18.4%) despite having HBcAb <100 C.O.I. Our results suggest that assessment of HBsAb and HBcAb levels prior to the chemotherapy is important for identifying patients at high risk of HBV-R, especially in solid cancers without HSCT and rituximab administration.


Subject(s)
Hepatitis B Antibodies , Hepatitis B Surface Antigens , Hepatitis B virus , Hepatitis B , Rituximab , Virus Activation , Humans , Male , Female , Middle Aged , Retrospective Studies , Hepatitis B Antibodies/blood , Hepatitis B Antibodies/immunology , Hepatitis B virus/immunology , Hepatitis B virus/physiology , Virus Activation/drug effects , Rituximab/therapeutic use , Rituximab/adverse effects , Adult , Aged , Hepatitis B/immunology , Hepatitis B Surface Antigens/blood , Hepatitis B Surface Antigens/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Young Adult , Neoplasms/drug therapy , Neoplasms/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/blood , Aged, 80 and over , Adolescent
3.
Nat Commun ; 15(1): 3729, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702330

ABSTRACT

The unique virus-cell interaction in Epstein-Barr virus (EBV)-associated malignancies implies targeting the viral latent-lytic switch is a promising therapeutic strategy. However, the lack of specific and efficient therapeutic agents to induce lytic cycle in these cancers is a major challenge facing clinical implementation. We develop a synthetic transcriptional activator that specifically activates endogenous BZLF1 and efficiently induces lytic reactivation in EBV-positive cancer cells. A lipid nanoparticle encapsulating nucleoside-modified mRNA which encodes a BZLF1-specific transcriptional activator (mTZ3-LNP) is synthesized for EBV-targeted therapy. Compared with conventional chemical inducers, mTZ3-LNP more efficiently activates EBV lytic gene expression in EBV-associated epithelial cancers. Here we show the potency and safety of treatment with mTZ3-LNP to suppress tumor growth in EBV-positive cancer models. The combination of mTZ3-LNP and ganciclovir yields highly selective cytotoxic effects of mRNA-based lytic induction therapy against EBV-positive tumor cells, indicating the potential of mRNA nanomedicine in the treatment of EBV-associated epithelial cancers.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Liposomes , Nanoparticles , Trans-Activators , Humans , Herpesvirus 4, Human/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/drug therapy , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virus Activation/drug effects , Xenograft Model Antitumor Assays , Gene Expression Regulation, Viral/drug effects , Mice, Nude , Female
4.
PLoS One ; 19(5): e0303995, 2024.
Article in English | MEDLINE | ID: mdl-38771836

ABSTRACT

BACKGROUNDS: In critically ill patients with COVID-19, secondary infections are potentially life-threatening complications. This study aimed to determine the prevalence, clinical characteristics, and risk factors of CMV reactivation among critically ill immunocompetent patients with COVID-19 pneumonia. METHODS: A retrospective cohort study was conducted among adult patients who were admitted to ICU and screened for quantitative real-time PCR for CMV viral load in a tertiary-care hospital during the third wave of the COVID-19 outbreak in Thailand. Cox regression models were used to identify significant risk factors for developing CMV reactivation. RESULTS: A total of 185 patients were studied; 133 patients (71.9%) in the non-CMV group and 52 patients (28.1%) in the CMV group. Of all, the mean age was 64.7±13.3 years and 101 patients (54.6%) were males. The CMV group had received a significantly higher median cumulative dose of corticosteroids than the non-CMV group (301 vs 177 mg of dexamethasone, p<0.001). Other modalities of treatments for COVID-19 including anti-viral drugs, anti-cytokine drugs and hemoperfusion were not different between the two groups (p>0.05). The 90-day mortality rate for all patients was 29.1%, with a significant difference between the CMV group and the non-CMV group (42.3% vs. 24.1%, p = 0.014). Median length of stay was longer in the CMV group than non-CMV group (43 vs 24 days, p<0.001). The CMV group has detectable CMV DNA load with a median [IQR] of 4,977 [1,365-14,742] IU/mL and 24,570 [3,703-106,642] in plasma and bronchoalveolar fluid, respectively. In multivariate analysis, only a cumulative corticosteroids dose of dexamethasone ≥250 mg (HR = 2.042; 95%CI, 1.130-3.688; p = 0.018) was associated with developing CMV reactivation. CONCLUSION: In critically ill COVID-19 patients, CMV reactivation is frequent and a high cumulative corticosteroids dose is a significant risk factor for CMV reactivation, which is associated with poor outcomes. Further prospective studies are warranted to determine optimal management.


Subject(s)
COVID-19 , Critical Illness , Cytomegalovirus Infections , Cytomegalovirus , Humans , Male , Middle Aged , COVID-19/epidemiology , COVID-19/virology , COVID-19/complications , Female , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/complications , Risk Factors , Aged , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/isolation & purification , Retrospective Studies , Prevalence , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Virus Activation/drug effects , Thailand/epidemiology , Viral Load
5.
J Int Med Res ; 52(5): 3000605241252580, 2024 May.
Article in English | MEDLINE | ID: mdl-38760056

ABSTRACT

Recombinant human type II tumour necrosis factor receptor-antibody fusion protein (rh TNFR:Fc) is an immunosuppressant approved for treating rheumatoid arthritis (RA). This case report describes a case of hepatitis B reactivation in a patient with drug-induced acute-on-chronic liver failure. A 58-year-old woman with a history of RA was treated with rh TNFR:Fc; and then subsequently received 25 mg rh TNFR:Fc, twice a week, as maintenance therapy. No anti-hepatitis B virus (HBV) preventive treatment was administered. Six months later, she was hospitalized with acute jaundice. HBV reactivation was observed, leading to acute-on-chronic liver failure. After active treatment, the patient's condition improved and she recovered well. Following careful diagnosis and treatment protocols are essential when treating RA with rh TNFR:Fc, especially in anti-hepatitis B core antigen antibody-positive patients, even when the HBV surface antigen and the HBV DNA are negative. In the case of HBV reactivation, liver function parameters, HBV surface antigen and HBV DNA should be closely monitored during treatment, and antiviral drugs should be used prophylactically when necessary, as fatal hepatitis B reactivation may occur in rare cases. A comprehensive evaluation and medication should be administered in a timely manner after evaluating the patient's physical condition and closely monitoring the patient.


Subject(s)
Hepatitis B virus , Hepatitis B , Recombinant Fusion Proteins , Virus Activation , Humans , Female , Middle Aged , Hepatitis B virus/pathogenicity , Hepatitis B virus/physiology , Virus Activation/drug effects , Recombinant Fusion Proteins/therapeutic use , Hepatitis B/virology , Hepatitis B/drug therapy , Hepatitis B/complications , Hepatitis B/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/virology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/complications , Liver Failure/virology , Liver Failure/etiology
6.
Rev Med Virol ; 34(3): e2538, 2024 May.
Article in English | MEDLINE | ID: mdl-38658176

ABSTRACT

Serious adverse events following vaccination include medical complications that require hospitalisation. The live varicella vaccine that was approved by the Food and Drug Administration in the United States in 1995 has an excellent safety record. Since the vaccine is a live virus, adverse events are more common in immunocompromised children who are vaccinated inadvertently. This review includes only serious adverse events in children considered to be immunocompetent. The serious adverse event called varicella vaccine meningitis was first reported in a hospitalised immunocompetent child in 2008. When we carried out a literature search, we found 15 cases of immunocompetent children and adolescents with varicella vaccine meningitis; the median age was 11 years. Eight of the children had received two varicella vaccinations. Most of the children also had a concomitant herpes zoster rash, although three did not. The children lived in the United States, Greece, Germany, Switzerland, and Japan. During our literature search, we found five additional cases of serious neurological events in immunocompetent children; these included 4 cases of progressive herpes zoster and one case of acute retinitis. Pulses of enteral corticosteroids as well as a lack of herpes simplex virus antibody may be risk factors for reactivation in immunocompetent children. All 20 children with adverse events were treated with acyclovir and recovered; 19 were hospitalised and one child was managed as an outpatient. Even though the number of neurological adverse events remains exceedingly low following varicella vaccination, we recommend documentation of those caused by the vaccine virus.


Subject(s)
Chickenpox Vaccine , Meningitis, Viral , Adolescent , Child , Child, Preschool , Female , Humans , Male , Acyclovir/therapeutic use , Antiviral Agents/therapeutic use , Chickenpox/prevention & control , Chickenpox/virology , Chickenpox Vaccine/administration & dosage , Chickenpox Vaccine/adverse effects , Chickenpox Vaccine/immunology , Herpesvirus 3, Human/immunology , Meningitis, Viral/virology , Nervous System Diseases/virology , Nervous System Diseases/etiology , Vaccination/adverse effects , Virus Activation/drug effects
7.
Transplant Cell Ther ; 30(5): 538.e1-538.e10, 2024 May.
Article in English | MEDLINE | ID: mdl-38331195

ABSTRACT

Cytomegalovirus (CMV) reactivations cause significant morbidity in allogeneic hematopoietic stem cell transplantation (HSCT) recipients. Graft-versus-host disease (GVHD) prophylaxis with post-transplantation cyclophosphamide (PTCy) is associated with an increased risk of CMV infections. Data are limited comparing HSCT with PTCy performed from matched sibling donors (MSDs), matched unrelated donors (MUDs), and haploidentical (Haplo) donors. In the present study, we aimed to characterize CMV reactivation and recurrence in patients with hematologic malignancies undergoing HSCT from MSD, MUD, and Haplo donors using PTCy as GVHD prophylaxis in the pre-letermovir era. We also analyzed risk factors of CMV reactivation, including GVHD as a time-dependent variable, on the incidence and mortality associated with CMV infections. We analyzed CMV reactivation in patients undergoing HSCT from 160 MSDs, 124 MUDs, and 82 Haplo donors from a single institution. Uniform GVHD prophylaxis with PTCy, sirolimus, and mycophenolate mofetil was given irrespective of donor type. Overall, 46% of patients had at least 1 CMV reactivation. The 1-year cumulative incidence of CMV infection was 39% for MSD, 44% for MUD, and 62% for Haplo donors (P < .001), with 96% of reactivations occurring before day +100. Multivariate analysis identified factors associated with the first CMV reactivation, including Haplo donor, positive recipient CMV serology, older patient age, and grade II-IV acute GVHD. The 1-year cumulative incidence of second reactivation from HSCT was 13%. Recipient CMV seropositivity, older patient age, and grade II-IV acute GVHD, but not type of donor, were identified as adverse factors for second CMV reactivation in multivariate analysis. The 1-year cumulative incidence of a third reactivation post HSCT was 4.4%. Ten cases of CMV disease were recorded, with no attributable deaths. Nevertheless, the risk for nonrelapse mortality was greater for patients who experienced CMV reactivation in multivariate time-dependent Cox model analysis. CMV reactivation is frequent in HSCT with PTCy in patients not receiving letermovir prophylaxis. Identified risk factors include the use of a Haplo donor, recipient CMV seropositivity, and grade II-IV acute GVHD. The prevalence of recurrent CMV reactivations is a noteworthy issue, especially after acute GVHD, warranting trials of secondary prophylaxis strategies.


Subject(s)
Cyclophosphamide , Cytomegalovirus Infections , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Virus Activation , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Female , Middle Aged , Adult , Virus Activation/drug effects , Cyclophosphamide/therapeutic use , Cyclophosphamide/adverse effects , Graft vs Host Disease/prevention & control , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/epidemiology , Transplantation, Homologous/adverse effects , Cytomegalovirus/immunology , Cytomegalovirus/drug effects , Aged , Young Adult , Tissue Donors , Adolescent , Transplantation, Haploidentical/adverse effects , Risk Factors , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Hematologic Neoplasms/therapy , Unrelated Donors , HLA Antigens/immunology , Siblings
8.
Antiviral Res ; 217: 105699, 2023 09.
Article in English | MEDLINE | ID: mdl-37549849

ABSTRACT

Epstein-Barr virus (EBV) is a highly prevalent human herpesvirus that persists for life in more than 95% of the adult population. EBV usually establishes an asymptomatic life-long infection, but it is also associated with malignancies affecting B lymphocytes and epithelial cells mainly. The virus alternates between a latent phase and a lytic phase, both of which contribute to the initiation of the tumor process. So far, there is only a limited number of antiviral molecules against the lytic phase, most of them targeting viral replication. Recent studies provided evidence that EBV uses components of the NLRP3 inflammasome to enter the productive phase of its cycle following activation in response to various stimuli. In the present work, we demonstrate that shikonin, a natural molecule with low toxicity which is known to inhibit inflammasome, can efficiently repress EBV reactivation. Similar results were obtained with apigenin and OLT 1177, two other NLRP3 inflammasome inhibitors. It is shown herein that shikonin repressed the transcription of reactivation-induced NLRP3 thereby inhibiting inflammasome activation and EBV lytic phase induction.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Herpesvirus 4, Human , Inflammasomes , Naphthoquinones , Virus Activation , Inflammasomes/antagonists & inhibitors , Virus Activation/drug effects , Herpesvirus 4, Human/drug effects , Naphthoquinones/pharmacology , Apigenin/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Humans , Cell Line , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Cell Line, Tumor
9.
J Agric Food Chem ; 71(10): 4394-4407, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36854107

ABSTRACT

As a continuation of our research on the development of pesticide active quinolizidine alkaloids (QAs) from the family Fabaceae, the chemical constituents of the root of Sophora tonkinensis Gagnep. were systematically investigated. Seventeen new matrine-type alkaloids (1-17), including one new naturally occurring compound (17), along with 20 known ones were isolated from the EtOH extract of S. tonkinensis. Notably, compound 5 possessed an unprecedented 6/6/5/4/6/6 hexacyclic system. Their structures were confirmed via comprehensive spectroscopic data analysis (IR, UV, NMR, HRESIMS), ECD calculation, and X-ray crystallography. Biological tests indicated that compounds 1, 4, 10, 12, 13, and 30 displayed significant anti-tomato spotted wilt virus (TSWV) activities compared with the positive control ningnanmycin. Moreover, compound 12 strongly inhibited the expression of the TSWV N, NSs, and NSm genes and TSWV NSs protein in plant host. Furthermore, compounds 4, 10, 12, 20, and 22 exhibited moderate insecticidal activities against TSWV thrip vector (Frankliniella occidentalis).


Subject(s)
Sophora , Tospovirus , Matrines/chemistry , Matrines/pharmacology , Tospovirus/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Spectrophotometry , Crystallography, X-Ray , Virus Activation/drug effects , Animals , Insecticides/chemistry , Insecticides/pharmacology , Viral Nonstructural Proteins/genetics , Models, Molecular , Molecular Structure , Plant Roots
10.
Turk J Gastroenterol ; 34(2): 156-160, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36445058

ABSTRACT

BACKGROUND: The risk of hepatitis B reactivation in hepatitis B surface antigen-negative phase of hepatitis B virus-infected patients exposed to biologic agents is not clear. We aimed to investigate the reactivation rate in hepatitis B surface antigen-negative phase of hepatitis B virus-infected patients after biologic therapy. METHODS: Patients followed at gastroenterology, rheumatology, and dermatology clinics with a diagnosis of immune-mediated inflam matory diseases were screened. Immune-mediated inflammatory diseases patients exposed to biologic agents with a negative hepatitis B surface antigen and positive hepatitis B core immunoglobulin G antibody were included in the study. RESULTS: We screened 8266 immune-mediated inflammatory disease patients, and 2484 patients were identified as exposed to biologic agents. Two hundred twenty-one patients were included in the study. The mean age was 54.08 ± 11.69 years, and 115 (52.0%) patients were female. The median number of different biologic subtype use was 1 (range: 1-6). The mean biologic agent exposure time was 55 (range: 2-179) months. One hundred and fifty-two (68.8%) patients used a concomitant immunomodulatory agent, and 84 (38.0%) patients were exposed to corticosteroids during biologic use. No hepatitis B reactivation with a reverse seroconversion of hepatitis B surface antigen positivity was seen. Antiviral prophylaxis for hepatitis B was applied to 48 (21.7%) patients. Hepatitis B virus-DNA was screened in 56 (25.3%) patients prior to the biologic exposure. Two patients without antiviral prophylaxis had hepatitis B virus-DNA reactivation with a negative hepatitis B surface antigen during exposure to the biologic agent. CONCLUSION: We found 2 reactivations and no hepatitis B surface antigen seroconversion in our cohort. Antiviral prophylaxis for patients exposed to biologic agents may need to be discussed in more detail.


Subject(s)
Biological Products , Hepatitis B Surface Antigens , Hepatitis B , Latent Infection , Virus Activation , Adult , Aged , Female , Humans , Male , Middle Aged , Antigens, Surface , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Biological Products/adverse effects , Biological Products/therapeutic use , Biological Therapy/adverse effects , Biological Therapy/methods , Hepatitis B/drug therapy , Hepatitis B/immunology , Hepatitis B/prevention & control , Hepatitis B/virology , Hepatitis B Antibodies , Hepatitis B Surface Antigens/immunology , Hepatitis B virus/physiology , Retrospective Studies , Latent Infection/etiology , Latent Infection/immunology , Virus Activation/drug effects , Virus Activation/immunology
11.
Hepatol Int ; 17(2): 281-290, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36580258

ABSTRACT

BACKGROUND AND AIMS: Programmed cell death protein-1 (PD-1) inhibitors plus tyrosine kinase inhibitor (TKI) have dramatically improved survival of patients with advanced hepatocellular carcinoma (HCC). However, the risk of hepatitis B virus (HBV) reactivation from these antitumor medications remains unclear. METHODS: Patients receiving TKI monotherapy (TKI group) or TKI combined with PD-1 inhibitors (combination group) were included. The primary endpoint was HBV reactivation as defined by an increase in HBV DNA titer by at least 1 log (tenfold) from baseline. The secondary endpoints included tumor progression and overall survival. RESULTS: Four hundred and ninety-nine patients met the inclusion criteria, including 296 patients in the TKI group and 203 patients in the combination group. The 3-, 6- and 12-month cumulative incidence rates of HBV reactivation in the TKI group vs. combination group were 7.8%, 12.8% and 21.3% vs. 9.9%, 19.2% and 30.0%, respectively (p = 0.02). The Cox proportional hazard model indicated that combination therapy (HR 1.41, 95% CI 1.00-1.99, p = 0.05), ALT > 40 U/ml (HR 1.50, 95% CI 1.05-2.16, p = 0.03), and tumor size > 5 cm (HR 1.58, 95% CI 1.10-2.28, p = 0.01) were independent risk factors for HBV reactivation. Compared with the HBV reactivation group, the progression-free survival and overall survival of patients in the HBV non-reactivation group were significantly prolonged (p < 0.001 and p = 0.001). CONCLUSIONS: Patients who received TKI combined with PD-1 inhibitors had a greater risk for HBV reactivation, and those with HBV reactivation had a higher rate of tumor progression and shorter survival time, than those receiving TKI alone.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B , Immune Checkpoint Inhibitors , Liver Neoplasms , Virus Activation , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Hepatitis B/physiopathology , Hepatitis B/virology , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/virology , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , /therapeutic use , Virus Activation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
13.
J Virol ; 96(15): e0037222, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35867565

ABSTRACT

Elimination of human immunodeficiency virus (HIV) reservoirs is a critical endpoint to eradicate HIV. One therapeutic intervention against latent HIV is "shock and kill." This strategy is based on the transcriptional activation of latent HIV with a latency-reversing agent (LRA) with the consequent killing of the reactivated cell by either the cytopathic effect of HIV or the immune system. We have previously found that the small molecule 3-hydroxy-1,2,3-benzotriazin-4(3H)-one (HODHBt) acts as an LRA by increasing signal transducer and activator of transcription (STAT) factor activation mediated by interleukin-15 (IL-15) in cells isolated from aviremic participants. The IL-15 superagonist N-803 is currently under clinical investigation to eliminate latent reservoirs. IL-15 and N-803 share similar mechanisms of action by promoting the activation of STATs and have shown some promise in preclinical models directed toward HIV eradication. In this work, we evaluated the ability of HODHBt to enhance IL-15 signaling in natural killer (NK) cells and the biological consequences associated with increased STAT activation in NK cell effector and memory-like functions. We showed that HODHBt increased IL-15-mediated STAT phosphorylation in NK cells, resulting in increases in the secretion of CXCL-10 and interferon gamma (IFN-γ) and the expression of cytotoxic proteins, including granzyme B, granzyme A, perforin, granulysin, FASL, and TRAIL. This increased cytotoxic profile results in increased cytotoxicity against HIV-infected cells and different tumor cell lines. HODHBt also improved the generation of cytokine-induced memory-like NK cells. Overall, our data demonstrate that enhancing the magnitude of IL-15 signaling with HODHBt favors NK cell cytotoxicity and memory-like generation, and thus, targeting this pathway could be further explored for HIV cure interventions. IMPORTANCE Several clinical trials targeting the HIV latent reservoir with LRAs have been completed. In spite of a lack of clinical benefit, they have been crucial to elucidate hurdles that "shock and kill" strategies have to overcome to promote an effective reduction of the latent reservoir to lead to a cure. These hurdles include low reactivation potential mediated by LRAs, the negative influence of some LRAs on the activity of natural killer and effector CD8 T cells, an increased resistance to apoptosis of latently infected cells, and an exhausted immune system due to chronic inflammation. To that end, finding therapeutic strategies that can overcome some of these challenges could improve the outcome of shock and kill strategies aimed at HIV eradication. Here, we show that the LRA HODHBt also improves IL-15-mediated NK cell effector and memory-like functions. As such, pharmacological enhancement of IL-15-mediated STAT activation can open new therapeutic avenues toward an HIV cure.


Subject(s)
HIV-1 , Immunologic Memory , Interleukin-15 , Killer Cells, Natural , STAT Transcription Factors , Triazines , Virus Latency , Humans , Cell Line, Tumor , Chemokine CXCL10 , Cytotoxicity Tests, Immunologic , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/growth & development , HIV-1/immunology , Immunologic Memory/drug effects , Interferon-gamma , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , STAT Transcription Factors/metabolism , Transcriptional Activation/drug effects , Triazines/pharmacology , Virus Activation/drug effects , Virus Latency/drug effects
14.
J Virol ; 96(12): e0044522, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35638831

ABSTRACT

HIV persistence requires lifelong antiretroviral therapy (ART), calling for a cure. The histone deacetylase inhibitor, romidepsin, is used in the "shock and kill" approach with the goal of reactivating virus and subsequently clearing infected cells through cell-mediated immune responses. We tested serial and double infusions of romidepsin in a rhesus macaque (RM) model of SIV functional cure, which controls virus without ART. Off ART, romidepsin reactivated SIV in all RMs. Subsequent infusions resulted in diminished reactivation, and two RMs did not reactivate the virus after the second or third infusions. Therefore, those two RMs received CD8-depleting antibody to assess the replication competence of the residual reservoir. The remaining RMs received double infusions, i.e., two doses separated by 48-h. Double infusions were well tolerated, induced immune activation, and effectively reactivated SIV. Although reactivation was gradually diminished, cell-associated viral DNA was minimally changed, and viral outgrowth occurred in 4/5 RMs. In the RM which did not reactivate after CD8 depletion, viral outgrowth was not detected in peripheral blood mononuclear cells (PBMC)-derived CD4+ cells. The frequency of SIV-specific CD8+ T cells increased after romidepsin administration, and the increased SIV-specific immune responses were associated, although not statistically, with the diminished reactivation. Thus, our data showing sequential decreases in viral reactivation with repeated romidepsin administrations with all RMs and absence of viral reactivation after CD8+ T-cell depletion in one animal suggest that, in the context of healthy immune responses, romidepsin affected the inducible viral reservoir and gradually increased immune-mediated viral control. Given the disparities between the results of romidepsin administration to ART-suppressed SIVmac239-infected RMs and HIV-infected normal progressors compared to our immune-healthy model, our data suggest that improving immune function for greater SIV-specific responses should be the starting point of HIV cure strategies. IMPORTANCE HIV cure is sought after due to the prevalence of comorbidities that occur in persons with HIV. One of the most investigated HIV cure strategies is the "shock and kill" approach. Our study investigated the use of romidepsin, a histone deacetylase (HDAC) inhibitor, in our rhesus macaque model of functional cure, which allows for better resolution of viral reactivation due to the lack of antiretroviral therapy. We found that repeated rounds of romidepsin resulted in gradually diminished viral reactivation. One animal inevitably lacked replication-competent virus in the blood. With the accompanying enhancement of the SIV-specific immune response, our data suggest that there is a reduction of the viral reservoir in one animal by the cell-mediated immune response. With the differences observed between our model and persons living with HIV (PWH) treated with romidepsin, specifically in the context of a healthy immune system in our model, our data thereby indicate the importance of restoring the immune system for cure strategies.


Subject(s)
Anti-Retroviral Agents , Depsipeptides , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes , Depsipeptides/pharmacology , HIV Infections , Leukocytes, Mononuclear/virology , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Viral Load , Virus Activation/drug effects , Virus Replication
15.
Pharmacol Res ; 178: 106181, 2022 04.
Article in English | MEDLINE | ID: mdl-35301112

ABSTRACT

To date, an estimated 300 million people worldwide have been infected with chronic hepatitis B virus (HBV). Although anti-HBV therapies have improved the long-term survival profile of chronic carriers, viral reactivation still poses a significant challenge for preventing HBV-related hepatitis, hepatocellular carcinoma (HCC), and death. Immuno-modulating drugs, which are widely applied in managing rheumatic conditions, are commonly associated with HBV reactivation (HBVr) as a result of drug-induced immune suppression. However, there are few reports on the risk of HBVr and the medication management plan for HBV carriers, especially rheumatic patients. In this review, we summarize immuno-modulating drug-induced HBVr during rheumatoid therapy and its preventive strategies for HBVr-induced liver diseases, especially cirrhosis and HCC. These findings will assist with developing treatments for rheumatic patients, and prevent HBV-related cirrhosis and HCC.


Subject(s)
Antirheumatic Agents , Carcinoma, Hepatocellular , Chemical and Drug Induced Liver Injury , Hepatitis B, Chronic , Herpesvirus 1, Cercopithecine , Liver Neoplasms , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/prevention & control , Chemical and Drug Induced Liver Injury/drug therapy , Hepatitis B virus , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/drug therapy , Humans , Liver Cirrhosis/drug therapy , Liver Neoplasms/drug therapy , Liver Neoplasms/prevention & control , Virus Activation/drug effects
16.
Nature ; 603(7900): 315-320, 2022 03.
Article in English | MEDLINE | ID: mdl-35197633

ABSTRACT

Colibactin is a chemically unstable small-molecule genotoxin that is produced by several different bacteria, including members of the human gut microbiome1,2. Although the biological activity of colibactin has been extensively investigated in mammalian systems3, little is known about its effects on other microorganisms. Here we show that colibactin targets bacteria that contain prophages, and induces lytic development through the bacterial SOS response. DNA, added exogenously, protects bacteria from colibactin, as does expressing a colibactin resistance protein (ClbS) in non-colibactin-producing cells. The prophage-inducing effects that we observe apply broadly across different phage-bacteria systems and in complex communities. Finally, we identify bacteria that have colibactin resistance genes but lack colibactin biosynthetic genes. Many of these bacteria are infected with predicted prophages, and we show that the expression of their ClbS homologues provides immunity from colibactin-triggered induction. Our study reveals a mechanism by which colibactin production could affect microbiomes and highlights a role for microbial natural products in influencing population-level events such as phage outbreaks.


Subject(s)
Bacteria , Bacterial Toxins , Peptides , Polyketides , Prophages , Virus Activation , Bacteria/drug effects , Bacteria/virology , Bacterial Toxins/metabolism , Bacterial Toxins/pharmacology , Bacteriolysis/drug effects , Microbial Interactions/drug effects , Peptides/metabolism , Peptides/pharmacology , Polyketides/metabolism , Polyketides/pharmacology , Prophages/drug effects , Prophages/physiology , SOS Response, Genetics/drug effects , Virus Activation/drug effects
17.
Int J Mol Sci ; 23(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35163532

ABSTRACT

Since the beginning of the HIV epidemic, lasting more than 30 years, the main goal of scientists was to develop effective methods for the prevention and treatment of HIV infection. Modern medicines have reduced the death rate from AIDS by 80%. However, they still have side effects and are very expensive, dictating the need to search for new drugs. Earlier, it was shown that phospholipases A2 (PLA2s) from bee and snake venoms block HIV replication, the effect being independent on catalytic PLA2 activity. However, the antiviral activity of human PLA2s against Lentiviruses depended on catalytic function and was mediated through the destruction of the viral membrane. To clarify the role of phospholipolytic activity in antiviral effects, we analyzed the anti-HIV activity of several snake PLA2s and found that the mechanisms of their antiviral activity were similar to that of mammalian PLA2. Our results indicate that snake PLA2s are capable of inhibiting syncytium formation between chronically HIV-infected cells and healthy CD4-positive cells and block HIV binding to cells. However, only dimeric PLA2s had pronounced virucidal and anti-HIV activity, which depended on their catalytic activity. The ability of snake PLA2s to inactivate the virus may provide an additional barrier to HIV infection. Thus, snake PLA2s might be considered as candidates for lead molecules in anti-HIV drug development.


Subject(s)
Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , Giant Cells/cytology , HIV-1/physiology , Phospholipases A2/pharmacology , Snake Venoms/enzymology , Snakes/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Giant Cells/drug effects , Giant Cells/virology , HIV-1/drug effects , Humans , Inhibitory Concentration 50 , Reptilian Proteins/pharmacology , Snakes/classification , Virus Activation/drug effects , Virus Attachment/drug effects
18.
PLoS Pathog ; 18(1): e1010245, 2022 01.
Article in English | MEDLINE | ID: mdl-35041707

ABSTRACT

Activation of the NF-κB signaling pathway by Protein Kinase C (PKC) agonists is a potent mechanism for human immunodeficiency virus (HIV) latency disruption in vitro. However, significant toxicity risks and the lack of evidence supporting their activity in vivo have limited further evaluation of PKC agonists as HIV latency-reversing agents (LRA) in cure strategies. Here we evaluated whether GSK445A, a stabilized ingenol-B derivative, can induce HIV/simian immunodeficiency virus (SIV) transcription and virus production in vitro and demonstrate pharmacological activity in nonhuman primates (NHP). CD4+ T cells from people living with HIV and from SIV+ rhesus macaques (RM) on antiretroviral therapy (ART) exposed in vitro to 25 nM of GSK445A produced cell-associated viral transcripts as well as viral particles at levels similar to those induced by PMA/Ionomycin, indicating that GSK445A can potently reverse HIV/SIV latency. Importantly, these concentrations of GSK445A did not impair the proliferation or survival of HIV-specific CD8+ T cells, but instead, increased their numbers and enhanced IFN-γ production in response to HIV peptides. In vivo, GSK445A tolerability was established in SIV-naïve RM at 15 µg/kg although tolerability was reduced in SIV-infected RM on ART. Increases in plasma viremia following GSK445A administration were suggestive of increased SIV transcription in vivo. Collectively, these results indicate that GSK445A is a potent HIV/SIV LRA in vitro and has a tolerable safety profile amenable for further evaluation in vivo in NHP models of HIV cure/remission.


Subject(s)
Diterpenes/pharmacology , HIV , Protein Kinase C/drug effects , Simian Immunodeficiency Virus , Virus Activation/drug effects , Virus Latency/drug effects , Animals , Humans , Macaca mulatta , Protein Kinase C/metabolism , RNA, Viral/drug effects , Transcription, Genetic
19.
J Virol ; 96(5): e0213021, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35019726

ABSTRACT

Acute infection of the ocular, oral, or nasal cavity by bovine herpesvirus 1 (BoHV-1) culminates in lifelong latency in sensory neurons within trigeminal ganglia. The BoHV-1 latency reactivation cycle, including calves latently infected with commercially available modified live vaccines, can lead to reproductive complications, including abortions. Recent studies demonstrated progesterone stimulated BoHV-1 productive infection and sporadically induced reactivation from latency in male rabbits. The progesterone receptor (PR) and progesterone transactivate the immediate early transcription unit 1 (IEtu1) promoter and the infected cell protein 0 (bICP0) early promoter. These viral promoters drive expression of two viral transcriptional regulatory proteins (bICP0 and bICP4) that are crucial for productive infection. Based on these observations, we hypothesize that progesterone induces reactivation in a subset of calves latently infected with BoHV-1. These studies demonstrated progesterone was less efficient than dexamethasone at initiating reactivation from latency in female calves. Notably, heat stress correlated with enhancing the ability of progesterone to induce reactivation from latency. Previous studies demonstrated that heat stress activates the glucocorticoid receptor (GR), which suggested GR activation augments progesterone-mediated reactivation from latency. Additional studies revealed GR and PR cooperatively stimulated productive infection and synergistically transactivated the IEtu1 promoter when cultures were treated with dexamethasone. Mutating one or both GR binding sites in the IEtu1 promoter blocked transactivation. Collectively, these studies indicated that progesterone intermittently triggered reactivation from latency, and heat stress augmented reactivation from reactivation. Finally, these studies suggest progesterone enhances virus spread in tissues and cells where PR is abundantly expressed. IMPORTANCE Steroid hormone fluctuations are predicted to enhance or initiate bovine herpesvirus 1 (BoHV-1) replication and virus spread in cattle. For example, stress increases the incidence of BoHV-1 reactivation from latency in cattle, and the synthetic corticosteroid dexamethasone consistently induces reactivation from latency. The glucocorticoid receptor (GR) and dexamethasone stimulate key viral regulatory promoters and productive infection, in part because the viral genome contains numerous consensus GR-responsive elements (GREs). The progesterone receptor (PR) and GR belong to the type I nuclear hormone receptor family. PR and progesterone specifically bind to and transactivate viral promoters that contain GREs and stimulate BoHV-1 productive infection. Although progesterone did not induce reactivation from latency in female calves as efficiently as dexamethasone, heat stress enhanced progesterone-mediated reactivation from latency. Consequently, we predict that low levels of stressful stimuli can cooperate with progesterone to induce reactivation from latency or promote virus spread.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Bovine , Progesterone , Animals , Cattle , Dexamethasone/pharmacology , Female , Heat-Shock Response , Herpesviridae Infections/virology , Herpesvirus 1, Bovine/physiology , Male , Progesterone/pharmacology , Rabbits , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Virus Activation/drug effects , Virus Latency/drug effects
20.
Ann Hematol ; 101(3): 631-641, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34981143

ABSTRACT

Hepatitis B virus reactivation (HBVr) is not uncommon in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. Hepatitis B surface antigen (HBsAg)-positive patients receiving allo-HSCT have a very high risk of HBVr. However, the validity of prophylactic antiviral treatment in HBsAg-positive allo-HSCT recipients has not been well studied. We aimed to add experience in dealing with HBsAg-positive patients following allo-HSCT. We conducted a cohort study that included 11 years of data of HBsAg-positive allo-HSCT patients in multiple centers. The cumulative incidence of HBVr with antiviral prophylaxis at 60 months following transplantation was 8.9%. Both lamivudine (LAM) and entecavir (ETV) effectively reduced the incidence of HBVr. Patients with absent-mild cGVHD had a lower HBVr rate than that of patients with moderate-severe cGVHD (HR = 0.201, P = 0.020). The incidence of HBsAg seroclearance at 60 months following transplantation was 34.3%. Recipients accepting from anti-HBs-negative donors were associated with a lower HBsAg seroclearance rate than that of those accepting from anti-HBs-positive donors (HR=0.255, P < 0.001). The peripheral blood stem cell (PBSC) donor source had a higher HBsAg seroclearance rates than that of the PBSC plus bone marrow stem cell source (HR = 4.700, P = 0.047). The prophylactic antiviral treatment effectively reduced HBVr in HBsAg-positive recipients receiving allo-HSCT. HBsAg-positive recipients accept anti-HBs-positive PBSC donor sources may facilitate the acquisition of HBsAg seroclearance after transplantation.


Subject(s)
Antiviral Agents/therapeutic use , Guanine/analogs & derivatives , Hematopoietic Stem Cell Transplantation , Hepatitis B virus/drug effects , Hepatitis B/prevention & control , Lamivudine/therapeutic use , Adult , Female , Guanine/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatitis B/blood , Hepatitis B/etiology , Hepatitis B/virology , Hepatitis B Surface Antigens/blood , Hepatitis B virus/physiology , Humans , Male , Middle Aged , Retrospective Studies , Transplantation, Homologous/adverse effects , Virus Activation/drug effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...