Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Lancet Microbe ; 5(3): e216-e225, 2024 03.
Article in English | MEDLINE | ID: mdl-38278167

ABSTRACT

BACKGROUND: Accurate quantitation of immune markers is crucial for ensuring reliable assessment of vaccine efficacy against infectious diseases. This study was designed to confirm standardised performance of SARS-CoV-2 assays used to evaluate COVID-19 vaccine candidates at the initial seven laboratories (in North America, Europe, and Asia) of the Coalition for Epidemic Preparedness Innovations (CEPI) Centralized Laboratory Network (CLN). METHODS: Three ELISAs (pre-spike protein, receptor binding domain, and nucleocapsid), a microneutralisation assay (MNA), a pseudotyped virus-based neutralisation assay (PNA), and an IFN-γ T-cell ELISpot assay were developed, validated or qualified, and transferred to participating laboratories. Immune responses were measured in ELISA laboratory units (ELU) for ELISA, 50% neuralisation dilution (ND50) for MNA, 50% neutralisation titre (NT50) for PNA, and spot-forming units for the ELISpot assay. Replicate assay results of well characterised panels and controls of blood samples from individuals with or without SARS-CoV-2 infection were evaluated by geometric mean ratios, standard deviation, linear regression, and Spearman correlation analysis for consistency, accuracy, and linearity of quantitative measurements across all laboratories. FINDINGS: High reproducibility of results across all laboratories was demonstrated, with interlaboratory precision of 4·1-7·7% coefficient of variation for all three ELISAs, 3·8-19·5% for PNA, and 17·1-24·1% for MNA, over a linear range of 11-30 760 ELU per mL for the three ELISAs, 14-7876 NT50 per mL for PNA, and 21-25 587 ND50 per mL for MNA. The MNA was also adapted for detection of neutralising antibodies against the major SARS-CoV-2 variants of concern. The results of PNA and MNA (r=0·864) and of ELISA and PNA (r=0·928) were highly correlated. The IFN-γ ELISpot interlaboratory variability was 15·9-49·9% coefficient of variation. Sensitivity and specificity were close to 100% for all assays. INTERPRETATION: The CEPI CLN provides accurate quantitation of anti-SARS-CoV-2 immune response across laboratories to allow direct comparisons of different vaccine formulations in different geographical areas. Lessons learned from this programme will serve as a model for faster responses to future pandemic threats and roll-out of effective vaccines. FUNDING: CEPI.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19 Vaccines , Laboratories , Reproducibility of Results , Antibodies, Viral , Immunity
2.
Front Immunol ; 13: 1032356, 2022.
Article in English | MEDLINE | ID: mdl-36532027

ABSTRACT

Oncolytic viruses (OVs) are promising anticancer treatments that specifically replicate in and kill cancer cells and have profound immunostimulatory effects. We previously reported the potential of vanadium-based compounds such as vanadyl sulfate (VS) as immunostimulatory enhancers of OV immunotherapy. These compounds, in conjunction with RNA-based OVs such as oncolytic vesicular stomatitis virus (VSVΔ51), improve viral spread and oncolysis, leading to long-term antitumor immunity and prolonged survival in resistant tumor models. This effect is associated with a virus-induced antiviral type I IFN response shifting towards a type II IFN response in the presence of vanadium. Here, we investigated the systemic impact of VS+VSVΔ51 combination therapy to understand the immunological mechanism of action leading to improved antitumor responses. VS+VSVΔ51 combination therapy significantly increased the levels of IFN-γ and IL-6, and improved tumor antigen-specific T-cell responses. Supported by immunological profiling and as a proof of concept for the design of more effective therapeutic regimens, we found that local delivery of IL-12 using VSVΔ51 in combination with VS further improved therapeutic outcomes in a syngeneic CT26WT colon cancer model.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Cytokines , Vanadium , Immunotherapy , Immunity , Chemokines
3.
Front Immunol ; 13: 1029269, 2022.
Article in English | MEDLINE | ID: mdl-36405739

ABSTRACT

Colorectal cancer is the third most diagnosed cancer and the second leading cause of cancer mortality worldwide, highlighting an urgent need for new therapeutic options and combination strategies for patients. The orchestration of potent T cell responses against human cancers is necessary for effective antitumour immunity. However, regression of a limited number of cancers has been induced by immune checkpoint inhibitors, T cell engagers (TCEs) and/or oncolytic viruses. Although one TCE has been FDA-approved for the treatment of hematological malignancies, many challenges exist for the treatment of solid cancers. Here, we show that TCEs targeting CEACAM5 and CD3 stimulate robust activation of CD4 and CD8-positive T cells in in vitro co-culture models with colorectal cancer cells, but in vivo efficacy is hindered by a lack of TCE retention in the tumour microenvironment and short TCE half-life, as demonstrated by HiBiT bioluminescent TCE-tagging technology. To overcome these limitations, we engineered Bispecific Engager Viruses, or BEVirs, a novel tumour-targeted vaccinia virus platform for intra-tumour delivery of these immunomodulatory molecules. We characterized virus-mediated TCE-secretion, TCE specificity and functionality from infected colorectal cancer cells and patient tumour samples, as well as TCE cytotoxicity in spheroid models, in the presence and absence of T cells. Importantly, we show regression of colorectal tumours in both syngeneic and xenograft mouse models. Our data suggest that a different profile of cytokines may contribute to the pro-inflammatory and immune effects driven by T cells in the tumour microenvironment to provide long-lasting immunity and abscopal effects. We establish combination regimens with immune checkpoint inhibitors for aggressive colorectal peritoneal metastases. We also observe a significant reduction in lung metastases of colorectal tumours through intravenous delivery of our oncolytic virus driven T-cell based combination immunotherapy to target colorectal tumours and FAP-positive stromal cells or CTLA4-positive Treg cells in the tumour microenvironment. In summary, we devised a novel combination strategy for the treatment of colorectal cancers using oncolytic vaccinia virus to enhance immune-payload delivery and boost T cell responses within tumours.


Subject(s)
Colorectal Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Mice , Animals , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Vaccinia virus , Disease Models, Animal , Colorectal Neoplasms/therapy , Tumor Microenvironment
4.
Mol Ther ; 30(10): 3270-3283, 2022 10 05.
Article in English | MEDLINE | ID: mdl-35619558

ABSTRACT

Profound natural killer (NK) cell suppression after cancer surgery is a main driver of metastases and recurrence, for which there is no clinically approved intervention available. Surgical stress is known to cause systemic postoperative changes that negatively modulate NK cell function including the expansion of surgery-induced myeloid-derived suppressor cells (Sx-MDSCs) and a marked reduction in arginine bioavailability. In this study, we determine that Sx-MDSCs regulate systemic arginine levels in the postoperative period and that restoring arginine imbalance after surgery by dietary intake alone was sufficient to significantly reduce surgery-induced metastases in our preclinical murine models. Importantly, the effects of perioperative arginine were dependent upon NK cells. Although perioperative arginine did not prevent immediate NK cell immunoparalysis after surgery, it did accelerate their return to preoperative cytotoxicity, interferon gamma secretion, and activating receptor expression. Finally, in a cohort of patients with colorectal cancer, postoperative arginine levels were shown to correlate with their Sx-MDSC levels. Therefore, this study lends further support for the use of perioperative arginine supplementation by improving NK cell recovery after surgery.


Subject(s)
Arginine , Myeloid-Derived Suppressor Cells , Animals , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Mice
5.
Nat Commun ; 13(1): 1898, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35393414

ABSTRACT

Recent advances in cancer therapeutics clearly demonstrate the need for innovative multiplex therapies that attack the tumour on multiple fronts. Oncolytic or "cancer-killing" viruses (OVs) represent up-and-coming multi-mechanistic immunotherapeutic drugs for the treatment of cancer. In this study, we perform an in-vitro screen based on virus-encoded artificial microRNAs (amiRNAs) and find that a unique amiRNA, herein termed amiR-4, confers a replicative advantage to the VSVΔ51 OV platform. Target validation of amiR-4 reveals ARID1A, a protein involved in chromatin remodelling, as an important player in resistance to OV replication. Virus-directed targeting of ARID1A coupled with small-molecule inhibition of the methyltransferase EZH2 leads to the synthetic lethal killing of both infected and uninfected tumour cells. The bystander killing of uninfected cells is mediated by intercellular transfer of extracellular vesicles carrying amiR-4 cargo. Altogether, our findings establish that OVs can serve as replicating vehicles for amiRNA therapeutics with the potential for combination with small molecule and immune checkpoint inhibitor therapy.


Subject(s)
Extracellular Vesicles , MicroRNAs , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , MicroRNAs/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics
6.
EClinicalMedicine ; 45: 101323, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35284808

ABSTRACT

Background: Production of affordable coronavirus disease 2019 (COVID-19) vaccines in low- and middle-income countries is needed. NDV-HXP-S is an inactivated egg-based recombinant Newcastle disease virus vaccine expressing the spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It's being developed by public sector manufacturers in Thailand, Vietnam, and Brazil; herein are initial results from Thailand. Methods: This phase 1 stage of a randomised, dose-escalation, observer-blind, placebo-controlled, phase 1/2 trial was conducted at the Vaccine Trial Centre, Mahidol University (Bangkok). Healthy males and non-pregnant females, aged 18-59 years and negative for SARS-CoV-2 antibodies, were eligible. Participants were randomised to receive one of six treatments by intramuscular injection twice, 28 days apart: 1 µg, 1 µg+CpG1018 (a toll-like receptor 9 agonist), 3 µg, 3 µg+CpG1018, 10 µg, or placebo. Participants and personnel assessing outcomes were masked to treatment. The primary outcomes were solicited and spontaneously reported adverse events (AEs) during 7 and 28 days after each vaccination, respectively. Secondary outcomes were immunogenicity measures (anti-S IgG and pseudotyped virus neutralisation). An interim analysis assessed safety at day 57 in treatment-exposed individuals and immunogenicity through day 43 per protocol. ClinicalTrials.gov (NCT04764422). Findings: Between March 20 and April 23, 2021, 377 individuals were screened and 210 were enroled (35 per group); all received dose one; five missed dose two. The most common solicited AEs among vaccinees, all predominantly mild, were injection site pain (<63%), fatigue (<35%), headache (<32%), and myalgia (<32%). The proportion reporting a vaccine-related AE ranged from 5·7% to 17·1% among vaccine groups and was 2·9% in controls; there was no vaccine-related serious adverse event. The 10 µg formulation's immunogenicity ranked best, followed by 3 µg+CpG1018, 3 µg, 1 µg+CpG1018, and 1 µg formulations. On day 43, the geometric mean concentrations of 50% neutralising antibody ranged from 122·23 international units per mL (IU/mL; 1 µg, 95% confidence interval (CI) 86·40-172·91) to 474·35 IU/mL (10 µg, 95% CI 320·90-701·19), with 93·9% to 100% of vaccine groups attaining a ≥ 4-fold increase over baseline. Interpretation: NDV-HXP-S had an acceptable safety profile and potent immunogenicity. The 3 µg and 3 µg+CpG1018 formulations advanced to phase 2. Funding: National Vaccine Institute (Thailand), National Research Council (Thailand), Bill & Melinda Gates Foundation, National Institutes of Health (USA).

7.
Mol Ther ; 30(5): 1885-1896, 2022 05 04.
Article in English | MEDLINE | ID: mdl-34687845

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic requires the continued development of safe, long-lasting, and efficacious vaccines for preventive responses to major outbreaks around the world, and especially in isolated and developing countries. To combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we characterize a temperature-stable vaccine candidate (TOH-Vac1) that uses a replication-competent, attenuated vaccinia virus as a vector to express a membrane-tethered spike receptor binding domain (RBD) antigen. We evaluate the effects of dose escalation and administration routes on vaccine safety, efficacy, and immunogenicity in animal models. Our vaccine induces high levels of SARS-CoV-2 neutralizing antibodies and favorable T cell responses, while maintaining an optimal safety profile in mice and cynomolgus macaques. We demonstrate robust immune responses and protective immunity against SARS-CoV-2 variants after only a single dose. Together, these findings support further development of our novel and versatile vaccine platform as an alternative or complementary approach to current vaccines.


Subject(s)
COVID-19 , Vaccines , Animals , Mice , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Immunity , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus , T-Lymphocytes
8.
medRxiv ; 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34580673

ABSTRACT

BACKGROUND: Production of affordable coronavirus disease 2019 (COVID-19) vaccines in low- and middle-income countries is needed. NDV-HXP-S is an inactivated egg-based Newcastle disease virus vaccine expressing the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It's being developed in Thailand, Vietnam, and Brazil; herein are initial results from Thailand. METHODS: This phase 1 stage of a randomised, dose-escalation, observer-blind, placebo-controlled, phase 1/2 trial was conducted at the Vaccine Trial Centre, Mahidol University (Bangkok). Healthy adults aged 18-59 years, non-pregnant and negative for SARS-CoV-2 antibodies were eligible. Participants were block randomised to receive one of six treatments by intramuscular injection twice, 28 days apart: 1 µg±CpG1018 (a toll-like receptor 9 agonist), 3 µg±CpG1018, 10 µg, or placebo. Participants and personnel assessing outcomes were masked to treatment. The primary outcomes were solicited and spontaneously reported adverse events (AEs) during 7 and 28 days after each vaccination, respectively. Secondary outcomes were immunogenicity measures (anti-S IgG and pseudotyped virus neutralisation). An interim analysis assessed safety at day 57 in treatment-exposed individuals and immunogenicity through day 43 per protocol. ClinicalTrials.gov ( NCT04764422 ). FINDINGS: Between March 20 and April 23, 2021, 377 individuals were screened and 210 were enrolled (35 per group); all received dose one; five missed dose two. The most common solicited AEs among vaccinees, all predominantly mild, were injection site pain (<63%), fatigue (<35%), headache (<32%), and myalgia (<32%). The proportion reporting a vaccine-related AE ranged from 5·7% to 17·1% among vaccine groups and was 2·9% in controls; there was no vaccine-related serious adverse event. The 10 µg formulation's immunogenicity ranked best, followed by 3 µg+CpG1018, 3 µg, 1 µg+CpG1018, and 1 µg formulations. On day 43, the geometric mean concentrations of 50% neutralising antibody ranged from 122·23 IU/mL (1 µg, 95% CI 86·40-172·91) to 474·35 IU/mL (10 µg, 95% CI 320·90-701·19), with 93·9% to 100% of vaccine groups attaining a ≥4-fold increase over baseline. INTERPRETATION: NDV-HXP-S had an acceptable safety profile and potent immunogenicity. The 3 µg and 3 µg+CpG1018 formulations advanced to phase 2. FUNDING: National Vaccine Institute (Thailand), National Research Council (Thailand), Bill & Melinda Gates Foundation, National Institutes of Health (USA).

9.
Hematol Oncol ; 39(4): 448-464, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33963789

ABSTRACT

Autologous cell vaccines use a patient's tumor cells to stimulate a broad antitumor response in vivo. This approach shows promise for treating hematologic cancers in early phase clinical trials, but overall safety and efficacy remain poorly described. We conducted a systematic review assessing the use of autologous cell vaccination in treating hematologic cancers. Primary outcomes of interest were safety and clinical response, with secondary outcomes including survival, relapse rate, correlative immune assays and health-quality related metrics. We performed a search of MEDLINE, Embase and the Cochrane Register of Controlled Trials including any interventional trial employing an autologous, whole cell product in any hematologic malignancy. Risk of bias was assessed using a modified Institute of Health Economics tool. Across 20 single arm studies, only 341 of 592 enrolled participants received one or more vaccinations. Primary reasons for not receiving vaccination included rapid disease progression/death and manufacturing challenges. Overall, few high-grade adverse events were observed. One death was reported and attributed to a GM-CSF producing allogeneic cell line co-administered with the autologous vaccine. Of 58 evaluable patients, the complete response rate was 21.0% [95% CI, 10.4%-37.8%)] and overall response rate was 35.8% (95% CI, 24.4%-49.0%). Of 97 evaluable patients for survival, the 5-years overall survival rate was 64.9% (95% CI, 52.6%-77.2%) and disease-free survival was 59.7% (95% CI, 47.7%-71.7%). We conclude that, in hematologic malignancies, based on limited available data, autologous cell vaccines are safe and display a trend towards efficacy but that challenges exist in vaccine manufacture and administration.


Subject(s)
Hematologic Neoplasms/therapy , Vaccines/therapeutic use , Adult , Aged , Female , Humans , Male , Middle Aged , Vaccines/pharmacology
10.
BMJ Open ; 10(6): e034714, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32518209

ABSTRACT

INTRODUCTION: Autologous cancer cell vaccines are promising personalised immunotherapeutic options for solid and haematological malignancies that uses the patient's own cells to arm an immune response. Evidence suggests that among patients receiving these vaccines, those who mount an immune response against their own tumour cells have better prognosis, and a myriad of preclinical studies have demonstrated the same. Recently, two autologous cell vaccines Vigil and OncoVAX have made it to phase III clinical trials. Here, we outline a protocol to be used for two separate systematic reviews using a parallel approach for inclusion criteria, data extraction and analysis for autologous cell vaccines in (1) solid and (2) haematological malignancies. We aim to review evidence from controlled and uncontrolled interventional studies of autologous cell vaccines administered to patients with cancer to determine their historical efficacy (with or without associated adjuvants or modifications) with clinical response rates and safety outcomes being of particular importance. METHODS AND ANALYSIS: We will search MEDLINE (OVID interface, including In-Process and Epub Ahead of Print), Embase (OVID interface) and the Cochrane Central Register of Controlled Trials (Wiley interface) for articles published from 1947 until 30 July 2018 (date search was performed). Studies will be screened first by title and abstract, then by full-text in duplicate. Interventional trials that report the use of an autologous cell vaccine to patients with cancer of any age will be included. The primary outcomes of interest in this review are clinical response (complete or overall/objective response) and safety outcomes (adverse events). Secondary outcomes include immune response, disease-free survival and overall survival. The risk of bias within studies will be assessed using the appropriate Cochrane Risk of Bias tool. If appropriate, a random effects meta-analysis will be performed to synthesise the data and report summary estimates of effect. Statistical heterogeneity will be assessed using the I2 statistic. ETHICS AND DISSEMINATION: Ethics approval is not required for this systematic review protocol as the review will solely use published literature. Results will be submitted to peer-reviewed journals for publication and presented to relevant stakeholders and scientific meetings. PROSPERO REGISTRATION NUMBER: CRD42019140187.


Subject(s)
Cancer Vaccines , Hematologic Neoplasms , Neoplasms , Humans , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Hematologic Neoplasms/therapy , Meta-Analysis as Topic , Neoplasms/therapy , Systematic Reviews as Topic
11.
J Clin Invest ; 129(9): 3499-3510, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31478911

ABSTRACT

Natural killer (NK) cells are innate cytotoxic lymphocytes involved in the surveillance and elimination of cancer. As we have learned more and more about the mechanisms NK cells employ to recognize and eliminate tumor cells, and how, in turn, cancer evades NK cell responses, we have gained a clear appreciation that NK cells can be harnessed in cancer immunotherapy. Here, we review the evidence for NK cells' critical role in combating transformed and malignant cells, and how cancer immunotherapies potentiate NK cell responses for therapeutic purposes. We highlight cutting-edge immunotherapeutic strategies in preclinical and clinical development such as adoptive NK cell transfer, chimeric antigen receptor-expressing NK cells (CAR-NKs), bispecific and trispecific killer cell engagers (BiKEs and TriKEs), checkpoint blockade, and oncolytic virotherapy. Further, we describe the challenges that NK cells face (e.g., postsurgical dysfunction) that must be overcome by these therapeutic modalities to achieve cancer clearance.


Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural , Neoplasms , Animals , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Killer Cells, Natural/transplantation , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
12.
Immunology ; 154(1): 156-165, 2018 05.
Article in English | MEDLINE | ID: mdl-29266204

ABSTRACT

Clearance of hepatitis C virus (HCV) is dependent on an effective virus-specific CD8+ T-cell response, which is dysfunctional in chronic HCV infection. Dysfunction in bulk or non-HCV-specific CD8+ T-cells in HCV infection has also been observed. This may contribute to observed reductions in immunity to other diseases (e.g. cancer, viral co-infections) in HCV-infected individuals. Evidence suggests that the HCV core protein (found in blood as free protein) may contribute to this impairment. To determine if HCV core contributes to the impairment of effector functions and survival potential of CD8+ T-cells, isolated human CD8+ T-cells from healthy donors were pre-incubated with recombinant HCV core protein for 72 hr and then stimulated in vitro to evaluate proliferation, survival potential and effector functions. Pre-incubation of stimulated CD8+ T-cells with HCV core significantly reduced their proliferation. Perforin production and degranulation were also decreased, but interferon-γ production was unchanged. Additionally, when CD8+ T-cells were treated with serum from HCV+ individuals, they produced less perforin than cells treated with healthy serum. Up-regulation of anti-apoptotic Bcl-2 was slightly lower in cells treated with HCV core, but signal transducer and activator of transcription 5 (STAT5) activation was increased, suggesting dysregulation downstream of STAT activation. Our study reveals that HCV core reduces the activity and target lysis-associated functions of CD8+ T-cells. This may contribute to the generalized impairment of CD8+ T-cells observed in HCV infection. These findings provide insight for the design of novel counteractive immune-mediated strategies including the design of effective therapeutic vaccines for use in HCV+ individuals.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cell Degranulation , Cell Proliferation , Hepacivirus/metabolism , Hepatitis C/metabolism , Lymphocyte Activation , Perforin/metabolism , STAT5 Transcription Factor/metabolism , Viral Core Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Cytotoxicity, Immunologic , Hepacivirus/immunology , Hepacivirus/pathogenicity , Hepatitis C/immunology , Hepatitis C/virology , Host-Pathogen Interactions , Humans , Interleukin-7/metabolism , Lysosomal Membrane Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
13.
Can Liver J ; 1(2): 94-105, 2018.
Article in English | MEDLINE | ID: mdl-35990718

ABSTRACT

Estimates are that more than 250,000 people in Canada are chronically infected with hepatitis C virus (HCV), and many more are unaware of their infection status. If untreated, chronic HCV infection can lead to cirrhosis and subsequent complications such as hepatocellular carcinoma. The Canadian Network on Hepatitis C, supported by the Public Health Agency of Canada and the Canadian Institutes of Health Research, has been committed to the scientific study of chronic hepatitis C and to supporting the advocacy work to improve diagnosis and access to HCV care in Canada. Although the treatment of HCV infection has been greatly advanced with direct-acting antivirals, with cure rates as high as 95%, many challenges remain in the implementation of HCV care. These issues include the lack of an effective vaccine, infection screening, treatment failure or resistance, post-cure health issues, limitations of treatment access despite increased provincial subsidization, complex needs of at-risk populations (ie, injection drug users, societal obstacles). At the 6th Canadian Symposium on HCV in March 2017, the theme "Delivering a Cure for Hepatitis C Infection: What Are the Remaining Gaps?" provided a framework in which basic scientists, clinicians, epidemiologists, social scientists, and community members interested in HCV research in Canada could showcase how they are working to address these ongoing challenges.

14.
Int J Mol Sci ; 18(8)2017 Aug 17.
Article in English | MEDLINE | ID: mdl-28817109

ABSTRACT

The physiological changes that occur immediately following cancer surgeries initiate a chain of events that ultimately result in a short pro-, followed by a prolonged anti-, inflammatory period. Natural Killer (NK) cells are severely affected during this period in the recovering cancer patient. NK cells play a crucial role in anti-tumour immunity because of their innate ability to differentiate between malignant versus normal cells. Therefore, an opportunity arises in the aftermath of cancer surgery for residual cancer cells, including distant metastases, to gain a foothold in the absence of NK cell surveillance. Here, we describe the post-operative environment and how the release of sympathetic stress-related factors (e.g., cortisol, prostaglandins, catecholamines), anti-inflammatory cytokines (e.g., IL-6, TGF-ß), and myeloid derived suppressor cells, mediate NK cell dysfunction. A snapshot of current and recently completed clinical trials specifically addressing NK cell dysfunction post-surgery is also discussed. In collecting and summarizing results from these different aspects of the surgical stress response, a comprehensive view of the NK cell suppressive effects of surgery is presented. Peri-operative therapies to mitigate NK cell suppression in the post-operative period could improve curative outcomes following cancer surgery.


Subject(s)
Cell Differentiation/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Catecholamines/immunology , Catecholamines/metabolism , Humans , Hydrocortisone/immunology , Hydrocortisone/metabolism , Killer Cells, Natural/pathology , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasms/pathology , Neoplasms/surgery , Postoperative Period , Prostaglandins/immunology , Prostaglandins/metabolism
15.
Clin Invest Med ; 40(2): E73-E80, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28447580

ABSTRACT

PURPOSE: Monitoring of hepatitis C virus (HCV) treatment response is performed by serial HCV RNA measurements; however, this may not be useful for predicting treatment success or failure with oral direct-acting antiviral agent (DAA) therapies. Liver enzyme levels, which are elevated in chronic HCV and tend to decline on therapy, may serve as a more logistically and economically feasible alternative for monitoring treatment response. SOURCE: The Ottawa Hospital Viral Hepatitis Clinic patients (n=219), receiving interferon-free oral DAA treatments, were assessed for liver enzymes and HCV RNA levels at baseline, week 4 and ≥12 weeks post-treatment. Suppression cut points used for this analysis were ALT ≤ 40U L-1 and AST ≤ 30U L-1. The primary outcome was week 12 sustained virologic response (SVR). By our analysis, all indicators had strong PPV (>90%) but limited NPV (.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Hepacivirus/pathogenicity , Hepatitis C/drug therapy , Hepatitis C/enzymology , Administration, Oral , Adult , Female , Hepatitis C/pathology , Humans , Liver/enzymology , Liver/virology , Male , Middle Aged , RNA, Viral/genetics , Treatment Outcome
16.
Aust N Z J Obstet Gynaecol ; 57(1): 57-62, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28251633

ABSTRACT

INTRODUCTION: Polyhydramnios is present in approximately 2% of pregnancies and it has been associated with a variety of adverse pregnancy outcomes. OBJECTIVES: The aim of our study was to evaluate the association between polyhydramnios and adverse pregnancy outcomes. METHODS: This was a retrospective case control study of 288 singleton pregnancies delivered in the Rotunda Hospital, Dublin, between 2013 and 2014. Polyhydramnios was defined as: (i) amniotic fluid index (AFI) ≥ 25 cm; (ii) maximal vertical pocket (MVP) of ≥8 cm; and (iii) a gestational age-specific threshold for AFI. Demographic information, obstetric and neonatal outcomes were obtained by review of hospital databases. Exclusion criteria included gestational or pre-existing diabetes, multiple pregnancy, carrying a fetus with structural or chromosomal abnormalities, Rhesus factor isoimmunisation, and TORCH screen positive. Outcomes were compared with outcomes of those without polyhydramnios. RESULTS: A total of 8798 deliveries occurred during the study period. The frequency of polyhydramnios was 1.6%. One hundred and forty-four women were selected in each group. There was no significant difference in preterm deliveries, low birth weight, low Apgar score at one minute and five minutes and perinatal mortality. However, increase in caesarean delivery rate (43.1% vs 21.5%), number of fetal distresses (17.4% vs 6.9%) and number of neonatal intensive care unit (NICU) admissions (17.4% vs 4.9%) were observed in our study. CONCLUSION: In conclusion, idiopathic polyhydramnios is associated with specific adverse outcomes, such as higher rate of caesarean delivery, fetal distress and NICU admissions. Therefore, close surveillance of these pregnancies is required, especially near term.


Subject(s)
Cesarean Section/statistics & numerical data , Fetal Distress/epidemiology , Intensive Care, Neonatal/statistics & numerical data , Polyhydramnios/diagnosis , Polyhydramnios/epidemiology , Adult , Case-Control Studies , Female , Humans , Incidence , Ireland/epidemiology , Pregnancy , Pregnancy Outcome , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...