Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.699
Filter
1.
Investig Clin Urol ; 65(3): 248-255, 2024 May.
Article in English | MEDLINE | ID: mdl-38714515

ABSTRACT

PURPOSE: This study investigated the efficacy of intravesical gemcitabine as an alternative to bacillus Calmette-Guérin (BCG) therapy. MATERIALS AND METHODS: Data were retrospectively collected across seven institutions from February 1999 to May 2023. Inclusion criteria included patients with intermediate- or high-risk non-muscle invasive bladder cancer (NMIBC) who underwent transurethral resection of bladder tumors (TURBT) and received at least four sessions of intravesical gemcitabine or BCG induction therapy. Patient characteristics, complete remission (CR), occurrence, and progression rates were compared. RESULTS: In total, 149 patients were included in this study (gemcitabine, 63; BCG, 86). No differences were apparent between the two groups in baseline characteristics, except for the follow-up period (gemcitabine, 9.2±5.9 months vs. BCG, 43.9±41.4 months, p<0.001). There were no consistent significant differences observed between the two groups in the 3-month (gemcitabine, 98.4% vs. BCG, 95.3%; p=0.848), 6-month (94.9% vs. 90.0%, respectively; p=0.793) and 1-year CR rates (84.2% vs. 83.3%, respectively; p=0.950). Also, there was no significant statistical difference in progression-free survival between the two groups (p=0.953). The occurrence rates of adverse events were similar between the groups (22.2% vs. 22.1%; p=0.989); however, the rate of Clavien-Dindo grade 2 or higher was significantly higher in the BCG group (1.6% vs. 16.3%, respectively; p<0.001). CONCLUSIONS: Intravesical gemcitabine demonstrated efficacy comparable to BCG therapy for the first year in patients with intermediate- and high-risk NMIBC. However, long-term follow-up studies are warranted.


Subject(s)
Adjuvants, Immunologic , Antimetabolites, Antineoplastic , BCG Vaccine , Deoxycytidine , Gemcitabine , Neoplasm Recurrence, Local , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Urinary Bladder Neoplasms/therapy , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Retrospective Studies , BCG Vaccine/administration & dosage , BCG Vaccine/therapeutic use , Male , Female , Administration, Intravesical , Aged , Antimetabolites, Antineoplastic/administration & dosage , Middle Aged , Adjuvants, Immunologic/administration & dosage , Cystectomy/methods , Risk Assessment , Urethra
2.
Hum Vaccin Immunother ; 20(1): 2348124, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38714332

ABSTRACT

South Korea's National Immunization Program administers the quadrivalent influenza vaccine (QIV) to manage seasonal influenza, with a particular focus on the elderly. After reviewing the safety and immune response triggered by the adjuvanted QIV (aQIV) in individuals aged 65 and older, the Ministry of Food and Drug Safety in Korea approved its use. However, the extensive impact of aQIV on public health is yet to be fully understood. This study assessed the cost-effectiveness of replacing QIV with aQIV in South Korean adults aged 65 years and older. A dynamic transmission model, calibrated with national influenza data, was applied to compare the influence of aQIV and QIV on older adults and the broader population throughout a single influenza season. This study considered both the direct and indirect effects of vaccination on the elderly. We derived the incremental cost-effectiveness ratios (ICERs) from quality-adjusted life-years (QALYs) and costs incurred, validated through a probabilistic sensitivity analysis with 5,000 simulations. Findings suggest that transitioning to aQIV from QIV in the elderly would be cost-effective, particularly if aQIV's efficacy reaches or exceeds 56.1%. With an ICER of $29,267/QALY, considerably lower than the $34,998/QALY willingness-to-pay threshold, aQIV presents as a cost-effective option. Thus, implementing aQIV with at least 56.1% efficacy is beneficial from both financial and public health perspectives in mitigating seasonal influenza in South Korea.


Subject(s)
Adjuvants, Immunologic , Cost-Benefit Analysis , Influenza Vaccines , Influenza, Human , Humans , Influenza Vaccines/economics , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Republic of Korea , Aged , Influenza, Human/prevention & control , Influenza, Human/economics , Aged, 80 and over , Adjuvants, Immunologic/economics , Adjuvants, Immunologic/administration & dosage , Male , Female , Quality-Adjusted Life Years
3.
BMC Vet Res ; 20(1): 173, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702665

ABSTRACT

Strangles is a highly contagious disease of the equine upper respiratory tract caused by Streptococcus equi subspecies. Streptococcus equi subsp. equi (S. equi) and Streptococcus equi subsp. zooepidemicus (S. zooepidemicus) was isolated, as local, hot, and field strains, from horses clinically suffering from respiratory distress. The isolated Streptococci were identified using bacteriological and molecular techniques. Four formulations of inactivated S. equi vaccines were developed and evaluated. The first formulation was prepared using the S. equi isolates, adjuvanted with MONTANIDE GEL adjuvant, while the second formulation was adjuvanted with MONTANIDE ISA-70 adjuvant. The other 2 formulations were inactivated combined vaccines prepared from both S. equi and S. zooepidemicus isolates. The 3rd formulation was the combined isolates adjuvanted with MONTANIDE GEL while the 4th formulation was the combined isolates adjuvanted with MONTANIDE ISA-70. The developed vaccines' physical properties, purity, sterility, safety, and potency were ensured. The immunizing efficacy was determined in isogenic BALB/c mice and white New Zealand rabbits using the passive hemagglutination test. Also, the antibodies' titer of the combined S. equi and S. zooepidemicus vaccine adjuvanted with MONTANIDE ISA-70 in foals was tracked using an indirect enzyme-linked immunosorbent assay. The protective efficacy of the developed vaccines was determined using a challenge test in both laboratory and field animal models, where a 75% protection rate was achieved. The combined vaccine proved to be more efficacious than the monovalent vaccine. Also, the MONTANIDE ISA-70 adjuvant provided significant protective efficacy than the MONTANIDE GEL. The current work is introducing a very promising mitigative and strategic controlling solution for strangles.


Subject(s)
Horse Diseases , Mice, Inbred BALB C , Streptococcal Infections , Streptococcal Vaccines , Streptococcus equi , Streptococcus , Animals , Streptococcus equi/immunology , Horses , Rabbits , Streptococcal Infections/veterinary , Streptococcal Infections/prevention & control , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Mice , Horse Diseases/prevention & control , Horse Diseases/microbiology , Horse Diseases/immunology , Streptococcal Vaccines/immunology , Streptococcal Vaccines/administration & dosage , Female , Antibodies, Bacterial/blood , Adjuvants, Immunologic/administration & dosage , Vaccines, Inactivated/immunology
4.
Hum Vaccin Immunother ; 20(1): 2346963, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38745461

ABSTRACT

COVID-19, caused by SARS-CoV-2, and meningococcal disease, caused by Neisseria meningitidis, are relevant infectious diseases, preventable through vaccination. Outer membrane vesicles (OMVs), released from Gram-negative bacteria, such as N. meningitidis, present adjuvant characteristics and may confer protection against meningococcal disease. Here, we evaluated in mice the humoral and cellular immune response to different doses of receptor binding domain (RBD) of SARS-CoV-2 adjuvanted by N. meningitidis C:2a:P1.5 OMVs and aluminum hydroxide, as a combined preparation for these pathogens. The immunization induced IgG antibodies of high avidity for RBD and OMVs, besides IgG that recognized the Omicron BA.2 variant of SARS-CoV-2 with intermediary avidity. Cellular immunity showed IFN-γ and IL-4 secretion in response to RBD and OMV stimuli, demonstrating immunologic memory and a mixed Th1/Th2 response. Offspring presented transferred IgG of similar levels and avidity as their mothers. Humoral immunity did not point to the superiority of any RBD dose, but the group immunized with a lower antigenic dose (0.5 µg) had the better cellular response. Overall, OMVs enhanced RBD immunogenicity and conferred an immune response directed to N. meningitidis too.


Subject(s)
Antibodies, Viral , COVID-19 , Immunoglobulin G , Neisseria meningitidis , SARS-CoV-2 , Animals , Mice , Immunoglobulin G/blood , Neisseria meningitidis/immunology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Adjuvants, Immunologic/administration & dosage , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunity, Cellular , Immunity, Humoral , Mice, Inbred BALB C , Meningococcal Infections/prevention & control , Meningococcal Infections/immunology , Spike Glycoprotein, Coronavirus/immunology , Adjuvants, Vaccine/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Hydroxide/immunology , Immunization/methods , Antibody Affinity , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Meningococcal Vaccines/immunology , Meningococcal Vaccines/administration & dosage , Immunologic Memory , Th1 Cells/immunology
5.
Sci Rep ; 14(1): 11101, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38750098

ABSTRACT

Mucosal immunity plays a major role not only in the prevention but probably also in the outcomes of COVID-19. An enhanced production of secretory immunoglobulin A (sIgA) might contribute to the activation of the immune response mechanisms. To assess the levels of sIgA produced by epithelial cells in the nasal and pharyngeal mucosa and those measured in salivary gland secretions and to study the course of COVID-19 following the combined scheme of intranasal and subcutaneous administration of a bacteria-based immunostimulant agent. This study included 69 patients, aged between 18 and 60, who had moderate COVID-19 infection. They were divided into two groups: Group 1 (control group) included 39 patients who received only background therapy, and Group 2 was made up of 30 patients who received background therapy in combination with the Immunovac VP4 vaccine, a bacteria-based immunostimulant agent, which was given for 11 days starting from the day of admission to hospital. The levels of sIgA were measured by ELISA in epithelial, nasal and pharyngeal swabs, and salivary gland secretions at baseline and on days 14 and 30. The combined scheme of intranasal and subcutaneous administration of the Immunovac VP4 vaccine in the complex therapy of patients with COVID-19 is accompanied by increased synthesis of sIgA in nasal and pharyngeal swabs, more intense decrease in the level of C-reactive protein (CRP) and reduction in the duration of fever and length of hospitalization compared to the control group. Prescribing a immunostimulant agent containing bacterial ligands in complex therapy for COVID-19 patients helps to enhance mucosal immunity and improves the course of the disease.


Subject(s)
Adjuvants, Immunologic , COVID-19 , Immunoglobulin A, Secretory , SARS-CoV-2 , Humans , Immunoglobulin A, Secretory/immunology , COVID-19/immunology , Female , Adult , Male , Middle Aged , SARS-CoV-2/immunology , Adjuvants, Immunologic/administration & dosage , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunity, Mucosal/drug effects , Young Adult , Adolescent , Administration, Intranasal
6.
Hum Vaccin Immunother ; 20(1): 2351664, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38757508

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is a lethal beta-coronavirus that emerged in 2012. The virus is part of the WHO blueprint priority list with a concerning fatality rate of 35%. Scientific efforts are ongoing for the development of vaccines, anti-viral and biotherapeutics, which are majorly directed toward the structural spike protein. However, the ongoing effort is challenging due to conformational instability of the spike protein and the evasion strategy posed by the MERS-CoV. In this study, we have expressed and purified the MERS-CoV pre-fusion spike protein in the Expi293F mammalian expression system. The purified protein was extensively characterized for its biochemical and biophysical properties. Thermal stability analysis showed a melting temperature of 58°C and the protein resisted major structural changes at elevated temperature as revealed by fluorescence spectroscopy and circular dichroism. Immunological assessment of the MERS-CoV spike immunogen in BALB/c mice with AddaVaxTM and Imject alum adjuvants showed elicitation of high titer antibody responses but a more balanced Th1/Th2 response with AddaVaxTM squalene like adjuvant. Together, our results suggest the formation of higher-order trimeric pre-fusion MERS-CoV spike proteins, which were able to induce robust immune responses. The comprehensive characterization of MERS-CoV spike protein warrants a better understanding of MERS spike protein and future vaccine development efforts.


Subject(s)
Antibodies, Viral , Mice, Inbred BALB C , Middle East Respiratory Syndrome Coronavirus , Spike Glycoprotein, Coronavirus , Viral Vaccines , Middle East Respiratory Syndrome Coronavirus/immunology , Animals , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Mice , Female , Coronavirus Infections/prevention & control , Coronavirus Infections/immunology , Immunogenicity, Vaccine , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine , Humans
7.
Nat Commun ; 15(1): 3738, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702297

ABSTRACT

Whole virus-based inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide have been critical to the COVID-19 pandemic response. Although these vaccines are protective against homologous coronavirus infection, the emergence of novel variants and the presence of large zoonotic reservoirs harboring novel heterologous coronaviruses provide significant opportunities for vaccine breakthrough, which raises the risk of adverse outcomes like vaccine-associated enhanced respiratory disease. Here, we use a female mouse model of coronavirus disease to evaluate inactivated vaccine performance against either homologous challenge with SARS-CoV-2 or heterologous challenge with a bat-derived coronavirus that represents a potential emerging disease threat. We show that inactivated SARS-CoV-2 vaccines adjuvanted with aluminum hydroxide can cause enhanced respiratory disease during heterologous infection, while use of an alternative adjuvant does not drive disease and promotes heterologous viral clearance. In this work, we highlight the impact of adjuvant selection on inactivated vaccine safety and efficacy against heterologous coronavirus infection.


Subject(s)
Aluminum Hydroxide , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccines, Inactivated , Animals , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Female , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Mice , Vaccines, Inactivated/immunology , SARS-CoV-2/immunology , Aluminum Hydroxide/administration & dosage , Disease Models, Animal , Adjuvants, Immunologic/administration & dosage , Adjuvants, Vaccine , Antibodies, Viral/immunology , Mice, Inbred BALB C , Humans , Severe acute respiratory syndrome-related coronavirus/immunology
8.
Front Immunol ; 15: 1370564, 2024.
Article in English | MEDLINE | ID: mdl-38711520

ABSTRACT

There are considerable avenues through which currently licensed influenza vaccines could be optimized. We tested influenza vaccination in a mouse model with two adjuvants: Sendai virus-derived defective interfering (SDI) RNA, a RIG-I agonist; and an amphiphilic imidazoquinoline (IMDQ-PEG-Chol), a TLR7/8 agonist. The negatively charged SDI RNA was formulated into lipid nanoparticles (LNPs) facilitating direct delivery of SDI RNA to the cytosol, where RIG-I sensing induces inflammatory and type I interferon responses. We previously tested SDI RNA and IMDQ-PEG-Chol as standalone and combination adjuvants for influenza and SARS-CoV-2 vaccines. Here, we tested two different ionizable lipids, K-Ac7-Dsa and S-Ac7-Dog, for LNP formulations. The LNPs were incorporated with SDI RNA to determine its potential as a combination adjuvant with IMDQ-PEG-Chol by evaluating the host immune response to vaccination and infection in immunized BALB/c mice. Adjuvanticity of IMDQ-PEG-Chol with and without empty or SDI-loaded LNPs was validated with quadrivalent inactivated influenza vaccine (QIV), showing robust induction of antibody titers and T-cell responses. Depending on the adjuvant combination and LNP formulation, humoral and cellular vaccine responses could be tailored towards type 1 or type 2 host responses with specific cytokine profiles that correlated with the protective responses to viral infection. The extent of protection conferred by different vaccine/LNP/adjuvant combinations was tested by challenging mice with a vaccine-matched strain of influenza A virus A/Singapore/gp1908/2015 IVR-180 (H1N1). Groups that received either LNP formulated with SDI or IMDQ-PEG-Chol, or both, showed very low levels of viral replication in their lungs at 5 days post-infection (DPI). These studies provide evidence that the combination of vaccines with LNPs and/or adjuvants promote antigen-specific cellular responses that can contribute to protection upon infection. Interestingly, we observed differences in humoral and cellular responses to vaccination between different groups receiving K-Ac7-Dsa or S-Ac7-Dog lipids in LNP formulations. The differences were also reflected in inflammatory responses in lungs of vaccinated animals to infection, depending on LNP formulations. Therefore, this study suggests that the composition of the LNPs, particularly the ionizable lipid, plays an important role in inducing inflammatory responses in vivo, which is important for vaccine safety and to prevent adverse effects upon viral exposure.


Subject(s)
Adjuvants, Immunologic , Influenza Vaccines , Liposomes , Mice, Inbred BALB C , Nanoparticles , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Adjuvants, Immunologic/administration & dosage , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Female , Lipids , Vaccination/methods , Adjuvants, Vaccine , Antibodies, Viral/blood , Antibodies, Viral/immunology , Disease Models, Animal , Sendai virus/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology
9.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729159

ABSTRACT

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes , Chitin , Membrane Proteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Signal Transduction , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Signal Transduction/drug effects , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Neoplasms/immunology , Neoplasms/therapy
10.
Front Immunol ; 15: 1354710, 2024.
Article in English | MEDLINE | ID: mdl-38726010

ABSTRACT

Cancer vaccines are gaining ground as immunotherapy options. We have previously demonstrated in cutaneous melanoma (CM) patients that adjuvant treatment with VACCIMEL, a mixture of four irradiated CM cell lines co-adjuvanted with BCG and GM-CSF, increases the cellular immune response to melanocyte differentiation antigens, cancer-testis antigens and neoantigens, with respect to basal levels. On the other hand, it is also known that treatment with anti-PD-1 monoclonal antibodies (MAbs), acting on pre-existing tumor-reactive lymphocytes, induces clinical responses in CM patients, albeit in a fraction of treated patients. A combination of both treatments would appear therefore desirable. In this paper, we describe CM patients who, having progressed even years after vaccination, were treated with anti-PD-1 MAbs. In 5/5 of such progressor patients, complete responses were obtained which lasted between 3 and 65+ months. Three of the patients remain disease-free and two recurred. One of the patients passed away after a recurrence of brain metastases. We suggest that clonally expanded reactive lymphocytes induced by VACCIMEL partially remain as memory cells, which may be recalled after tumor recurrence and may foster ulterior activity of anti-PD-1 MAbs.


Subject(s)
Cancer Vaccines , Melanoma , Programmed Cell Death 1 Receptor , Skin Neoplasms , Humans , Melanoma/immunology , Melanoma/therapy , Melanoma/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Skin Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cancer Vaccines/administration & dosage , Male , Female , Middle Aged , Aged , Immune Checkpoint Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Melanoma, Cutaneous Malignant , Treatment Outcome , Adjuvants, Immunologic/therapeutic use , Adjuvants, Immunologic/administration & dosage
11.
Lancet Rheumatol ; 6(6): e352-e360, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38710192

ABSTRACT

BACKGROUND: The adjuvanted herpes zoster subunit vaccine has shown good efficacy and safety in the general population. However, its effectiveness has not been comprehensively assessed in patients with systemic lupus erythematosus (SLE). This study aimed to evaluate the immunogenicity and safety of the adjuvanted herpes zoster subunit vaccine in patients with SLE. METHODS: This single-centre, randomised, double-blind, placebo-controlled, trial was done at the rheumatology outpatient clinic at Seoul National University Hospital, South Korea. Patients (aged ≥19 years) with clinically stable SLE and previous exposure (≥4 weeks) to immunosuppressive drugs were randomly assigned (4:1) via a central interactive web response system to receive herpes zoster subunit vaccine or placebo (0·5 mL intramuscular injection) at weeks 0 and 8. Investigators and participants were masked to intervention and group assignment. Anti-glycoprotein E antibody titres and glycoprotein E-specific cell-mediated vaccine responses were evaluated at baseline and at week 8 after the first dose, and at week 4, week 26, and week 52 after the second dose using enzyme-linked immunosorbent assay and flow cytometry, respectively. Reactogenicity, SLE disease activity, including Systemic Lupus Erythematosus Disease Activity Index 2000 and British Isles Lupus Assessment Group-flare rate, were examined. The primary outcome was the proportion of patients with a positive humoral vaccine response 4 weeks after the second dose. The primary and safety analyses were done in a modified intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT06001606. FINDINGS: Between June 14, and July 19, 2023, 65 patients with SLE were enrolled, of whom 52 were randomly assigned to the herpes zoster subunit vaccine and 13 to placebo. 49 patients in the vaccine group and 11 patients in the placebo group were included in the modified intention-to-treat population. 56 (93%) of 60 patients were women and four (7%) were men. Mean age was 48·7 years (SD 11·4). The proportion of participants with a humoral vaccine response at 4 weeks after the second dose was significantly higher in the vaccine group (48 [98%] of 49 participants) than the placebo group (none [0%] of 11 patients; p<0·0001). More patients in the vaccine group than placebo group reported injection site reactions (42 patients vs two patients), fever (ten vs none), and fatigue (26 vs two). There were no differences in Systemic Lupus Erythematosus Disease Activity Index 2000 and British Isles Lupus Assessment Group-flare rates between the groups. There were no treatment-related deaths. INTERPRETATION: The herpes zoster subunit vaccine induces humoral and cellular immunity against herpes zoster with a good safety profile in patients with SLE. A larger study is warranted to assess the efficacy of vaccines to prevent herpes zoster in patients with SLE. FUNDING: Ministry of Science and ICT, The Government of the Republic of Korea.


Subject(s)
Herpes Zoster Vaccine , Lupus Erythematosus, Systemic , Vaccines, Subunit , Humans , Lupus Erythematosus, Systemic/immunology , Female , Double-Blind Method , Male , Herpes Zoster Vaccine/immunology , Herpes Zoster Vaccine/administration & dosage , Herpes Zoster Vaccine/adverse effects , Republic of Korea/epidemiology , Adult , Middle Aged , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/therapeutic use , Herpes Zoster/prevention & control , Herpes Zoster/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Immunogenicity, Vaccine , Antibodies, Viral/blood
12.
Front Immunol ; 15: 1383476, 2024.
Article in English | MEDLINE | ID: mdl-38799439

ABSTRACT

None of the typhoid Vi Polysaccharide (ViPS) subunit vaccines incorporate adjuvants, and the immunogenicity of ViPS vaccines (e.g. Typbar TCV® and Typhim Vi®) is in part due to associated TLR4 ligands such as endotoxin present in these vaccines. Since endotoxin content in vaccines is variable and kept very low due to inherent toxicity, it was hypothesized that incorporating a defined amount of a non-toxic TLR4-ligand such as monophosphoryl lipid A in ViPS vaccines would improve their immunogenicity. To test this hypothesis, a monophosphoryl lipid A-based adjuvant formulation named Turbo was developed. Admixing Turbo with Typbar TCV® (ViPS-conjugated to tetanus toxoid) increased the levels of anti-ViPS IgM, IgG1, IgG2b, IgG2a/c, and IgG3 in inbred and outbred mice. In infant mice, a single immunization with Turbo adjuvanted Typbar TCV® resulted in a significantly increased and durable IgG response and improved the control of bacterial burden compared to mice immunized without Turbo. Similarly, when adjuvanted with Turbo, the antibody response and control of bacteremia were also improved in mice immunized with Typhim Vi®, an unconjugated vaccine. The immunogenicity of unconjugated ViPS is inefficient in young mice and is lost in adult mice when immunostimulatory ligands in ViPS are removed. Nevertheless, when adjuvanted with Turbo, poorly immunogenic ViPS induced a robust IgG response in young and adult mice, and this was observed even under antigen-limiting conditions. These data suggest that incorporation of Turbo as an adjuvant will make typhoid vaccines more immunogenic regardless of their intrinsic immunogenicity or conjugation status and maximize the efficacy across all ages.


Subject(s)
Adjuvants, Immunologic , Antibodies, Bacterial , Lipid A , Toll-Like Receptor 4 , Typhoid Fever , Typhoid-Paratyphoid Vaccines , Vaccines, Subunit , Animals , Typhoid-Paratyphoid Vaccines/immunology , Typhoid-Paratyphoid Vaccines/administration & dosage , Mice , Toll-Like Receptor 4/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Adjuvants, Immunologic/administration & dosage , Lipid A/analogs & derivatives , Lipid A/immunology , Typhoid Fever/prevention & control , Typhoid Fever/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Female , Ligands , Polysaccharides, Bacterial/immunology , Immunogenicity, Vaccine , Adjuvants, Vaccine , Salmonella typhi/immunology , Mice, Inbred BALB C
13.
N Engl J Med ; 390(15): 1359-1371, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38631003

ABSTRACT

BACKGROUND: Adjuvant pembrolizumab therapy after surgery for renal-cell carcinoma was approved on the basis of a significant improvement in disease-free survival in the KEYNOTE-564 trial. Whether the results regarding overall survival from the third prespecified interim analysis of the trial would also favor pembrolizumab was uncertain. METHODS: In this phase 3, double-blind, placebo-controlled trial, we randomly assigned (in a 1:1 ratio) participants with clear-cell renal-cell carcinoma who had an increased risk of recurrence after surgery to receive pembrolizumab (at a dose of 200 mg) or placebo every 3 weeks for up to 17 cycles (approximately 1 year) or until recurrence, the occurrence of unacceptable toxic effects, or withdrawal of consent. A significant improvement in disease-free survival according to investigator assessment (the primary end point) was shown previously. Overall survival was the key secondary end point. Safety was a secondary end point. RESULTS: A total of 496 participants were assigned to receive pembrolizumab and 498 to receive placebo. As of September 15, 2023, the median follow-up was 57.2 months. The disease-free survival benefit was consistent with that in previous analyses (hazard ratio for recurrence or death, 0.72; 95% confidence interval [CI], 0.59 to 0.87). A significant improvement in overall survival was observed with pembrolizumab as compared with placebo (hazard ratio for death, 0.62; 95% CI, 0.44 to 0.87; P = 0.005). The estimated overall survival at 48 months was 91.2% in the pembrolizumab group, as compared with 86.0% in the placebo group; the benefit was consistent across key subgroups. Pembrolizumab was associated with a higher incidence of serious adverse events of any cause (20.7%, vs. 11.5% with placebo) and of grade 3 or 4 adverse events related to pembrolizumab or placebo (18.6% vs. 1.2%). No deaths were attributed to pembrolizumab therapy. CONCLUSIONS: Adjuvant pembrolizumab was associated with a significant and clinically meaningful improvement in overall survival, as compared with placebo, among participants with clear-cell renal-cell carcinoma at increased risk for recurrence after surgery. (Funded by Merck Sharp and Dohme, a subsidiary of Merck; KEYNOTE-564 ClinicalTrials.gov number, NCT03142334.).


Subject(s)
Antineoplastic Agents, Immunological , Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/surgery , Double-Blind Method , Kidney Neoplasms/drug therapy , Kidney Neoplasms/mortality , Kidney Neoplasms/surgery , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Disease-Free Survival , Combined Modality Therapy , Survival Analysis
14.
Influenza Other Respir Viruses ; 18(4): e13288, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38644564

ABSTRACT

BACKGROUND: Adults ≥ 65 years of age have suboptimal influenza vaccination responses compared to younger adults due to age-related immunosenescence. Two vaccines were specifically developed to enhance protection: MF59-adjuvanted trivalent influenza vaccine (aIIV3) and high-dose egg-based trivalent influenza vaccine (HD-IIV3e). METHODS: In a retrospective cohort study conducted using US electronic medical records linked to claims data during the 2019-2020 influenza season, we compared the relative vaccine effectiveness (rVE) of aIIV3 with HD-IIV3e and a standard-dose non-adjuvanted egg-based quadrivalent inactivated influenza vaccine (IIV4e) for the prevention of cardiorespiratory hospitalizations, including influenza hospitalizations. We evaluated outcomes in the "any" diagnosis position and the "admitting" position on the claim. A doubly robust methodology using inverse probability of treatment weighting and logistic regression was used to adjust for covariate imbalance. rVE was calculated as 100 * (1 - ORadjusted). RESULTS: The study included 4,299,594 adults ≥ 65 years of age who received aIIV3, HD-IIV3e, or IIV4e. Overall, aIIV3 was associated with lower proportions of cardiorespiratory hospitalizations with diagnoses in any position compared to HD-IIV3e (rVE = 3.9% [95% CI, 2.7-5.0]) or IIV4e (9.0% [95% CI, 7.7-10.4]). Specifically, aIIV3 was more effective compared with HD-IIV3e and IIV4e in preventing influenza hospitalizations (HD-IIV3e: 9.7% [95% CI, 1.9-17.0]; IIV4e: 25.3% [95% CI, 17.7-32.2]). Consistent trends were observed for admitting diagnoses. CONCLUSION: Relative to both HD-IIV3e and IIV4e, aIIV3 provided improved protection from cardiorespiratory or influenza hospitalizations.


Subject(s)
Adjuvants, Immunologic , Hospitalization , Influenza Vaccines , Influenza, Human , Polysorbates , Squalene , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Aged , Hospitalization/statistics & numerical data , Male , Retrospective Studies , Female , Squalene/administration & dosage , Polysorbates/administration & dosage , Middle Aged , United States/epidemiology , Adjuvants, Immunologic/administration & dosage , Aged, 80 and over , Vaccine Efficacy , Seasons , Adult , Vaccination/statistics & numerical data
15.
Front Immunol ; 15: 1387534, 2024.
Article in English | MEDLINE | ID: mdl-38650936

ABSTRACT

For several years, we have been committed to exploring the potential of Bordetella pertussis-derived outer membrane vesicles (OMVBp) as a promising third-generation vaccine against the reemerging pertussis disease. The results of our preclinical trials not only confirm its protective capacity against B. pertussis infection but also set the stage for forthcoming human clinical trials. This study delves into the examination of OMVBp as an adjuvant. To accomplish this objective, we implemented a two-dose murine schedule to evaluate the specific immune response induced by formulations containing OMVBp combined with 3 heterologous immunogens: Tetanus toxoid (T), Diphtheria toxoid (D), and the SARS-CoV-2 Spike protein (S). The specific levels of IgG, IgG1, and IgG2a triggered by the different tested formulations were evaluated using ELISA in dose-response assays for OMVBp and the immunogens at varying levels. These assays demonstrated that OMVBp exhibits adjuvant properties even at the low concentration employed (1.5 µg of protein per dose). As this effect was notably enhanced at medium (3 µg) and high concentrations (6 µg), we chose the medium concentration to determine the minimum immunogen dose at which the OMV adjuvant properties are significantly evident. These assays demonstrated that OMVBp exhibits adjuvant properties even at the lowest concentration tested for each immunogen. In the presence of OMVBp, specific IgG levels detected for the lowest amount of antigen tested increased by 2.5 to 10 fold compared to those found in animals immunized with formulations containing adjuvant-free antigens (p<0.0001). When assessing the adjuvant properties of OMVBp compared to the widely recognized adjuvant alum, we detected similar levels of specific IgG against D, T and S for both adjuvants. Experiments with OMVs derived from E. coli (OMVE.coli) reaffirmed that the adjuvant properties of OMVs extend across different bacterial species. Nonetheless, it's crucial to highlight that OMVBp notably skewed the immune response towards a Th1 profile (p<0.05). These collective findings emphasize the dual role of OMVBp as both an adjuvant and modulator of the immune response, positioning it favorably for incorporation into combined vaccine formulations.


Subject(s)
Adjuvants, Immunologic , Bordetella pertussis , Immunoglobulin G , Th1 Cells , Whooping Cough , Bordetella pertussis/immunology , Animals , Adjuvants, Immunologic/administration & dosage , Mice , Th1 Cells/immunology , Whooping Cough/immunology , Whooping Cough/prevention & control , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Pertussis Vaccine/immunology , Pertussis Vaccine/administration & dosage , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Spike Glycoprotein, Coronavirus/immunology , Mice, Inbred BALB C , SARS-CoV-2/immunology , Bacterial Outer Membrane Proteins/immunology , Humans , COVID-19/immunology , COVID-19/prevention & control , Tetanus Toxoid/immunology
16.
Hum Vaccin Immunother ; 20(1): 2337987, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38658133

ABSTRACT

There is a growing interest in development of novel vaccines against respiratory tract infections, due to COVID-19 pandemic. Here, we examined mucosal adjuvanticity and the mucosal booster effect of membrane vesicles (MVs) of a novel probiotic E. coli derivative lacking both flagella and potentially carcinogenic colibactin (ΔflhDΔclbP). ΔflhDΔclbP-derived MVs showed rather strong mucosal adjuvanticity as compared to those of a single flagellar mutant strain (ΔflhD-MVs). In addition, glycoengineered ΔflhDΔclbP-MVs displaying serotype-14 pneumococcal capsular polysaccharide (CPS14+MVs) were well-characterized based on biological and physicochemical parameters. Subcutaneous (SC) and intranasal (IN) booster effects of CPS14+MVs on systemic and mucosal immunity were evaluated in mice that have already been subcutaneously prime-immunized with the same MVs. With a two-dose regimen, an IN boost (SC-IN) elicited stronger IgA responses than homologous prime-boost immunization (SC-SC). With a three-dose regimen, serum IgG levels were comparable among all tested regimens. Homologous immunization (SC-SC-SC) elicited the highest IgM responses among all regimens tested, whereas SC-SC-SC failed to elicit IgA responses in blood and saliva. Furthermore, serum IgA and salivary SIgA levels were increased with an increased number of IN doses administrated. Notably, SC-IN-IN induced not only robust IgG response, but also the highest IgA response in both serum and saliva among the groups. The present findings suggest the potential of a heterologous three-dose administration for building both systemic and mucosal immunity, e.g. an SC-IN-IN vaccine regimen could be beneficial. Another important observation was abundant packaging of colibactin in MVs, suggesting increased applicability of ΔflhDΔclbP-MVs in the context of vaccine safety.


Subject(s)
Adjuvants, Immunologic , Escherichia coli , Immunity, Mucosal , Immunization, Secondary , Mice, Inbred BALB C , Polyketides , Probiotics , Animals , Mice , Probiotics/administration & dosage , Escherichia coli/immunology , Immunization, Secondary/methods , Female , Adjuvants, Immunologic/administration & dosage , Immunoglobulin A , Peptides/immunology , Administration, Intranasal , Immunoglobulin G/blood , Immunoglobulin M , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage
17.
Int J Pharm ; 656: 124076, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38569976

ABSTRACT

Vaccines represent a pivotal health advancement for preventing infection. However, because carrier systems with repeated administration can invoke carrier-targeted immune responses that diminish subsequent immune responses (e.g., PEG antibodies), there is a continual need to develop novel vaccine platforms. Zinc carnosine microparticles (ZnCar MPs), which are composed of a one-dimensional coordination polymer formed between carnosine and the metal ion zinc, have exhibited efficacy in inducing an immune response against influenza. However, ZnCar MPs' limited suspendability hinders clinical application. In this study, we address this issue by mixing mannan, a polysaccharide derived from yeast, with ZnCar MPs. We show that the addition of mannan increases the suspendability of this promising vaccine formulation. Additionally, since mannan is an adjuvant, we illustrate that the addition of mannan increases the antibody response and T cell response when mixed with ZnCar MPs. Mice vaccinated with mannan + OVA/ZnCar MPs had elevated serum IgG and IgG1 levels in comparison to vaccination without mannan. Moreover, in the mannan + OVA/ZnCar MPs vaccinated group, mucosal washes demonstrated increased IgG, IgG1, and IgG2c titers, and antigen recall assays showed enhanced IFN-γ production in response to MHC-I and MHC-II immunodominant peptide restimulation, compared to the vaccination without mannan. These findings suggest that the use of mannan mixed with ZnCar MPs holds potential for subunit vaccination and its improved suspendability further promotes clinical translation.


Subject(s)
Carnosine , Mannans , Vaccines, Subunit , Zinc , Mannans/chemistry , Mannans/administration & dosage , Mannans/immunology , Animals , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Zinc/chemistry , Zinc/administration & dosage , Carnosine/administration & dosage , Carnosine/chemistry , Female , Immunoglobulin G/blood , Mice , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Ovalbumin/immunology , Ovalbumin/administration & dosage , Mice, Inbred C57BL , Polymers/chemistry , Polymers/administration & dosage , Mice, Inbred BALB C , Drug Carriers/chemistry
18.
BMC Med ; 22(1): 170, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649867

ABSTRACT

BACKGROUND: The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS: In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS: Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS: R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION: The trial is registered with ClinicalTrials.gov under identifier NCT04862416.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Membrane Glycoproteins , Plasmodium falciparum , Protozoan Proteins , Adolescent , Adult , Animals , Female , Humans , Male , Middle Aged , Young Adult , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Antibodies, Protozoan , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/immunology , Netherlands , Plasmodium falciparum/immunology , Protozoan Proteins/immunology
19.
Expert Rev Vaccines ; 23(1): 535-545, 2024.
Article in English | MEDLINE | ID: mdl-38664959

ABSTRACT

INTRODUCTION: Zebrafishes represent a proven model for human diseases and systems biology, exhibiting physiological and genetic similarities and having innate and adaptive immune systems. However, they are underexplored for human vaccinology, vaccine development, and testing. Here we summarize gaps and challenges. AREAS COVERED: Zebrafish models have four potential applications: 1) Vaccine safety: The past successes in using zebrafishes to test xenobiotics could extend to vaccine and adjuvant formulations for general safety or target organs due to the zebrafish embryos' optical transparency. 2) Innate immunity: The zebrafish offers refined ways to examine vaccine effects through signaling via Toll-like or NOD-like receptors in zebrafish myeloid cells. 3) Adaptive immunity: Zebrafishes produce IgM, IgD,and two IgZ immunoglobulins, but these are understudied, due to a lack of immunological reagents for challenge studies. 4) Systems vaccinology: Due to the availability of a well-referenced zebrafish genome, transcriptome, proteome, and epigenome, this model offers potential here. EXPERT OPINION: It remains unproven whether zebrafishes can be employed for testing and developing human vaccines. We are still at the hypothesis-generating stage, although it is possible to begin outlining experiments for this purpose. Through transgenic manipulation, zebrafish models could offer new paths for shaping animal models and systems vaccinology.


Subject(s)
Adaptive Immunity , Adjuvants, Immunologic , Immunity, Innate , Models, Animal , Vaccine Development , Vaccines , Zebrafish , Zebrafish/immunology , Animals , Adjuvants, Immunologic/administration & dosage , Humans , Vaccines/immunology , Vaccines/administration & dosage , Vaccinology/methods
20.
Colloids Surf B Biointerfaces ; 238: 113920, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38688058

ABSTRACT

Mucosal immunization is a powerful weapon against viral infection. In this paper, large pore mesoporous silica nanoparticles (LMSN) with different particle sizes were synthesized for loading influenza split vaccine (SV) to explore the effect of nanoparticle sizes on mucosal immunization and adjuvant efficacy. Interestingly, it was found that among the three particle sizes of nanoparticles, only LMSN-M with around 250 nm could significantly enhance the mucosal immune effect of SV, possessing adjuvant effect. The results indicated that particle size affected the adjuvant effect of LMSN. There was no apparent difference in vaccine loading capacity of LMSN with different particle sizes, but the release of SV depended on the pore length of LMSN. The adjuvant effect of LMSN-M was attributed to its higher cellular uptake performance, intestine absorption and transport efficiency, and the ability to stimulate the maturation of dendritic cells. Simultaneously, compared with LMSN-S and LMSN-L, the more retention of LMSN-M in mesenteric lymph nodes increased the chance of interaction between vaccine and immune system, resulting in the enhanced immunity. This is the first time to study the impact of particle size of LMSN adjuvant on improving mucosal immunity of oral influenza vaccine, and the present work provides a scientific reference for adjuvant design of oral vaccine.


Subject(s)
Influenza Vaccines , Nanoparticles , Particle Size , Silicon Dioxide , Silicon Dioxide/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/administration & dosage , Nanoparticles/chemistry , Animals , Administration, Oral , Porosity , Mice , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Mice, Inbred BALB C , Female , Immunity, Mucosal/drug effects , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...