Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Molecules ; 26(11)2021 May 28.
Article in English | MEDLINE | ID: mdl-34071302

ABSTRACT

The 3-hydroxyquinone derivative of the non-psychotrophic phytocannabinoid cannabigerol, so-called VCE-003.2, and some other derivatives have been recently investigated for neuroprotective properties in experimental models of Parkinson's disease (PD) in mice. The pharmacological effects in those models were related to the activity on the peroxisome proliferator-activated receptor-γ (PPAR-γ) and possibly other pathways. In the present study, we investigated VCE-004.8 (formulated as EHP-101 for oral administration), the 3-hydroxyquinone derivative of cannabidiol (CBD), with agonist activity at the cannabinoid receptor type-2 (CB2) receptor in addition to its activity at the PPAR-γ receptor. Studies were conducted in both in vivo (lesioned-mice) and in vitro (SH-SY5Y cells) models using the classic parkinsonian neurotoxin 6-hydroxydopamine (6-OHDA). Our data confirmed that the treatment with VCE-004.8 partially reduced the loss of tyrosine hydroxylase (TH)-positive neurons measured in the substantia nigra of 6-OHDA-lesioned mice, in parallel with an almost complete reversal of the astroglial (GFAP) and microglial (CD68) reactivity occurring in this structure. Such neuroprotective effects attenuated the motor deficiencies shown by 6-OHDA-lesioned mice in the cylinder rearing test, but not in the pole test. Next, we explored the mechanism involved in the beneficial effect of VCE-004.8 in vivo, by analyzing cell survival in cultured SH-SY5Y cells exposed to 6-OHDA. We found an important cytoprotective effect of VCE-004.8 at a concentration of 10 µM, which was completely reversed by the addition of antagonists, T0070907 and SR144528, aimed at blocking PPAR-γ and CB2 receptors, respectively. The treatment with T0070907 alone only caused a partial reversal, whereas SR144528 alone had no effect, indicating a major contribution of PPAR-γ receptors in the cytoprotective effect of VCE-004.8 at 10 µM. In summary, our data confirmed the neuroprotective potential of VCE-004.8 in 6-OHDA-lesioned mice, and in vitro studies confirmed a greater relevance for PPAR-γ receptors rather than CB2 receptors in these effects.


Subject(s)
Cannabidiol/chemistry , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Quinones/chemistry , Administration, Oral , Animals , Benzamides/pharmacology , Camphanes/pharmacology , Cannabinoids/chemistry , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuroprotection , Oxidopamine/chemistry , PPAR gamma/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Tyrosine 3-Monooxygenase/metabolism
2.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804243

ABSTRACT

A growing body of preclinical evidence indicates that certain cannabinoids, including cannabidiol (CBD) and synthetic derivatives, may play a role in the myelinating processes and are promising small molecules to be developed as drug candidates for management of demyelinating diseases such as multiple sclerosis (MS), stroke and traumatic brain injury (TBI), which are three of the most prevalent demyelinating disorders. Thanks to the properties described for CBD and its interesting profile in humans, both the phytocannabinoid and derivatives could be considered as potential candidates for clinical use. In this review we will summarize current advances in the use of CBD and other cannabinoids as future potential treatments. While new research is accelerating the process for the generation of novel drug candidates and identification of druggable targets, the collaboration of key players such as basic researchers, clinicians and pharmaceutical companies is required to bring novel therapies to the patients.


Subject(s)
Cannabidiol/therapeutic use , Cannabinoids/therapeutic use , Cannabis/chemistry , Demyelinating Diseases/drug therapy , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Demyelinating Diseases/pathology , Humans , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Stroke/drug therapy , Stroke/pathology
3.
Mol Cell Neurosci ; 110: 103583, 2021 01.
Article in English | MEDLINE | ID: mdl-33338634

ABSTRACT

The quinone derivative of the non-psychotropic cannabinoid cannabigerol (CBG), so-called VCE-003.2, has been recently investigated for its neuroprotective properties in inflammatory models of Parkinson's disease (PD) in mice. Such potential derives from its activity at the peroxisome proliferator-activated receptor-γ (PPAR-γ). In the present study, we investigated the neuroprotective properties of VCE-003.2 against the parkinsonian neurotoxin 6-hydroxydopamine (6-OHDA), in comparison with two new CBG-related derivatives, the cannabigerolic acid quinone (CBGA-Q) and its sodium salt CBGA-Q-Salt, which, similarly to VCE-003.2, were found to be active at the PPAR-γ receptor, but not at the cannabinoid CB1 and CB2 receptors. First, we investigated their cytoprotective properties in vitro by analyzing cell survival in cultured SH-SY5Y cells exposed to 6-OHDA. We found an important cytoprotective effect of VCE-003.2 at a concentration of 20 µM, which was not reversed by the blockade of PPAR-γ receptors with GW9662, supporting its activity at an alternative site (non-sensitive to classic antagonists) in this receptor. We also found CBGA-Q and CBGA-Q-Salt being cytoprotective in this cell assay, but their effects were completely eliminated by GW9662, thus indicating that they are active at the canonical site in the PPAR-γ receptor. Then, we moved to in vivo testing using mice unilaterally lesioned with 6-OHDA. Our data confirmed that VCE-003.2 administered orally (20 mg/kg) preserved tyrosine hydroxylase (TH)-positive nigral neurons against 6-OHDA-induced damage, whereas it completely attenuated the astroglial (GFAP) and microglial (CD68) reactivity found in the substantia nigra of lesioned mice. Such neuroprotective effects caused an important recovery in the motor deficiencies displayed by 6-OHDA-lesioned mice in the pole test and the cylinder rearing test. We also investigated CBGA-Q, given orally (20 mg/kg) or intraperitoneally (10 mg/kg, i.p.), having similar benefits compared to VCE-003.2 against the loss of TH-positive nigral neurons, glial reactivity and motor defects caused by 6-OHDA. Lastly, the sodium salt of CBGA-Q, given orally (40 mg/kg) to 6-OHDA-lesioned mice, also showed benefits at behavioral and histopathological levels, but to a lower extent compared to the other two compounds. In contrast, when given i.p., CBGA-Q-Salt (10 mg/kg) was poorly active. We also analyzed the concentrations of dopamine and its metabolite DOPAC in the striatum of 6-OHDA-lesioned mice after the treatment with the different compounds, but recovery in the contents of both dopamine and DOPAC was only found after the treatment with VCE-003.2. In summary, our data confirmed the neuroprotective potential of VCE-003.2 in 6-OHDA-lesioned mice, which adds to its previous activity found in an inflammatory model of PD (LPS-lesioned mice). Additional phytocannabinoid derivatives, CBGA-Q and CBGA-Q-Salt, also afforded neuroprotection in 6-OHDA-lesioned mice, but their effects were lower compared to VCE-003.2, in particular in the case of CBGA-Q-Salt. In vitro studies confirmed the relevance of PPAR-γ receptors for these effects.


Subject(s)
Antiparkinson Agents/therapeutic use , Cannabinoids/chemistry , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Quinones/chemistry , Animals , Antiparkinson Agents/chemical synthesis , Antiparkinson Agents/pharmacology , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Parkinson Disease/etiology , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
4.
Int J Hyperthermia ; 31(1): 1-4, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25599964

ABSTRACT

We report two cases of pulmonary arterial pseudoaneurysms (PAs) following percutaneous radiofrequency ablation (PRFA). The first patient was a 74-year-old Caucasian man who was treated for a secondary location of an advanced melanoma. A computed tomography scan at 72 h after the procedure, performed for basithoracic pain, hyperthermia and haemoptysis, revealed a 17-mm PA within the ablative zone. A lobectomy was performed. The second patient was an 80-year-old white man followed up for a right apical lung adenocarcinoma. Massive haemoptysis occurred 24 h after PRFA; emergent contrast-enhanced CT and pulmonary arteriography revealed a pulmonary artery PA (20 mm diameter), which was embolised with coils. The initial clinical course was satisfactory; however, 15 days after the procedure, the patient unfortunately presented a new massive haemoptysis and died a few hours later. The long ablation duration and the multiple repositioning of the electrodes might have been risk factors for this rare and potentially lethal complication.


Subject(s)
Adenocarcinoma/therapy , Aneurysm, False/etiology , Catheter Ablation/adverse effects , Embolization, Therapeutic/adverse effects , Lung Neoplasms/therapy , Melanoma/therapy , Skin Neoplasms/therapy , Adenocarcinoma/secondary , Adenocarcinoma of Lung , Aged , Aged, 80 and over , Humans , Lung Neoplasms/secondary , Male , Melanoma/pathology , Pulmonary Artery , Skin Neoplasms/pathology
6.
Hum Vaccin Immunother ; 9(6): 1333-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23857272

ABSTRACT

Increasing the potency and supply of seasonal and pandemic influenza vaccines remains an important unmet medical need which may be effectively accomplished with adjuvanted egg- or cell culture-derived vaccines. Vaxfectin, a cationic lipid-based adjuvant with a favorable safety profile in phase 1 plasmid DNA vaccines trials, was tested in combination with seasonal split, trivalent and pandemic whole virus, monovalent influenza vaccines produced in Vero cell cultures. Comparison of hemagglutination inhibition (HI) antibody titers in Vaxfectin-adjuvanted to nonadjuvanted vaccinated mice and guinea pigs revealed 3- to 20-fold increases in antibody titers against each of the trivalent influenza virus vaccine strains and 2- to 8-fold increases in antibody titers against the monovalent H5N1 influenza virus vaccine strain. With the vaccine doses tested, comparable antibody responses were induced with formulations that were freshly prepared or refrigerated at conventional 2-8°C storage conditions for up to 6 mo. Comparison of T-cell frequencies measured by interferon-gamma ELISPOT assay between groups revealed increases of between 2- to 10-fold for each of the adjuvanted trivalent strains and up to 22-fold higher with monovalent H5N1 strain. Both trivalent and monovalent vaccines were easy to formulate with Vaxfectin by simple mixing. These preclinical data support further testing of Vaxfectin-adjuvanted Vero cell culture vaccines toward clinical studies designed to assess safety and immunogenicity of these vaccines in humans.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Influenza A virus/immunology , Influenza Vaccines/immunology , Phosphatidylethanolamines/administration & dosage , Animals , Antibodies, Viral/blood , Female , Guinea Pigs , Hemagglutination Inhibition Tests , Influenza Vaccines/administration & dosage , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
7.
J Virol ; 87(12): 6560-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23552419

ABSTRACT

DNA vaccines formulated with the cationic lipid-based adjuvant Vaxfectin induce protective immunity in macaques after intradermal (i.d.) or intramuscular (i.m.) delivery of 0.5 to 1 mg of codon-optimized DNA encoding the hemagglutinin (H) and fusion (F) proteins of measles virus (MeV). To characterize the effect of Vaxfectin at lower doses of H+F DNA, rhesus macaques were vaccinated twice with 20 µg of DNA plus Vaxfectin i.d., 100 µg of DNA plus Vaxfectin i.d., 100 µg of DNA plus Vaxfectin i.m. or 100 µg of DNA plus phosphate-buffered saline (PBS) i.m. using a needleless Biojector device. The levels of neutralizing (P = 0.036) and binding (P = 0.0001) antibodies were higher after 20 or 100 µg of DNA plus Vaxfectin than after 100 µg of DNA plus PBS. Gamma interferon (IFN-γ)-producing T cells were induced more rapidly than antibody, but were not improved with Vaxfectin. At 18 months after vaccination, monkeys were challenged with wild-type MeV. None developed rash or viremia, but all showed evidence of infection. Antibody levels increased, and IFN-γ- and interleukin-17-producing T cells, including cells specific for the nucleoprotein absent from the vaccine, were induced. At 3 months after challenge, MeV RNA was detected in the leukocytes of two monkeys. The levels of antibody peaked 2 to 4 weeks after challenge and then declined in vaccinated animals reflecting low numbers of bone marrow-resident plasma cells. Therefore, Vaxfectin was dose sparing and substantially improved the antibody response to the H+F DNA vaccine. This immune response led to protection from disease (rash/viremia) but not from infection. Antibody responses after challenge were more transient in vaccinated animals than in an unvaccinated animal.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies, Viral/blood , Antibody Formation/immunology , Hemagglutinins, Viral/immunology , Macaca mulatta/immunology , Measles Vaccine/immunology , Phosphatidylethanolamines/immunology , Vaccines, DNA/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Hemagglutinins, Viral/genetics , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Measles/immunology , Measles/prevention & control , Measles/virology , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Phosphatidylethanolamines/administration & dosage , T-Lymphocytes/immunology , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Fusion Proteins/genetics
9.
Vaccines (Basel) ; 1(4): 398-414, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-26344340

ABSTRACT

2013 marks a milestone year for plasmid DNA vaccine development as a first-in-class cytomegalovirus (CMV) DNA vaccine enters pivotal phase 3 testing. This vaccine consists of two plasmids expressing CMV antigens glycoprotein B (gB) and phosphoprotein 65 (pp65) formulated with a CRL1005 poloxamer and benzalkonium chloride (BAK) delivery system designed to enhance plasmid expression. The vaccine's planned initial indication under investigation is for prevention of CMV reactivation in CMV-seropositive (CMV⁺) recipients of an allogeneic hematopoietic stem cell transplant (HCT). A randomized, double-blind placebo-controlled phase 2 proof-of-concept study provided initial evidence of the safety of this product in CMV⁺ HCT recipients who underwent immune ablation conditioning regimens. This study revealed a significant reduction in viral load endpoints and increased frequencies of pp65-specific interferon-γ-producing T cells in vaccine recipients compared to placebo recipients. The results of this endpoint-defining trial provided the basis for defining the primary and secondary endpoints of a global phase 3 trial in HCT recipients. A case study is presented here describing the development history of this vaccine from product concept to initiation of the phase 3 trial.

10.
Hum Vaccin Immunother ; 8(11): 1595-606, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22922766

ABSTRACT

Human cytomegalovirus (CMV) establishes a lifelong persistent infection characterized by periods of latency and sporadic viral replication and is a major infectious cause of birth defects following congenital infection. Currently, no licensed vaccine is available that would prevent CMV infection. In an effort to develop a prophylactic CMV vaccine, the effects of different formulations, immunization routes and delivery devices on the immunogenicity of plasmid DNA (pDNA)-based vaccines were evaluated in rabbits and mice. Compared with PBS- and poloxamer-based formulations, significantly higher antibody responses were obtained with pDNA formulated with Vaxfectin (®) , a cationic lipid-based adjuvant. With low vaccine doses, the intradermal (ID) route resulted in higher antibody responses than obtained when the same dose was administered intramuscularly (IM). Since the IM route allowed injection of larger volumes and higher doses than could be administered at a single ID site, better antibody responses were obtained using the IM route. The needle-free injection system Biojector (®) 2000 and electroporation devices enhanced antibody responses only marginally compared with responses obtained with Vaxfectin (®) -formulated pDNA injected IM with a needle. A single-vial Vaxfectin (®) formulation was developed in a dosage form ready for use after thawing at room temperature. Finally, in a GLP-compliant repeat-dose toxicology study conducted in rabbits, single-vial Vaxfectin (®) -formulated vaccines, containing pDNA and Vaxfectin (®) up to 4.5 mg and 2 mg/injection, respectively, showed a favorable safety profile and were judged as well-tolerated. The results support further development of a Vaxfectin (®) -formulated pDNA vaccine to target congenital CMV infection.


Subject(s)
Cytomegalovirus Vaccines/immunology , Plasmids/genetics , Vaccines, DNA/therapeutic use , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Female , Injections, Intradermal , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Rabbits , Vaccines, DNA/immunology
11.
J Gen Virol ; 93(Pt 6): 1305-1315, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22398318

ABSTRACT

The herpes simplex type 2 (HSV-2) envelope glycoprotein (gD2) was evaluated as a potential antigen candidate for a plasmid DNA (pDNA)-based HSV-2 vaccine. The pDNA was formulated with Vaxfectin, a cationic lipid-based adjuvant, and tested in a murine HSV-2 lethal challenge model. gD2 was expressed as full-length (FL) and secreted (S) gD2 forms. A 0.1 µg pDNA dose was tested to distinguish treatment conditions for survival and a 100 µg pDNA dose was tested to distinguish treatment conditions for reduction in vaginal and latent HSV-2 copies. Vaxfectin-formulated gD2 pDNA significantly increased serum IgG titres and survival for both FL gD2 and S gD2 compared with gD2 pDNA alone. Mice immunized with FL gD2 formulated with Vaxfectin showed reduction in vaginal and dorsal root ganglia (DRG) HSV-2 copies. The stringency of this protection was further evaluated by testing Vaxfectin-formulated FL gD2 pDNA at a high 500 LD(50) inoculum. At this high viral challenge, the 0.1 µg dose of FL gD2 Vaxfectin-formulated pDNA yielded 80 % survival compared with no survival for FL gD2 pDNA alone. Vaxfectin-formulated FL gD2 pDNA, administered at a 100 µg pDNA dose, significantly reduced HSV-2 DNA copy number, compared with FL gD2 DNA alone. In addition, 40 % of mice vaccinated with adjuvanted FL pDNA had no detectable HSV-2 viral genomes in the DRG, whereas all mice vaccinated with gD2 pDNA alone were positive for HSV-2 viral genomes. These results show the potential contribution of Vaxfectin-gD2 pDNA to a future multivalent HSV-2 vaccine.


Subject(s)
Herpes Genitalis/prevention & control , Herpesvirus 2, Human/immunology , Phosphatidylethanolamines/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/immunology , Female , Herpes Genitalis/immunology , Herpes Genitalis/virology , Herpesvirus 2, Human/genetics , Mice , Mice, Inbred BALB C , Phosphatidylethanolamines/administration & dosage , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
12.
Lancet Infect Dis ; 12(4): 290-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22237175

ABSTRACT

BACKGROUND: Cytomegalovirus reactivation occurs within 6 months in 60-70% of cytomegalovirus-seropositive patients after allogeneic haemopoietic stem-cell transplantation (HSCT), mainly due to immunosuppression associated with the procedure. Pre-emptive antiviral therapy reduces incidence of cytomegalovirus disease but can be toxic. To reduce the potential for disease and subsequent need for such antiviral drugs, we aimed to assess safety and efficacy of a cytomegalovirus therapeutic DNA vaccine compared with placebo. METHODS: In this exploratory double-blind, placebo-controlled, parallel group, phase 2 trial, up to 80 donor-recipient pairs and 80 unpaired recipients undergoing allogeneic HSCT were planned for enrolment at 16 transplant centres in the USA. Eligible recipients were cytomegalovirus-seropositive, 18-65 years old, without high-risk primary disease, T-cell depletion, previous vaccination for cytomegalovirus, or autoimmune diseases. We randomly allocated participants in both parallel groups in a 1:1 ratio to receive a cytomegalovirus therapeutic DNA vaccine (TransVax; Vical, San Diego, CA, USA) or placebo before conditioning and at 1, 3, and 6 months after transplantation. The vaccine contains plasmids encoding cytomegalovirus glycoprotein B and phosphoprotein 65 formulated with poloxamer CRL1005 and benzalkonium chloride. Randomisation was done by sequential allocation based on Pocock and Simon's method, and stratified by site, donor-recipient HLA matching status, and donor's cytomegalovirus serostatus. The primary outcome was the occurrence rate of clinically significant viraemia resulting in initiation of cytomegalovirus-specific antiviral therapy in the per-protocol assessable population. We assessed rates of adverse events in all participants who received at least one dose of vaccine or placebo. This study is registered with ClinicalTrials.gov, number NCT00285259. FINDINGS: We randomly allocated 108 participants (94 HSCT recipients and 14 paired donors) between June 29, 2006, and Dec 11, 2009. Enrolment of the paired arm was halted in February 2008 for logistical reasons. Safety was assessed in all participants; the efficacy population was restricted to 74 unpaired recipients. Groups were balanced for demographic and clinical variables. 19 (48%) of 40 vaccine recipients required cytomegalovirus-specific antiviral therapy, compared with 21 (62%) of 34 controls (p=0·145). However, during follow-up vaccine significantly reduced the occurrence and recurrence of cytomegalovirus viraemia and improved the time-to-event for viraemia episodes compared with placebo. The vaccine was well-tolerated; only one participant discontinued after an allergic reaction. Incidence of common adverse events after HSCT (eg, graft-versus-host disease or secondary infections) did not differ between groups. INTERPRETATION: We show proof of concept for an immunotherapeutic cytomegalovirus vaccine (TransVax) for clinically significant viraemia in the HSCT setting. The reported safety and efficacy outcomes support further development in a phase 3 trial, notwithstanding a lack of significant reduction in the use of cytomegalovirus-specific antiviral therapy compared with placebo in this phase 2 trial. FUNDING: Vical and US National Institute of Allergy and Infectious Diseases.


Subject(s)
Cytomegalovirus Infections/therapy , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus/immunology , Hematopoietic Stem Cell Transplantation/methods , Vaccines, DNA/administration & dosage , Adult , Aged , Antibodies, Viral/blood , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/immunology , Double-Blind Method , Humans , Male , Middle Aged , Regression Analysis , Vaccines, DNA/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viremia/immunology , Viremia/therapy , Viremia/virology , Young Adult
13.
Vaccine ; 29(33): 5443-52, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21641955

ABSTRACT

Vaxfectin(®) is a lipid-based adjuvant initially developed for use with plasmid DNA (pDNA) vaccines. Here we present detailed nonclinical assessments performed prior to Vaxfectin(®)'s first-in-man use, as an adjuvant in the H5N1 influenza vaccine VCL-IPT1. Following IM delivery to rabbits, VCL-IPT1 pDNA localized primarily to injection sites, where levels steadily declined over the 2 months examined. Risk of pDNA integration into genomic DNA was negligible. Toxicology studies in rabbits revealed mild inflammatory/immune responses at injection sites characteristic of IM vaccine delivery; Vaxfectin(®) directly contributed to these responses. These data support clinical development of H5N1 pDNA vaccines, and also present an encouraging profile for further development of Vaxfectin(®) as an adjuvant for vaccines in general.


Subject(s)
Adjuvants, Immunologic/pharmacokinetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/pharmacokinetics , Phosphatidylethanolamines/pharmacokinetics , Vaccines, DNA/pharmacokinetics , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Animals , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Influenza Vaccines/genetics , Injections, Intramuscular , Male , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/adverse effects , Rabbits , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects , Vaccines, DNA/genetics
14.
Expert Opin Drug Deliv ; 7(12): 1433-46, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21118032

ABSTRACT

IMPORTANCE OF THE FIELD: Many vaccines require the use of an adjuvant to achieve immunity. So far, few adjuvants have advanced successfully through clinical trials to become part of licensed vaccines. Vaxfectin® (Vical, CA, USA) represents a next-generation adjuvant with promise as a platform technology, showing utility with both plasmid DNA (pDNA) and protein-based vaccines. AREAS COVERED IN THIS REVIEW: This review describes the chemical, physical, preclinical and clinical development of Vaxfectin for pDNA-based vaccines. Also included is the preclinical development of Vaxfectin-adjuvanted protein- and peptide-based vaccines. WHAT THE READER WILL GAIN: The reader will gain knowledge of vaccine adjuvant development from bench to bedside. TAKE HOME MESSAGE: Vaxfectin has effectively boosted the immune response against a range of pDNA-expressed pathogenic antigens in preclinical models extending from rodents to non-human primates. In the clinic, Vaxfectin-adjuvanted pDNA-based H5N1 influenza vaccines have been shown to be well tolerated and to result in durable immune responses within the predicted protective range reported for protein-based vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Viral/immunology , Phosphatidylethanolamines/administration & dosage , Vaccines, DNA/administration & dosage , Viral Vaccines/administration & dosage , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/chemistry , Animals , Antigens, Viral/genetics , Humans , Immunization, Secondary , Phosphatidylethanolamines/adverse effects , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/immunology , Plasmids , Vaccines, DNA/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Vaccines/immunology
15.
Vaccine ; 28(13): 2565-72, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20117262

ABSTRACT

BACKGROUND: Development of vaccines against highly pathogenic avian influenza virus H5N1 subtypes posing a pandemic threat remains a priority. Limitations in manufacturing capacity and production time of conventional inactivated vaccines highlight the need for additional approaches. METHODS: We conducted two double-blind, placebo-controlled phase 1 studies involving a total of 103 healthy adults who received two intramuscular injections of Vaxfectin-adjuvanted plasmid DNA vaccine or placebo 21 days apart. Vaccine cohorts received either a monovalent vaccine containing an A/Vietnam/1203/04 H5 hemagglutinin-encoding plasmid or a trivalent vaccine with plasmids encoding H5, NP, and M2 proteins in doses from 0.1 to 1mg of DNA/injection. RESULTS: All doses were well tolerated without vaccine-related serious adverse events or discontinuations. In the monovalent cohorts, hemagglutination inhibition (HI) titers of > or =40 and 4-fold rises from baseline were achieved in 47-67% of subjects and H5-specific T-cell responses in 75-100%. Trivalent cohorts had lower HI response rates (< or = 20%), but 72% of subjects achieved T-cell and/or antibody responses to one or more antigens. CONCLUSIONS: Vaxfectin-adjuvanted monovalent H5 DNA vaccines were well tolerated and induced HI response rates and titers in the reported range of inactivated protein-based H5 vaccines, suggesting that adjuvanted DNA vaccines with rapid vaccine production could be useful for pandemic control.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/adverse effects , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adult , Antibodies, Viral/blood , Double-Blind Method , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunization, Secondary/methods , Influenza A virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Injections, Intramuscular , Male , Nucleocapsid Proteins , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/adverse effects , Placebos/administration & dosage , Plasmids , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology
16.
Vaccine ; 27(52): 7409-17, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-19735757

ABSTRACT

Cationic lipids have been used as delivery systems to enhance the performance of vaccines and immunotherapeutics. However, little is known about the effect of administration of cationic lipid-formulated vaccines on gene expression. This study used DNA microarrays (39,000 transcripts) to characterize early changes in gene expression patterns in mouse muscle 1 and 2 days after intramuscular (i.m.) injection of a hCMV gB plasmid DNA (pDNA) vaccine formulated with the cationic lipid system Vaxfectin; gene expression profiles were compared to those obtained after i.m. injection of pDNA in PBS. Analysis of the DNA microarray data indicated that approximately 1% of the represented transcripts were modulated at least 2-fold compared to the PBS samples at both time points. Functional analysis of the modulated genes revealed that transcripts involved in antigen processing and presentation, apoptosis and the Toll-like receptor pathway were significantly enriched. In addition, confirmation of local and systemic modulation of subsets of biomarkers was achieved using Real-Time PCR and Cytometric Bead Assays. Time course and magnitude of cellular infiltration (F4/80+ and CD11b+ cells) to the injection site was changed in response to formulation of hCMVgB pDNA with Vaxfectin. Since the expression level of the pDNA-encoded transgene in the muscle was not affected by formulation Vaxfectin mechanism of action is expected to rely primarily on modulation of immune pathways and not on an increase in transfection of the antigen-encoding pDNA. Taken together, these data help explain the Vaxfectin-dependent robust enhancement of immune responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Biomarkers/blood , Cytomegalovirus Vaccines/immunology , Phosphatidylethanolamines/immunology , Viral Envelope Proteins/immunology , Animals , Cytokines/blood , Cytomegalovirus Vaccines/administration & dosage , Female , Gene Expression Profiling , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Plasmids , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
17.
Vaccine ; 27(46): 6399-403, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19552895

ABSTRACT

Mice were immunized either with unadjuvanted seasonal trivalent influenza vaccine (TIV) or TIV formulated with Vaxfectin, a cationic lipid-based adjuvant. Increasing doses of Vaxfectin resulted in increased hemagglutination-inhibition or anti-TIV ELISA antibody titers, with up to a 200-fold increase obtained with 900 microg of Vaxfectin. A >or=10-fold dose-sparing effect was demonstrated with Vaxfectin formulations. Vaxfectin preferentially increased IgG2 titers compared to IgG1 titers, resulting in a balanced IgG isotype distribution. Lower doses of Vaxfectin (30 microg) did not enhance antibody responses, but increased the number of IFN-gamma secreting T-cells by up to 18-fold. The data demonstrate that Vaxfectin enhances Th1 responses with protein-based seasonal influenza vaccine, and suggest that cellular or humoral immune responses may be preferentially induced by modifying the Vaxfectin:antigen ratio in the vaccine formulation.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Phosphatidylethanolamines/pharmacology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Antibody Formation , Antigens, Viral/immunology , Cations/immunology , Cations/pharmacology , Cells, Cultured , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination Inhibition Tests , Immunity, Cellular , Immunity, Humoral , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Interferon-gamma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Phosphatidylethanolamines/administration & dosage , Phosphatidylethanolamines/immunology , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology
18.
Hum Vaccin ; 5(8): 536-44, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19458488

ABSTRACT

VCL-AB01, a cationic lipid-formulated plasmid DNA (pDNA)-based vaccine that contains genes encoding genetically detoxified Bacillus anthracis protective antigen (PA) and lethal factor (LF), was assessed in a Phase 1, dose-escalating clinical trial in healthy adults for safety and immunogenicity, and in nonhuman primates for immunogenicity and efficacy against challenge with a lethal dose of B. anthracis spores. Healthy 18-45 year old subjects were randomly assigned to receive either the investigational vaccine containing 0.2 mg, 0.6 mg, or 2 mg of total pDNA per dose, or saline placebo, administered at 0, 1 and 2 months. The 0.2 mg and 0.6 mg dose levels were generally well tolerated; however, dose-limiting reactogenicity was observed among subjects given the first 2 mg dose and the remaining two injections in the 2 mg group were reduced to 0.6 mg. Dose-related increases in seroconversion frequencies were observed. Overall, 10%, 33.3% and 80% of subjects in the 0.2, 0.6 and 2 mg groups, respectively, developed antibodies to PA and/or LF as measured by ELISA; however, antibodies with toxin neutralizing activity (TNA) were detected in only one subject. In monkeys that received a 0.6 mg dose three times at 2 week intervals, low levels of antibodies were detected by ELISA but not by the TNA assay in all animals just prior to challenge. Despite the absence of TNA, 75% animals survived the lethal challenge. In summary, VCL-AB01 was generally well tolerated in humans at a dose that provided immunity in monkeys despite the lack of robust TNA titers in either species.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/immunology , Anthrax/prevention & control , Bacillus anthracis/immunology , Vaccines, DNA/immunology , Adolescent , Adult , Animals , Anthrax/blood , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/adverse effects , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Drug Administration Schedule , Drug Evaluation, Preclinical , Female , Humans , Injections, Intramuscular , Macaca fascicularis , Male , Middle Aged , New York , Rabbits , Texas , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/adverse effects
19.
Clin Vaccine Immunol ; 15(8): 1214-21, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524884

ABSTRACT

A measles virus vaccine for infants under 6 months of age would help control measles. DNA vaccines hold promise, but none has provided full protection from challenge. Codon-optimized plasmid DNAs encoding the measles virus hemagglutinin and fusion glycoproteins were formulated with the cationic lipid-based adjuvant Vaxfectin. In mice, antibody and gamma interferon (IFN-gamma) production were increased by two- to threefold. In macaques, juveniles vaccinated at 0 and 28 days with 500 microg of DNA intradermally or with 1 mg intramuscularly developed sustained neutralizing antibody and H- and F-specific IFN-gamma responses. Infant monkeys developed sustained neutralizing antibody and T cells secreting IFN-gamma and interleukin-4. Twelve to 15 months after vaccination, vaccinated monkeys were protected from an intratracheal challenge: viremia was undetectable by cocultivation and rashes did not appear, while two naïve monkeys developed viremia and rashes. The use of Vaxfectin-formulated DNA is a promising approach to the development of a measles vaccine for young infants.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hemagglutinins, Viral/immunology , Measles Vaccine/administration & dosage , Measles/prevention & control , Phosphatidylethanolamines/administration & dosage , Vaccines, DNA/administration & dosage , Viral Fusion Proteins/immunology , Animals , Antibodies, Viral/blood , Female , Hemagglutinins, Viral/genetics , Humans , Interferon-gamma/metabolism , Macaca mulatta , Measles/immunology , Measles Vaccine/genetics , Measles Vaccine/immunology , Measles virus/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Phosphatidylethanolamines/immunology , Sequence Analysis, DNA , T-Lymphocytes/immunology , Vaccination , Vaccines, DNA/immunology , Viral Fusion Proteins/genetics
20.
J Infect Dis ; 197(12): 1643-52, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18513153

ABSTRACT

Plasmid DNA (pDNA) vaccines represent an alternative to conventional inactivated influenza vaccines that are likely to experience supply constraints during a pandemic. Several Vaxfectin-formulated pDNA vaccines were tested in mice and ferrets for efficacy against a lethal challenge with the highly pathogenic A/Vietnam/1203/04 (H5N1) influenza virus strain; the vaccines encoded influenza A virus hemagglutinin (HA), and/or nucleoprotein (NP), and M2 protein. Complete protection from death and disease was achieved in mice and ferrets with 2 doses of a Vaxfectin-formulated vaccine containing H5 HA, NP, and M2 plasmids and in ferrets with only 1 dose. A Vaxfectin-formulated vaccine containing NP and M2 pDNA provided significant protection against death in mice and provided some benefit in ferrets (i.e., 17% survival, delayed time to illness and death, and significant reduction in viral load compared with that in negative control animals). These experiments support the clinical testing of pDNA vaccine candidates that may ultimately increase global vaccine supply options during pandemics.


Subject(s)
Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Plasmids , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Body Weight , Chemistry, Pharmaceutical , Ferrets , Mice , Mice, Inbred BALB C , Nasal Lavage Fluid/virology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Phosphatidylethanolamines , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...