Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 727
1.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Article En | MEDLINE | ID: mdl-33751208

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Antigens, Neoplasm/therapeutic use , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Esophageal Neoplasms/drug therapy , Glucans/therapeutic use , Membrane Proteins/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Adjuvants, Immunologic/therapeutic use , Aged , Aged, 80 and over , Animals , Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/therapeutic use , Esophageal Neoplasms/immunology , Female , Glucans/immunology , Humans , Interferon Inducers/immunology , Interferon Inducers/therapeutic use , Male , Membrane Proteins/immunology , Mice , Middle Aged , Nanoparticles , Poly I-C/immunology , Polylysine/immunology , Polylysine/therapeutic use
2.
Minerva Med ; 112(1): 144-152, 2021 Feb.
Article En | MEDLINE | ID: mdl-33104300

The world is now entering its 9th month of combat against a pandemic of deadly pneumonia. Started out from China in December 2019, the disease has been declared as caused by infection with a so far unknown RNA Coronavirus of the respiratory family, then named severe acute respiratory syndrome coronavirus SARS-CoV-2. In the absence of a vaccine, and with scientists still struggling for an effective therapy, COVID-19 (the SARS-dependent syndrome) carries up to now, a death toll of more than 590,000 (July 18,2020) undermining jobs and finance of contemporary society in all continents. Social distancing, the only measure hitherto shown to restrain virus spread, has been progressively loosened from May 2020 in some countries, leaving us in the fear of repeat attacks from the unchecked virus. We discuss the problem and propose to tentatively boost the antivirus cell machinery by using lab-made viral mimics to engage cell receptors.


COVID-19/therapy , SARS-CoV-2 , COVID-19/complications , COVID-19/epidemiology , Carboxymethylcellulose Sodium/analogs & derivatives , Carboxymethylcellulose Sodium/therapeutic use , Cytokine Release Syndrome/etiology , Humans , Immunization, Passive , Interferon Inducers/therapeutic use , Mucocutaneous Lymph Node Syndrome/etiology , Physical Distancing , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Polylysine/therapeutic use , Practice Guidelines as Topic , RNA, Double-Stranded/drug effects , RNA, Viral/drug effects , Recurrence , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Secondary Prevention , COVID-19 Drug Treatment , COVID-19 Serotherapy
3.
Int J Med Sci ; 17(12): 1803-1810, 2020.
Article En | MEDLINE | ID: mdl-32714083

Since the end of 2019, a new type of coronavirus pneumonia (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) has been spreading rapidly throughout the world. Previously, there were two outbreaks of severe coronavirus caused by different coronaviruses worldwide, namely Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and the Middle East Respiratory Syndrome Coronavirus (MERS-CoV). This article introduced the origin, virological characteristics and epidemiological overview of SARS-CoV-2, reviewed the currently known drugs that may prevent and treat coronavirus, explained the characteristics of the new coronavirus and provided novel information for the prevention and treatment of COVID-19.


Betacoronavirus , Coronavirus Infections/drug therapy , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/drug therapy , Pneumonia, Viral/prevention & control , Amides/pharmacology , Amides/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Betacoronavirus/isolation & purification , Betacoronavirus/physiology , COVID-19 , Chloroquine/analogs & derivatives , Chloroquine/therapeutic use , Chlorpromazine/therapeutic use , Coronavirus/genetics , Coronavirus Infections/genetics , Cyclophilins/antagonists & inhibitors , Drug Development , Drug Repositioning , Drugs, Chinese Herbal/therapeutic use , Endocytosis/drug effects , Humans , Immune Sera , Interferon Inducers/therapeutic use , Nucleic Acid Synthesis Inhibitors/pharmacology , Nucleic Acid Synthesis Inhibitors/therapeutic use , Pneumonia, Viral/genetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Resveratrol/pharmacology , Resveratrol/therapeutic use , SARS-CoV-2 , Viral Vaccines/therapeutic use , COVID-19 Drug Treatment
4.
DNA Cell Biol ; 39(9): 1730-1740, 2020 Sep.
Article En | MEDLINE | ID: mdl-32580635

Polyinosinic-polycytidylic acid (PIC) is a potent double-stranded RNA (dsRNA) adjuvant useful in intranasal influenza vaccination. In mice, the intensity and duration of immune responses to PIC correlated with the double-stranded chain length. A rational method to avoid PIC chain extension in PIC production is to use multiple short poly(I) molecules and one long poly(C) molecule for PIC assembly. In this study, we elucidate that a newly developed uPIC100-400 molecule comprising multiple 0.1 kb poly(I) molecules and one 0.4 kb poly(C) molecule effectively enhanced the immune responses in mice, by preventing the challenged viral propagation and inducing hemagglutinin-specific IgA, after intranasal A(H1N1)pdm09 influenza vaccination. Reduced intraperitoneal toxicity of PIC prepared with multiple short poly(I) molecules in mice indicates the widened effective range of uPIC100-400 as an adjuvant. In contrast to uPIC100-400, the PIC molecule comprising multiple 0.05 kb poly(I) molecules failed to elicit mouse mucosal immunity. These results were consistent with TLR3 response but not retinoic acid inducible gene I (RIG-I)-like receptor response in the cell assays, which suggests that the adjuvant effect of PIC in mouse intranasal immunization depends on TLR3 signaling. In conclusion, the double-stranded PIC with reduced toxicity developed in this study would contribute to the development of PIC-adjuvanted vaccines.


Adjuvants, Immunologic/therapeutic use , Interferon Inducers/therapeutic use , Orthomyxoviridae Infections/immunology , Poly I-C/therapeutic use , Toll-Like Receptor 3/metabolism , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Animals , Cells, Cultured , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin A/immunology , Influenza Vaccines/immunology , Interferon Inducers/administration & dosage , Interferon Inducers/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae Infections/prevention & control , Poly I-C/administration & dosage , Poly I-C/adverse effects , Signal Transduction
6.
Vestn Otorinolaringol ; 84(3): 82-88, 2019.
Article Ru | MEDLINE | ID: mdl-31486434

Medical scientific sources about randomized clinical trials of Cycloferon were studied as a single plot of 531 articles. AIM was to get the generalized cycloferon efficacy's assessment in comparison with basic therapies curing the otorhinolaryngologic diseases. Groups heterogeneity and responding parameters' variability were estimated also. Comparison groups were symmetric enough to annihilate end-point variabilities, so the results' interpretations were clear enough and vector of clinical effects was detectable. Comparison groups were integrated to increase statistical power of metaanalysis. In result, cycloferon additional administration in treatment of otorhinolaryngologic diseases added 25% to absolute and relative usefulness of medical intervention. Also cycloferon increased 3,5 times the chances of recovery and absence of recurrent exacerbation.


Acridines/therapeutic use , Interferon Inducers/therapeutic use , Respiratory Tract Infections/drug therapy , Humans , Nose Diseases
7.
Int J Mol Sci ; 20(9)2019 May 07.
Article En | MEDLINE | ID: mdl-31067687

Rhinovirus (RV) is the predominant virus causing respiratory tract infections. Bronchobini® is a low dose multi component, multi target preparation used to treat inflammatory respiratory diseases such as the common cold, described to ease severity of symptoms such as cough and viscous mucus production. The aim of the study was to assess the efficacy of Bronchobini® in RV infection and to elucidate its mode of action. Therefore, Bronchobini®'s ingredients (BRO) were assessed in an ex vivo model of RV infection using mouse precision-cut lung slices, an organotypic tissue capable to reflect the host immune response to RV infection. Cytokine profiles were assessed using enzyme-linked immunosorbent assay (ELISA) and mesoscale discovery (MSD). Gene expression analysis was performed using Affymetrix microarrays and ingenuity pathway analysis. BRO treatment resulted in the significant suppression of RV-induced antiviral and pro-inflammatory cytokine release. Transcriptome analysis revealed a multifactorial mode of action of BRO, with a strong inhibition of the RV-induced pro-inflammatory and antiviral host response mediated by nuclear factor kappa B (NFkB) and interferon signaling pathways. Interestingly, this was due to priming of these pathways in the absence of virus. Overall, BRO exerted its beneficial anti-inflammatory effect by priming the antiviral host response resulting in a reduced inflammatory response to RV infection, thereby balancing an otherwise excessive inflammatory response.


Antiviral Agents/pharmacology , Interferon Inducers/pharmacology , Interferons/metabolism , Lung/drug effects , Picornaviridae Infections/drug therapy , Plant Extracts/pharmacology , Transcriptome , Animals , Antiviral Agents/therapeutic use , Female , Interferon Inducers/therapeutic use , Lung/metabolism , Lung/virology , Mice , Mice, Inbred BALB C , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Plant Extracts/therapeutic use , Rhinovirus/drug effects , Rhinovirus/pathogenicity , Signal Transduction
8.
Antiviral Res ; 163: 179-184, 2019 03.
Article En | MEDLINE | ID: mdl-30611774

The potential protection of poly-ICLC (Hiltonol®) a double stranded RNA (dsRNA) against EBOV infection was assessed with prophylactic and therapeutic administration to wild type and TLR3-negative mice, and in non-human primates (NHPs) by measuring EBOL serum titers, survival extension, and serum liver and kidney function markers. Various doses of aqueous and liposomal poly-ICLC monotherapy provided robust protection in otherwise lethal murine EBOV challenge models, when treatment is started on the day 0 or one day after virus challenge. There was no advantage of liposomal vs. the aqueous poly-ICLC form. Protection appeared to be independent of TLR-3. NHPs treated with poly-ICLC and challenged with EBOV survived longer but eventually succumbed to Ebola infection. Nevertheless, the liver and kidney serum markers were markedly reduced in the infected and treated NHPs. In the two longest surviving poly-ICLC- treated NHPs, the day 10 serum EBOV titer was reduced 2.1 and 30 fold respectively.


Carboxymethylcellulose Sodium/analogs & derivatives , Hemorrhagic Fever, Ebola/drug therapy , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Animals , Carboxymethylcellulose Sodium/therapeutic use , Democratic Republic of the Congo , Female , Macaca fascicularis , Mice , Mice, Inbred BALB C , Polylysine/therapeutic use
9.
PLoS One ; 13(11): e0206827, 2018.
Article En | MEDLINE | ID: mdl-30383838

Hyperuricaemia is associated with various metabolic dysfunctions including obesity, type 2 diabetes mellitus, hypertension and in general metabolic syndrome, which are all associated with increased risk of cancer. However, the direct association between elevated uricemia and cancer mortality still remains unclear. In this study, we used a mouse model of hyperuricemia, the Urahplt2/plt2 (PLT2) mouse, to investigate the effect of high uric acid levels on anti-tumor immune responses and tumor growth. In normo-uricaemic C57BL/6 mice injected with B16 melanomas, immunotherapy by treatment with Poly I:C at the tumor site delayed tumor growth compared to PBS treatment. In contrast, Poly I:C-treated hyper-uricaemic PLT2 mice were unable to delay tumor growth. Conventional and monocyte-derived dendritic cells in the tumor-draining lymph nodes (dLN) of C57BL/6 and PLT2 mice were similarly increased after Poly I:C immunotherapy, and expressed high levels of CD40 and CD86. CD8+ T cells in the tumor-dLN and tumor of both WT and PLT2 mice were also increased after Poly I:C immunotherapy, and were able to secrete increased IFNγ upon in vitro restimulation. Surprisingly, tumor-specific CD8+ T cells in dLN were less abundant in PLT2 mice compared to C57BL/6, but showed a greater ability to proliferate even in the absence of cognate antigen. These data suggest that hyperuricaemia may affect the functionality of CD8+ T cells in vivo, leading to dysregulated T cell proliferation and impaired anti-tumor activity.


CD8-Positive T-Lymphocytes/immunology , Hyperuricemia/immunology , Interferon Inducers/pharmacology , Melanoma, Experimental/immunology , Poly I-C/pharmacology , Skin Neoplasms/immunology , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Carcinogenesis/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Dendritic Cells/immunology , Female , Humans , Hyperuricemia/blood , Hyperuricemia/genetics , Hyperuricemia/metabolism , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunotherapy/methods , Interferon Inducers/therapeutic use , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Melanoma, Experimental/blood , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/therapeutic use , Skin Neoplasms/blood , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Treatment Outcome , Uric Acid/blood , Uric Acid/metabolism
10.
Hum Vaccin Immunother ; 14(7): 1815-1819, 2018 07 03.
Article En | MEDLINE | ID: mdl-29505317

Genital warts are one of the most common sexually transmitted diseases worldwide. The disease is a result of infection with low-risk types of human papillomaviruses, mostly type 6 and 11. Current therapies for genital warts are mainly ablative, or alternatively topical application of imiquimod cream and sinecatechin (polyphenon E) ointment to the warts. However, low patient compliance and high recurrence rate are significant problems for the treatment of genital warts by imiquimod and ablative therapies. We summarise recent literature in this area and propose combining imiquimod with other therapies to increase the efficacy of imiquimod.


Condylomata Acuminata/therapy , Imiquimod/therapeutic use , Interferon Inducers/therapeutic use , Administration, Topical , Anus Diseases/virology , Catechin/analogs & derivatives , Catechin/therapeutic use , Condylomata Acuminata/drug therapy , Drug Therapy, Combination , Humans , Interleukin-10/antagonists & inhibitors , Papillomaviridae , Recurrence , Treatment Outcome
12.
J Immunother Cancer ; 5(1): 92, 2017 11 21.
Article En | MEDLINE | ID: mdl-29157306

BACKGROUND: Breast cancer remains a leading cause of cancer death worldwide. There is evidence that immunotherapy may play a role in the eradication of residual disease. Peptide vaccines for immunotherapy are capable of durable immune memory, but vaccines alone have shown sparse clinical activity against breast cancer to date. Toll-like receptor (TLR) agonists and helper peptides are excellent adjuvants for vaccine immunotherapy and they are examined in this human clinical trial. METHODS: A vaccine consisting of 9 MHC class I-restricted breast cancer-associated peptides (from MAGE-A1, -A3, and -A10, CEA, NY-ESO-1, and HER2 proteins) was combined with a TLR3 agonist, poly-ICLC, along with a helper peptide derived from tetanus toxoid. The vaccine was administered on days 1, 8, 15, 36, 57, 78. CD8+ T cell responses to the vaccine were assessed by both direct and stimulated interferon gamma ELIspot assays. RESULTS: Twelve patients with breast cancer were treated: five had estrogen receptor positive disease and five were HER2 amplified. There were no dose-limiting toxicities. Toxicities were limited to Grade 1 and Grade 2 and included mild injection site reactions and flu-like symptoms, which occurred in most patients. The most common toxicities were injection site reaction/induration and fatigue, which were experienced by 100% and 92% of participants, respectively. In the stimulated ELIspot assays, peptide-specific CD8+ T cell responses were detected in 4 of 11 evaluable patients. Two patients had borderline immune responses to the vaccine. The two peptides derived from CEA were immunogenic. No difference in immune response was evident between patients receiving endocrine therapy and those not receiving endocrine therapy during the vaccine series. CONCLUSIONS: Peptide vaccine administered in the adjuvant breast cancer setting was safe and feasible. The TLR3 adjuvant, poly-ICLC, plus helper peptide mixture provided modest immune stimulation. Further optimization is required for this multi-peptide vaccine/adjuvant combination. TRIAL REGISTRATION: ClinicalTrials.gov (posted 2/15/2012): NCT01532960. Registered 2/8/2012. https://clinicaltrials.gov/show/NCT01532960.


Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Adjuvants, Immunologic , Adult , Cancer Vaccines/pharmacology , Carboxymethylcellulose Sodium/pharmacology , Carboxymethylcellulose Sodium/therapeutic use , Female , Humans , Immunotherapy , Interferon Inducers/pharmacology , Middle Aged , Pilot Projects , Poly I-C/pharmacology , Polylysine/pharmacology , Polylysine/therapeutic use
13.
Cell Rep ; 21(7): 1870-1882, 2017 Nov 14.
Article En | MEDLINE | ID: mdl-29141219

Liver cancer has become the second most deadly malignant disease, with no efficient targeted or immune therapeutic agents available yet. While dissecting the roles of cytoplasmic signaling molecules in hepatocarcinogenesis using an inducible mouse gene targeting system, Mx1-cre, we identified a potent liver tumor-inhibitory effect of synthetic double-stranded RNA (dsRNA), polyinosinic-polycytidylic acid (pIC), an inducer of the Mx1-cre system. Injection of pIC at the pre-cancer stage robustly suppressed liver tumorigenesis either induced by chemical carcinogens or by Pten loss and associated hepatosteatosis. The immunostimulatory dsRNA inhibited liver cancer initiation, apparently by boosting multiple anti-tumor activities of innate immunity, including induction of immunoregulatory cytokines, activation of NK cells and dendritic cells, and reprogramming of macrophage polarization. This study paves the way for the development of preventive and early interfering strategies for liver cancer to reduce the rapidly increasing incidences of liver cancer in an ever-growing population with chronic liver disorders.


Carcinogenesis/immunology , Immunity, Innate , Interferon Inducers/pharmacology , Liver Neoplasms/immunology , Poly I-C/pharmacology , Animals , Carcinogenesis/drug effects , Dendritic Cells/immunology , Interferon Inducers/therapeutic use , Killer Cells, Natural/immunology , Liver Neoplasms/prevention & control , Macrophage Activation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Poly I-C/therapeutic use , RNA, Double-Stranded/pharmacology
15.
Antiviral Res ; 139: 1-12, 2017 Mar.
Article En | MEDLINE | ID: mdl-27956136

Hiltonol®, (Poly IC:LC), a potent immunomodulator, is a synthetic, double-stranded polyriboinosinic-polyribocytidylic acid (poly IC) stabilized with Poly-L-lysine and carboxymethyl cellulose (LC). Hiltonol® was tested for efficacy in a lethal SARS-CoV-infected BALB/c mouse model. Hiltonol® at 5, 1, 0.5 or 0.25 mg/kg/day by intranasal (i.n.) route resulted in significant survival benefit when administered at selected times 24 h prior to challenge with a lethal dose of mouse-adapted severe acute respiratory syndrome coronavirus (SARS-CoV). The infected BALB/c mice receiving the Hiltonol® treatments were also significantly effective in protecting mice against weight loss due to infection (p < 0.001). Groups of 20 mice were dosed with Hiltonol® at 2.5 or 0.75 mg/kg by intranasal instillation 7, 14, and 21 days before virus exposure and a second dose was given 24 h later, prophylactic Hiltonol® treatments (2.5 mg/kg/day) were completely protective in preventing death, and in causing significant reduction in lung hemorrhage scores, lung weights and lung virus titers. Hiltonol® was also effective as a therapeutic when give up to 8 h post virus exposure; 100% of the-infected mice were protected against death when Hiltonol® was administered at 5 mg/kg/day 8 h after infection. Our data suggest that Hiltonol® treatment of SARS-CoV infection in mice leads to substantial prophylactic and therapeutic effects and could be used for treatment of other virus disease such as those caused by MERS-CoV a related coronavirus. These properties might be therapeutically advantageous if Hiltonol® is considered for possible clinical use.


Carboxymethylcellulose Sodium/analogs & derivatives , Immunomodulation , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/prevention & control , Adjuvants, Immunologic , Administration, Intranasal , Animals , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/therapeutic use , Disease Models, Animal , Interferon Inducers/administration & dosage , Lung/drug effects , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Poly I-C/administration & dosage , Polylysine/administration & dosage , Polylysine/therapeutic use , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe Acute Respiratory Syndrome/mortality , Severe Acute Respiratory Syndrome/virology , Survival Analysis
16.
Sex Transm Infect ; 93(3): 155-161, 2017 05.
Article En | MEDLINE | ID: mdl-27803240

BACKGROUND: Anogenital warts (AGWs, condylomata acuminata) are among the most common STIs and may severely impact quality of life (QoL). Available treatment options are characterised by a high proportion of non-responders and recurrences. OBJECTIVE: To systematically review and meta-analyse the available evidence from randomised controlled trials (RCTs) on topical treatments for AGWs considering short-term and long-term efficacy, effects on QoL and adverse events (AE). METHODS: A comprehensive literature search was performed in Cochrane Central Register of Controlled Trials, Embase and MEDLINE. Included studies were evaluated with the Cochrane Collaboration's risk of bias tool. The confidence in the pooled effect estimates was evaluated according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach and categorised as 'very low', 'low', 'moderate' or 'high'. RESULTS: Eighteen RCTs met the inclusion criteria. Regarding complete clearance (CC), imiquimod 3.75% and 5% cream, podophyllotoxin 0.5% solution and gel and polyphenon E 10% and 15% ointment were superior to placebo. Although more local AE and pain occurred in the actively treated groups, differences regarding dropouts due to AE were not statistically significant. For podophyllotoxin 0.15% cream, no placebo-controlled trials were available; however, in an active-controlled trial, it was inferior to podophyllotoxin 0.5% solution with respect to CC. No significant differences were detected between imiquimod 5% cream and podophyllotoxin 0.5% solution and between polyphenon E 10% and 15% ointment. No data on the influence on health-related QoL were available. CONCLUSION: Our confidence in the pooled estimates (GRADE quality of the evidence) ranged from very low to high. Apart from the given results, other aspects such as availability, costs or patient preference have to be considered when making a treatment choice. Due to the limited number of direct comparisons, conclusions on the relative efficacy of the different treatment options are restricted.


Anus Diseases/drug therapy , Condylomata Acuminata/drug therapy , Interferon Inducers/therapeutic use , Administration, Topical , Anus Diseases/virology , Condylomata Acuminata/virology , Humans , Immunocompetence , Interferon Inducers/administration & dosage , Papillomaviridae/drug effects , Papillomaviridae/isolation & purification , Randomized Controlled Trials as Topic , Recurrence , Self Administration
17.
BMC Infect Dis ; 16(1): 621, 2016 Nov 03.
Article En | MEDLINE | ID: mdl-27809794

BACKGROUND: Bacillus anthracis, the causative agent of anthrax, is a spore forming and toxin producing rod-shaped bacterium that is classified as a category A bioterror agent. This pathogenic microbe can be transmitted to both animals and humans. Clinical presentation depends on the route of entry (direct contact, ingestion, injection or aerosolization) with symptoms ranging from isolated skin infections to more severe manifestations such as cardiac or pulmonary shock, meningitis, and death. To date, anthrax is treatable if antibiotics are administered promptly and continued for 60 days. However, if treatment is delayed or administered improperly, the patient's chances of survival are decreased drastically. In addition, antibiotics are ineffective against the harmful anthrax toxins and spores. Therefore, alternative therapeutics are essential. In this review article, we explore and discuss advances that have been made in anthrax therapy with a primary focus on alternative pre-approved and novel antibiotics as well as anti-toxin therapies. METHODS: A literature search was conducted using the University of Manitoba search engine. Using this search engine allowed access to a greater variety of journals/articles that would have otherwise been restricted for general use. In order to be considered for discussion for this review, all articles must have been published later than 2009. RESULTS: The alternative pre-approved antibiotics demonstrated high efficacy against B. anthracis both in vitro and in vivo. In addition, the safety profile and clinical pharmacology of these drugs were already known. Compounds that targeted underexploited bacterial processes (DNA replication, RNA synthesis, and cell division) were also very effective in combatting B. anthracis. In addition, these novel compounds prevented bacterial resistance. Targeting B. anthracis virulence, more specifically the anthrax toxins, increased the length of which treatment could be administered. CONCLUSIONS: Several novel and pre-existing antibiotics, as well as toxin inhibitors, have shown increasing promise. A combination treatment that targets both bacterial growth and toxin production would be ideal and probably necessary for effectively combatting this armed bacterium.


Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Antitoxins/therapeutic use , Alpha-Globulins/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antigens, Bacterial , Bacillus anthracis , Bacterial Toxins , DNA Helicases/antagonists & inhibitors , Daunorubicin/analogs & derivatives , Daunorubicin/therapeutic use , Doxorubicin/therapeutic use , Drug Discovery , Fluoroquinolones , Humans , Interferon Inducers/therapeutic use , Levofloxacin , Linezolid , Moxifloxacin , Ofloxacin , Polyketides/therapeutic use , Serine Proteinase Inhibitors/therapeutic use , Tilorone/therapeutic use , Virulence
18.
Dermatol Surg ; 42(12): 1340-1346, 2016 Dec.
Article En | MEDLINE | ID: mdl-27598444

BACKGROUND: Conventional treatments for warts like cryotherapy are limited by the pain during procedures, especially in pediatric patients. Imiquimod is a topical immune response modifier, but the thick stratum corneum of common warts prevents drug permeation through skin. OBJECTIVE: To evaluate the efficacy and safety of fractional laser/topical 5% imiquimod cream for the treatment of warts in children. METHODS: Eleven pediatric patients with multiple recalcitrant common warts were included. Lesions were treated using an ablative fractional 2,940-nm Er:YAG laser at 1- or 2-week interval. After each laser treatment session, imiquimod 5% cream was self-applied once daily 5 days a week. Response and adverse effects were assessed 2 weekly until complete clearance or up to maximum of 48 weeks. Pain during fractional laser was assessed using a visual analogue scale (0-10). RESULTS: Eight of the 11 (72.7%) children experienced complete clearance. Mean duration was 29.7 (16-48) weeks, and the mean number of fractional laser was 17.5 (8-37). No significant adverse effect was observed. Pain visual analogue scale during fractional laser was 2.4 (1-4) compared to 6.2 (5-8) during cryotherapy. CONCLUSION: This pilot study indicates that fractional laser-assisted topical imiquimod may provide benefit for recalcitrant warts in children.


Aminoquinolines/therapeutic use , Interferon Inducers/therapeutic use , Lasers, Solid-State/therapeutic use , Warts/drug therapy , Administration, Topical , Adolescent , Aminoquinolines/administration & dosage , Anesthesia, Local/methods , Child , Female , Humans , Imiquimod , Interferon Inducers/administration & dosage , Male , Pain Measurement , Pilot Projects , Treatment Outcome
19.
Neuro Oncol ; 18(8): 1157-68, 2016 08.
Article En | MEDLINE | ID: mdl-26984745

BACKGROUND: Low-grade gliomas (LGGs) are the most common brain tumors of childhood. Although surgical resection is curative for well-circumscribed superficial lesions, tumors that are infiltrative or arise from deep structures are therapeutically challenging, and new treatment approaches are needed. Having identified a panel of glioma-associated antigens (GAAs) overexpressed in these tumors, we initiated a pilot trial of vaccinations with peptides for GAA epitopes in human leukocyte antigen-A2+ children with recurrent LGG that had progressed after at least 2 prior regimens. METHODS: Peptide epitopes for 3 GAAs (EphA2, IL-13Rα2, and survivin) were emulsified in Montanide-ISA-51 and administered subcutaneously adjacent to intramuscular injections of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-lymphocyte responses against GAA epitopes. Treatment response was evaluated clinically and by MRI. RESULTS: Fourteen children were enrolled. Other than grade 3 urticaria in one child, no regimen-limiting toxicity was encountered. Vaccination induced immunoreactivity to at least one vaccine-targeted GAA in all 12 evaluable patients: to IL-13Rα2 in 3, EphA2 in 11, and survivin in 3. One child with a metastatic LGG had asymptomatic pseudoprogression noted 6 weeks after starting vaccination, followed by dramatic disease regression with >75% shrinkage of primary tumor and regression of metastatic disease, persisting >57 months. Three other children had sustained partial responses, lasting >10, >31, and >45 months, and one had a transient response. CONCLUSIONS: GAA peptide vaccination in children with recurrent LGGs is generally well tolerated, with preliminary evidence of immunological and clinical activity.


Antigens, Neoplasm/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Glioma/drug therapy , Glioma/immunology , Interferon Inducers/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Vaccination/methods , Adolescent , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/adverse effects , Antigens, Neoplasm/immunology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/adverse effects , Carboxymethylcellulose Sodium/therapeutic use , Child , Child, Preschool , Disease-Free Survival , Epitopes , Female , Humans , Infant , Inhibitor of Apoptosis Proteins/immunology , Interferon Inducers/administration & dosage , Interferon Inducers/adverse effects , Interferon Inducers/immunology , Interleukin-13 Receptor alpha2 Subunit/immunology , Male , Neoplasm Grading , Pilot Projects , Poly I-C/administration & dosage , Poly I-C/adverse effects , Poly I-C/immunology , Polylysine/administration & dosage , Polylysine/adverse effects , Polylysine/immunology , Polylysine/therapeutic use , Receptor, EphA2/immunology , Survivin , Treatment Outcome
...