Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
BMC Oral Health ; 24(1): 525, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702623

ABSTRACT

OBJECTIVE: To evaluate the antibacterial effectiveness of a combination of ε-poly-L-lysine (ε-PL), funme peptide (FP) as well as domiphen against oral pathogens, and assess the efficacy of a BOP® mouthwash supplemented with this combination in reducing halitosis and supragingival plaque in a clinical trial. MATERIALS AND METHODS: The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the compound against Fusobacterium nucleatum, Porphyromonas gingivalis, Streptococcus mutans, and Aggregatibacter actinomycetemcomitans were determined by the gradient dilution method. Subsequently, the CCK-8 assay was used to detect the toxicity of mouthwash on human gingival fibroblastst, and the effectiveness in reducing halitosis and supragingival plaque of the mouthwash supplemented with the combination was analyzed by a randomized, double-blind, parallel-controlled clinical trial. RESULTS: The combination exhibited significant inhibitory effects on tested oral pathogens with the MIC < 1.56% (v/v) and the MBC < 3.13% (v/v), and the mouthwash containing this combination did not inhibit the viability of human gingival fibroblasts at the test concentrations. The clinical trial showed that the test group displayed notably lower volatile sulfur compounds (VSCs) at 0, 10, 24 h, and 7 d post-mouthwash (P < 0.05), compared with the baseline. After 7 days, the VSC levels of the and control groups were reduced by 50.27% and 32.12%, respectively, and notably cutting severe halitosis by 57.03% in the test group. Additionally, the Plaque Index (PLI) of the test and control group decreased by 54.55% and 8.38%, respectively, and there was a significant difference in PLI between the two groups after 7 days (P < 0.01). CONCLUSIONS: The combination of ε-PL, FP and domiphen demonstrated potent inhibitory and bactericidal effects against the tested oral pathogens, and the newly formulated mouthwash added with the combination exhibited anti-dental plaque and anti-halitosis properties in a clinical trial and was safe. TRIAL REGISTRATION: The randomized controlled clinical trial was registered on Chinese Clinical Trial Registry (No. ChiCTR2300073816, Date: 21/07/2023).


Subject(s)
Dental Plaque , Halitosis , Mouthwashes , Polylysine , Humans , Halitosis/prevention & control , Halitosis/drug therapy , Halitosis/microbiology , Mouthwashes/therapeutic use , Dental Plaque/microbiology , Dental Plaque/prevention & control , Double-Blind Method , Male , Female , Polylysine/therapeutic use , Adult , Microbial Sensitivity Tests , Young Adult , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Porphyromonas gingivalis/drug effects , Fusobacterium nucleatum/drug effects , Fibroblasts/drug effects , Peptides/therapeutic use , Peptides/pharmacology , Aggregatibacter actinomycetemcomitans/drug effects , Streptococcus mutans/drug effects
2.
Mil Med ; 188(Suppl 6): 52-60, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37948238

ABSTRACT

INTRODUCTION: Because antibiotic resistance is increasing worldwide and the leading cause of death in burn patients is an infection, an urgent need exists for nonantibiotic approaches to eliminate multidrug-resistant bacteria from burns to prevent their systemic dissemination and sepsis. We previously demonstrated the significant antibiofilm activity of a chitosan (CS) hydrogel containing the antimicrobial peptide epsilon-poly-l-lysine (EPL) against multidrug-resistant Pseudomonas aeruginosa using ex vivo porcine skin. In this study, we evaluated the in vivo antibacterial efficacy of a CS/EPL hydrogel against P. aeruginosa in a murine burn wound infection model. MATERIALS AND METHODS: Full-thickness burns were created on the dorsum using a heated brass rod and were inoculated with bioluminescent, biofilm-forming P. aeruginosa (Xen41). Mice were treated with CS/EPL, CS, or no hydrogel applied topically 2 or 24 hours after inoculation to assess the ability to prevent or eradicate existing biofilms, respectively. Dressing changes occurred daily for 3 days, and in vivo bioluminescence imaging was performed to detect and quantitate bacterial growth. Blood samples were cultured to determine systemic infection. In vitro antibacterial activity and cytotoxicity against human primary dermal fibroblasts, keratinocytes, and mesenchymal stem cells were also assessed. RESULTS: CS/EPL treatment initiated at early or delayed time points showed a significant reduction in bioluminescence imaging signal compared to CS on days 2 and 3 of treatment. Mice administered CS/EPL had fewer bloodstream infections, lower weight loss, and greater activity than the untreated and CS groups. CS/EPL reduced bacterial burden by two orders of magnitude in vitro and exhibited low cytotoxicity against human cells. CONCLUSION: A topical hydrogel delivering the antimicrobial peptide EPL demonstrates in vivo efficacy to reduce but not eradicate established P. aeruginosa biofilms in infected burn wounds. This biocompatible hydrogel shows promise as an antimicrobial barrier dressing for the sustained protection of burn wounds from external bacterial contamination.


Subject(s)
Anti-Infective Agents , Burns , Chitosan , Pseudomonas Infections , Wound Infection , Swine , Mice , Humans , Animals , Hydrogels/pharmacology , Hydrogels/therapeutic use , Pseudomonas aeruginosa , Chitosan/pharmacology , Chitosan/therapeutic use , Polylysine/pharmacology , Polylysine/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Wound Infection/prevention & control , Burns/complications , Burns/drug therapy , Burns/microbiology , Antimicrobial Peptides , Pseudomonas Infections/complications , Pseudomonas Infections/drug therapy
3.
J Thromb Haemost ; 21(6): 1478-1492, 2023 06.
Article in English | MEDLINE | ID: mdl-36871669

ABSTRACT

BACKGROUND: Hemorrhage, in particular noncompressible hemorrhage, is the leading cause of casualties in combat trauma and civilian trauma. Although systemic agents can stop bleeding at both inaccessible and accessible injury sites, the application of systemic hemostats in clinics is strictly limited by the nontargeting ability of hemostats and their subsequent potential for thromboembolic complications. OBJECTIVES: To engineer an anticoagulant/procoagulant self-converting and bleeding site-targeting systemic nanohemostat to rapidly control noncompressible bleeding without thrombosis risk. METHODS: A multiscale computer simulation was taken to guide the self-assembly of sulindac (SUL, a prodrug of the antiplatelets agent) and poly-L-lysine (a cation polymer with platelets activation ability) for forming poly-L-lysine/SUL nanoparticles (PSNs). In vitro platelet-adhering ability, platelet activation effect, and hemostasis activity of PSNs were evaluated. Then, the biosafety, level of thrombosis, targeting ability, and hemostasis effect of systemic applied PSNs were carefully evaluated in various hemorrhage models. RESULTS: PSNs were successfully prepared and showed good platelet adhesion and activation in vitro. The bleeding site-targeting ability and hemostatic efficiency in different bleeding models were leveled up by PSNs markedly compared with vitamin K and etamsylate in vivo. SUL in PSNs could be metabolized into sulindac sulfide at clot sites in 4 hours for antiplatelet aggregation, thus reducing thrombotic risk compared with other hemostatic agents, via the ingenious utilization of prodrug metabolism in terms of time intervals and the adhesion on platelets. CONCLUSION: PSNs are expected to be a low-cost, safe, efficient, clinically translatable first-aid hemostat for first-aid scenarios.


Subject(s)
Hemostatics , Prodrugs , Thrombosis , Humans , Anticoagulants/therapeutic use , Computer Simulation , Polylysine/pharmacology , Polylysine/therapeutic use , Prodrugs/pharmacology , Prodrugs/therapeutic use , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Hemostasis , Hemostatics/therapeutic use , Thrombosis/drug therapy
4.
Arch Gynecol Obstet ; 306(1): 101-108, 2022 07.
Article in English | MEDLINE | ID: mdl-35246717

ABSTRACT

Bacterial vaginosis (BV) is the most common vaginal infection affecting women of childbearing age, and is associated with a substantial burden on women's physical, emotional, sexual and social lives, as well as being linked to a number of gynaecological and obstetrical complications and adverse pregnancy outcomes. Antibiotics, such as metronidazole or clindamycin, are recommended as first-line treatment for BV, but may be associated with antibiotic resistance, high rates of recurrence and poor patient treatment satisfaction. Astodrimer sodium gel is a novel, non-antibiotic treatment for BV that is not systemically absorbed. It prevents pathogenic bacteria from adhering to the vaginal wall, and disrupts and inhibits the formation of pathogenic bacterial biofilms. Clinical cure rates of 50-57% were observed in patients with BV treated with astodrimer sodium compared with 17-21% treated with placebo (p < 0.001) in Phase 3 trials. In a separate Phase 3 trial, recurrence of BV occurred in 44% of patients treated with astodrimer sodium compared with 54% of patients who received placebo (p = 0.015). Astodrimer sodium is well tolerated, with vulvovaginal candidosis being the only treatment-related adverse event reported to occur more often than with placebo. The availability of astodrimer sodium, a well-tolerated, convenient, non-antibiotic treatment for BV, represents significant progress in the treatment of this burdensome condition.


Subject(s)
Anti-Bacterial Agents , Dendrimers , Polylysine , Vaginosis, Bacterial , Anti-Bacterial Agents/therapeutic use , Bacteria , Clindamycin/therapeutic use , Dendrimers/therapeutic use , Female , Humans , Metronidazole/therapeutic use , Polylysine/therapeutic use , Pregnancy , Vaginosis, Bacterial/drug therapy , Vaginosis, Bacterial/microbiology
5.
Viruses ; 13(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34452519

ABSTRACT

Strategies to combat COVID-19 require multiple ways to protect vulnerable people from infection. SARS-CoV-2 is an airborne pathogen and the nasal cavity is a primary target of infection. The K18-hACE2 mouse model was used to investigate the anti-SARS-CoV-2 efficacy of astodrimer sodium formulated in a mucoadhesive nasal spray. Animals received astodrimer sodium 1% nasal spray or PBS intranasally, or intranasally and intratracheally, for 7 days, and they were infected intranasally with SARS-CoV-2 after the first product administration on Day 0. Another group was infected intranasally with SARS-CoV-2 that had been pre-incubated with astodrimer sodium 1% nasal spray or PBS for 60 min before the neutralisation of test product activity. Astodrimer sodium 1% significantly reduced the viral genome copies (>99.9%) and the infectious virus (~95%) in the lung and trachea vs. PBS. The pre-incubation of SARS-CoV-2 with astodrimer sodium 1% resulted in a significant reduction in the viral genome copies (>99.9%) and the infectious virus (>99%) in the lung and trachea, and the infectious virus was not detected in the brain or liver. Astodrimer sodium 1% resulted in a significant reduction of viral genome copies in nasal secretions vs. PBS on Day 7 post-infection. A reduction in the viral shedding from the nasal cavity may result in lower virus transmission rates. Viraemia was low or undetectable in animals treated with astodrimer sodium 1% or infected with treated virus, correlating with the lack of detectable viral replication in the liver. Similarly, low virus replication in the nasal cavity after treatment with astodrimer sodium 1% potentially protected the brain from infection. Astodrimer sodium 1% significantly reduced the pro-inflammatory cytokines IL-6, IL-1α, IL-1ß, TNFα and TGFß and the chemokine MCP-1 in the serum, lung and trachea vs. PBS. Astodrimer sodium 1% nasal spray blocked or reduced SARS-CoV-2 replication and its sequelae in K18-hACE2 mice. These data indicate a potential role for the product in preventing SARS-CoV-2 infection or for reducing the severity of COVID-19.


Subject(s)
Antiviral Agents/administration & dosage , COVID-19 Drug Treatment , Dendrimers/administration & dosage , Nasal Cavity/virology , Nasal Sprays , Polylysine/administration & dosage , SARS-CoV-2/drug effects , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antiviral Agents/therapeutic use , Brain/virology , COVID-19/prevention & control , COVID-19/virology , Dendrimers/therapeutic use , Disease Models, Animal , Female , Liver/virology , Male , Mice , Mice, Transgenic , Polylysine/therapeutic use , Respiratory System/virology , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Viral Load/drug effects , Viremia , Virus Replication/drug effects
6.
ACS Appl Mater Interfaces ; 13(28): 32716-32728, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34227797

ABSTRACT

Bacterial cellulose (BC) holds several unique properties such as high water retention capability, flexibility, biocompatibility, and high absorption capacity. All these features make it a potential material for wound healing applications. However, it lacks antibacterial properties, which hampers its applications for infectious wound healings. This study reported BC-based dressings containing ε-polylysine (ε-PL), cross-linked by a biocompatible and mussel-inspired polydopamine (PDA) for promoting infectious wound healing. BC membranes were coated with PDA by a simple self-polymerization process, followed by treating with different contents of ε-PL. The resulted membranes showed strong antibacterial properties against tested bacteria by both in vitro and in vivo evaluations. The membranes also exhibited hemocompatibility and cytocompatibility by in vitro investigations. Moreover, the functionalized membranes promoted infected wound healing using Sprague-Dawley rats as a model animal. A complete wound healing was observed in the group treated with functionalized membranes, while wounds were still open for control and pure BC groups in the same duration. Histological investigations indicated that the thickness of newborn skin was greater and smoother in the groups treated with modified membranes in comparison to neat BC or control groups. These results revealed that the functionalized membranes have great potential as a dressing material for infected wounds in future clinical applications.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bandages , Cellulose/chemistry , Polylysine/therapeutic use , Staphylococcal Skin Infections/drug therapy , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/toxicity , Cellulose/toxicity , Escherichia coli/drug effects , Indoles/chemistry , Indoles/therapeutic use , Indoles/toxicity , Male , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Polylysine/analogs & derivatives , Polylysine/toxicity , Polymers/chemistry , Polymers/therapeutic use , Polymers/toxicity , Rats, Sprague-Dawley , Skin/drug effects , Skin/pathology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/drug effects , Wound Infection/drug therapy , Wound Infection/pathology
7.
J Exp Clin Cancer Res ; 40(1): 213, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34172082

ABSTRACT

Immunotherapy is currently under intensive investigation as a potential breakthrough treatment option for glioblastoma. Given the anatomical and immunological complexities surrounding glioblastoma, lymphocytes that infiltrate the brain to develop durable immunity with memory will be key. Polyinosinic:polycytidylic acid, or poly(I:C), and its derivative poly-ICLC could serve as a priming or boosting therapy to unleash lymphocytes and other factors in the (immuno)therapeutic armory against glioblastoma. Here, we present a systematic review on the effects and efficacy of poly(I:C)/poly-ICLC for glioblastoma treatment, ranging from preclinical work on cellular and murine glioblastoma models to reported and ongoing clinical studies. MEDLINE was searched until 15 May 2021 to identify preclinical (glioblastoma cells, murine models) and clinical studies that investigated poly(I:C) or poly-ICLC in glioblastoma. A systematic review approach was conducted according to PRISMA guidelines. ClinicalTrials.gov was queried for ongoing clinical studies. Direct pro-tumorigenic effects of poly(I:C) on glioblastoma cells have not been described. On the contrary, poly(I:C) changes the immunological profile of glioblastoma cells and can also kill them directly. In murine glioblastoma models, poly(I:C) has shown therapeutic relevance as an adjuvant therapy to several treatment modalities, including vaccination and immune checkpoint blockade. Clinically, mostly as an adjuvant to dendritic cell or peptide vaccines, poly-ICLC has been demonstrated to be safe and capable of eliciting immunological activity to boost therapeutic responses. Poly-ICLC could be a valuable tool to enhance immunotherapeutic approaches for glioblastoma. We conclude by proposing several promising combination strategies that might advance glioblastoma immunotherapy and discuss key pre-clinical aspects to improve clinical translation.


Subject(s)
Brain Neoplasms/drug therapy , Carboxymethylcellulose Sodium/analogs & derivatives , Glioblastoma/drug therapy , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Animals , Brain Neoplasms/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/therapeutic use , Clinical Trials as Topic , Glioblastoma/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Mice , Polylysine/therapeutic use
8.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33751208

ABSTRACT

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.


Subject(s)
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
9.
Minerva Med ; 112(1): 144-152, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33104300

ABSTRACT

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.


Subject(s)
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
10.
Inflammation ; 44(1): 174-185, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32803665

ABSTRACT

Cytokines like IL-6, TNF-α, and IL-1ß are important mediators of inflammation in many inflammatory diseases, as well as in cellular processes like cell proliferation and cell adhesion. Finding new molecules that decrease cell proliferation, adhesion (inflammatory infiltrate), and pro-inflammatory cytokine release could help in the treatment of many inflammatory diseases. The naturally derived poly(gallic acid) (PGAL), produced enzymatically from gallic acid in aqueous medium, is a non-toxic, thermostable multiradical polyanion that is antioxidant and has potential biomedical uses. Experimental evidence has demonstrated that PGAL reduces pro-inflammatory cytokines, which are the target of some inflammatory diseases. PGAL decreased IL-6, TNF-α, and IL-1ß production in human monocytes exposed to PMA without affecting cell viability. Additionally, PGAL reduced cell proliferation by affecting the transition from the S phase to the G2 phase of the cell cycle. Cell adhesion experiments showed that PMA-induced cell adhesion was diminished with the presence of PGAL, particularly at a concentration of 200 µg/mL. These properties of PGAL show a potential use for treating inflammatory diseases, such as psoriasis or arthritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Polyglutamic Acid/analogs & derivatives , Polylysine/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Humans , Inflammation/drug therapy , Inflammation/metabolism , Polyglutamic Acid/pharmacology , Polyglutamic Acid/therapeutic use , Polylysine/pharmacology , Polylysine/therapeutic use , THP-1 Cells
11.
J Mater Chem B ; 8(41): 9492-9501, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33001118

ABSTRACT

Developing new strategies to enhance drug accumulation in the tumor and therapeutic efficacy is of great importance in the field of tumor therapy. Herein, a peanut-like multifunctional nanomedicine (CuS-PGH NMs) made of CuS nanoparticles encapsulated in poly(l-lysine)(PLL)/glucose oxidase (GOx)-hyaluronic acid (HA) shells has been constructed via layer-by-layer (LbL) assembly, and shows good biocompatibility and effective multi-gradient therapy. Because of the enhanced permeability and retention (EPR) effect, the CuS-PGH NMs could significantly enhance the cellular uptake by tumors overexpressing CD44 receptors, which respond to hyaluronidase (HAase)-triggered surface charge conversion. Once internalized by the tumor, GOx was the first to be exposed and could effectively deplete endogenous glucose for starvation therapy, and the excess H2O2 was then converted into highly toxic hydroxyl radicals (˙OH) via a Cu+-mediated Fenton-like reaction for chemodynamic therapy (CDT). Meanwhile, the as-obtained Cu+ ions accompanied the regenerated less-active Cu2+ ions. Interestingly, the high content of H2O2 could, in turn, accelerate Cu2+/Cu+ conversion to promote the Cu+-H2O2 reaction for enhanced chemodynamic therapy (CDT), thereby achieving efficient tumor growth suppression via synergistic starvation/CDT therapy. Subsequently, owing to the strong NIR-II absorption capability of CuS-PGH NMs, effective photothermal tumor ablation of the weakened tumor cells could be realized with the precise guidance of NIR-II PAI. This multi-gradient therapeutic strategy has been demonstrated to have excellent antitumor activity with minimal nonspecific damages, and offers a new avenue to precise tumor therapy.


Subject(s)
Breast Neoplasms/therapy , Nanoparticles/therapeutic use , Theranostic Nanomedicine/methods , Animals , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Copper/therapeutic use , Female , Glucose Oxidase/therapeutic use , Hyaluronic Acid/therapeutic use , Mice , Photoacoustic Techniques , Polylysine/therapeutic use
12.
Semin Immunol ; 49: 101414, 2020 06.
Article in English | MEDLINE | ID: mdl-33011064

ABSTRACT

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Carboxymethylcellulose Sodium/analogs & derivatives , Immunologic Factors/pharmacology , Immunomodulation , Poly I-C/immunology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Carboxymethylcellulose Sodium/pharmacology , Carboxymethylcellulose Sodium/therapeutic use , Cytokines/metabolism , Humans , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Immunomodulation/drug effects , Interferon-Induced Helicase, IFIH1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Poly I-C/therapeutic use , Polylysine/immunology , Polylysine/pharmacology , Polylysine/therapeutic use , Receptors, Pattern Recognition/metabolism , Toll-Like Receptor 3/metabolism
13.
Urol Clin North Am ; 47(4S): e1-e8, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33446322

ABSTRACT

Relatively simple, synthetic, double-stranded RNAs can be powerful viral pathogen-associated molecular pattern (PAMP) mimics, inducing a panoply of antiviral and antitumor responses that act at multiple stages of host defense. Their mechanisms of action and uses are beginning to be understood, alone, in combination with other therapeutics, or as novel PAMP-adjuvants providing the critical danger signal that has been missing from most cancer and other modern vaccines. Dose, timing, route of administration combinations, and other clinical variables can have a critical impact on immunogenicity. This article reviews advances in the use of polyinosinic-polycytidylic acid and derivatives, in particular poly-ICLC.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Immunologic Factors/therapeutic use , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Prostatic Neoplasms/therapy , RNA, Double-Stranded/therapeutic use , Adjuvants, Immunologic/physiology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/therapeutic use , Clinical Trials as Topic , Humans , Immunologic Factors/immunology , Male , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/therapeutic use , Poly I-C/immunology , Polylysine/immunology , Polylysine/therapeutic use , Prostatic Neoplasms/immunology , RNA, Double-Stranded/immunology
14.
Nano Lett ; 19(8): 5515-5523, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31362507

ABSTRACT

Designing simple-structured nanomedicine without lacking key functionalities, thereby avoiding incomplete damage or relapse of tumor with the administration of a safe dose, is pivotal for successful cancer nanotherapy. We herein presented a nanomedicine of photodynamic therapy (PDT) that simply assembled amphiphilic macromolecules of poly-l-lysine conjugating with photosensitizers onto hydrophobic upconverting nanoparticles. We demonstrated that the nanoformulation, despite its simple structure and synthesis, simultaneously possesses multiple features, including substantial payload of photosensitizers, avid cellular internalization both in vitro and in vivo, efficient diffusion and broad distribution in tumor lesion, and potent fatality for cancer stem cells that are refractory to other therapy modalities. Because of the combination of these functionalities, the tumors in mice were eradicated and no relapse was observed after at least 40 days, just with an extremely low intraperitoneal injection dose of 5.6 mg/kg. Our results suggested a strategy for designing multifunctional nanomedicines with simple construct and efficacious therapeutic response and presented the promising potential of PDT for a radical cure of cancer.


Subject(s)
Nanoconjugates/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Animals , Cell Line, Tumor , HeLa Cells , Humans , Mice , Nanoconjugates/administration & dosage , Nanoconjugates/chemistry , Neoplasm Recurrence, Local/prevention & control , Neoplastic Stem Cells/drug effects , Photochemotherapy , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Polylysine/administration & dosage , Polylysine/analogs & derivatives , Polylysine/therapeutic use
15.
Nat Med ; 25(5): 814-824, 2019 05.
Article in English | MEDLINE | ID: mdl-30962585

ABSTRACT

Indolent non-Hodgkin's lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial ( NCT01976585 ). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.


Subject(s)
Cancer Vaccines , Lymphoma, B-Cell/therapy , Adult , Aged , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Carboxymethylcellulose Sodium/analogs & derivatives , Carboxymethylcellulose Sodium/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/immunology , Female , Humans , Immunotherapy, Adoptive , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Male , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Polylysine/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Toll-Like Receptor 3/agonists , Vaccination
16.
Front Immunol ; 10: 725, 2019.
Article in English | MEDLINE | ID: mdl-31024557

ABSTRACT

Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n = 15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4 mg subcutaneously) vs. placebo. Subjects were observed for adverse events, immune activation, and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA, and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA, and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 h after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: www.ClinicalTrials.gov, identifier: NCT02071095.


Subject(s)
Carboxymethylcellulose Sodium/analogs & derivatives , HIV Infections/drug therapy , HIV Infections/immunology , Immunity, Innate/drug effects , Poly I-C/therapeutic use , Polylysine/analogs & derivatives , Toll-Like Receptor 3/metabolism , Adult , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carboxymethylcellulose Sodium/therapeutic use , Double-Blind Method , Female , HIV/immunology , HIV Infections/metabolism , Humans , Male , Middle Aged , Polylysine/therapeutic use
17.
Antiviral Res ; 163: 179-184, 2019 03.
Article in English | MEDLINE | ID: mdl-30611774

ABSTRACT

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.


Subject(s)
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
18.
BMB Rep ; 52(5): 324-329, 2019 May.
Article in English | MEDLINE | ID: mdl-30293549

ABSTRACT

Recent progress in cellular reprogramming technology and lineage-specific cell differentiation has provided great opportunities for translational research. Because virus-based gene delivery is not a practical reprogramming protocol, protein-based reprogramming has been receiving attention as a safe way to generate reprogrammed cells. However, the poor efficiency of the cellular uptake of reprogramming proteins is still a major obstacle. Here, we reported key factors which improve the cellular uptake of these proteins. Purified red fluorescent proteins fused with 9xLysine (dsRED-9K) as a cell penetrating peptide were efficiently delivered into the diverse primary cells. Protein delivery was improved by the addition of amodiaquine. Furthermore, purified dsRED-9K was able to penetrate all cell lineages derived from mouse embryonic stem cells efficiently. Our data may provide important insights into the design of protein-based reprogramming or differentiation protocols [BMB Reports 2019; 52(5): 324-329].


Subject(s)
Cell-Penetrating Peptides/metabolism , Cellular Reprogramming Techniques/methods , Polylysine/metabolism , Amodiaquine/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/genetics , Cell-Penetrating Peptides/pharmacology , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Gene Transfer Techniques , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Peptides/therapeutic use , Polylysine/therapeutic use , Transcription Factors/metabolism
19.
Kidney Blood Press Res ; 43(4): 1273-1284, 2018.
Article in English | MEDLINE | ID: mdl-30078011

ABSTRACT

BACKGROUND/AIMS: Myocardial ischemia/reperfusion (I/R) injury (MI/RI) is a critical cause of death in patients with heart disease. However, the pharmaco-therapeutical outcome for MI/RI remains unsatisfactory. Innovative approaches for enhancing drug sensitivity and recovering myocardial function in MI/RI treatment are urgently needed. The purpose of this study was to evaluate the protective effects of exenatide-loaded poly(L-lysine)-poly(ethylene glycol)-poly(L-lysine) (PLL-PEG-PLL) nanoparticles (NPs) against MI/RI. METHODS: The size of PLL-PEG-PLL NPs and the loading and release rates of exenatide were determined. The in vitro NP cytotoxicity was evaluated using newborn rat cardiomyocytes. Rats pretreated with free exenatide or exenatide/PLL-PEG-PLL polyplexes were subjected to 0.5-h ischemia and 2-h reperfusion in the left anterior descending coronary artery. The histopathologic lesions were assessed using hematoxylin-eosin staining. The general physiological indices, including blood pressure (BP), heart rate (HR), the left ventricular ejection fraction (LVEF) and end-diastolic pressure (LEVDP), and the left ventricular pressure maximal rate of rising (dp/dtmax), were monitored using a non-invasive blood pressure analyzer and color Doppler echocardiography. The antioxidative activity in the myocardial tissue was measured. The myocardial enzymatic activity was further estimated by determining the serum levels of creatine kinase (CK), lactate dehydrogenase (LDH), cardiac troponin T (cTnT), and glucagon-like peptide-1 (GLP-1), as well as the expression of GLP-1R in the myocardial tissue. RESULTS: Exenatide preconditioning attenuated the oxidative stress injury and promoted the myocardial function in I/R-induced myocardial injury, while the application of block copolymer PLL-PEG-PLL as a potential exenatide nanocarrier with sustained release significantly enhanced the bioavailability of exenatide. CONCLUSION: The block copolymer PLL-PEG-PLL may function as a potent exenatide nanocarrier for augmenting pharmacotherapy against MI/RI with unprecedented clinical benefits. Further study is needed to better clarify the underlying mechanisms.


Subject(s)
Ischemic Preconditioning, Myocardial/methods , Myocardial Reperfusion Injury/drug therapy , Nanoparticles/chemistry , Peptides/pharmacology , Venoms/pharmacology , Animals , Animals, Newborn , Drug Carriers/chemistry , Exenatide , Hypoglycemic Agents , Incretins , Male , Oxidative Stress/drug effects , Peptides/pharmacokinetics , Peptides/therapeutic use , Polyethylene Glycols/therapeutic use , Polylysine/analogs & derivatives , Polylysine/therapeutic use , Rats , Rats, Sprague-Dawley , Venoms/pharmacokinetics , Venoms/therapeutic use
20.
Theranostics ; 8(11): 2974-2987, 2018.
Article in English | MEDLINE | ID: mdl-29896297

ABSTRACT

Although there is ample evidence that the chemotherapeutic drugs trigger an immune response, the efficient tumor rejection or regression is not guaranteed probably due to the massive immunosuppression within the tumor microenvironment. Thus, a rational delivery platform that overcomes immunosuppression is needed to maximally achieve both cytotoxic and immune-modulatory functions of chemotherapeutics. Accumulating evidence suggests that platinum-based drugs might be suitable for this application. Methods: The dendrigraft polylysine (DGL) with its uniform size and multifunctional groups was employed as the polymeric core and conjugated with platinum-based compounds as therapeutics and WKYMVm peptide (Wpep) as a targeting ligand to construct the novel delivery platform Wpep-DGL/Pt. A series of in vitro and in vivo analyses, including physical and chemical characterizations, targeting property, biosafety, and antitumor efficacy of Wpep-DGL/Pt were systematically carried out. Results: Wpep-DGL/Pt showed potent antitumor efficacy in MDA-MB-231 cells tumor-bearing nude mice with a deficient immune system, demonstrating targeted delivery of chemotherapeutics and the resultant cytotoxicity. Furthermore, in immunocompetent mice bearing 4T1 cells tumors, Wpep-DGL/Pt activated immune cells and induced cell death proving their dual function of chemotherapeutic and immunomodulatory efficacy. Conclusion: This work represents a novel approach for cancer immunotherapy by integrating nanotechnology and platinum-based therapeutics which not only efficiently exerts the chemotherapeutic cytotoxic effect on tumor cell but also restores immune response of immunological cells within the tumor microenvironment.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/therapy , Peptides/therapeutic use , Platinum/therapeutic use , Adjuvants, Immunologic/chemistry , Animals , Antineoplastic Agents/chemistry , Drug Compounding , Drug Delivery Systems , Female , Immunity, Humoral , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles , Neoplasms/immunology , Peptides/chemistry , Platinum/chemistry , Polylysine/chemistry , Polylysine/therapeutic use , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...