WO2017175132A1 - The use of cationic derivative of dextran for inhibition of herpes simplex viruses - Google Patents

The use of cationic derivative of dextran for inhibition of herpes simplex viruses Download PDF

Info

Publication number
WO2017175132A1
WO2017175132A1 PCT/IB2017/051918 IB2017051918W WO2017175132A1 WO 2017175132 A1 WO2017175132 A1 WO 2017175132A1 IB 2017051918 W IB2017051918 W IB 2017051918W WO 2017175132 A1 WO2017175132 A1 WO 2017175132A1
Authority
WO
WIPO (PCT)
Prior art keywords
hsv
virus
dextran
cells
polymer
Prior art date
Application number
PCT/IB2017/051918
Other languages
French (fr)
Inventor
Maria Nowakowska
Krzysztof Pyrc
Krzysztof Szczubialka
Justyna CIEJKA
Aleksandra MILEWSKA
Magdalena PACHOTA
Original Assignee
Uniwersytet Jagiellonski
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Uniwersytet Jagiellonski filed Critical Uniwersytet Jagiellonski
Publication of WO2017175132A1 publication Critical patent/WO2017175132A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/721Dextrans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses

Definitions

  • the subject of the invention is the use of dextran polymer for the prophylaxis and treatment of infections caused by human herpesvirus.
  • Herpes simplex viruses type 1 and 2 (HSV-1 and HSV-2, respectively) belong to the Herpesviridae family encompassing more than 100 viruses, eight of which affect humans 1 . They are also widespread in human population, as it is estimated that 50-90% of the population worldwide may be infected 2 .
  • HSV-1 is mainly associated with the watery blisters in the mouth area (lips, tongue, oral mucosa), while HSV-2 mostly affects genital region 3 4 . Both of them are, however, also associated with potentially fatal viral encephalitis and stromal keratitis, an ocular disease, which is a leading cause of cornea-derived blindness in developed countries 5"7 .
  • the HSV virion is a spherical, complex structure, about 200 nm in diameter.
  • the inner core, containing the viral DNA stabilized by polyamines, is encapsulated by an icosahedral capsid, which is further coated with an unstructured protein layer, called the tegument.
  • the whole structure is surrounded by a lipid bilayer decorated with viral surface glycoproteins.
  • HSV is a linear dsDNA virus, with genome of 152 and 155 kbp for HSV-1 for HSV-2, respectively. Two major genomic regions - L (long) and S (short) - both flanked with inverted repeats may be distinguished.
  • the HSV genome encodes for at least 90 transcriptional units, which are sorted into 3 classes: a - immediate early, ⁇ - early, and ⁇ - late genes.
  • the a genes are located in the inverted repeats of both L and S components and encode for proteins regulating expression of early and late genes.
  • the ⁇ and ⁇ genes are distributed within the L and S region
  • HSV infection begins with epithelial cells, usually oral or ocular for HSV-1 and genital for HSV-2.
  • the virus is easily spread through direct contact, i.e., kissing or sexual intercourse.
  • HSV proceeds to enter nerve endings and, employing retrograde transport, makes its way to the dorsal root neurons nuclei, where it establishes latency - a state of lifelong infection.
  • This possibility a hallmark of the Herpesviridae family, is facilitated by a failure of immediate early genes transcription caused by ineffective axonal transport of viral regulation factors, such as VP 16 and IFN-a activity in sensory neurons 9 10 .
  • the virus Once the virus reaches the latent state, the infection seems to be incurable and may be reactivated, either spontaneously or by stress stimuli, such as fever, UV exposure, or hormonal changes u .
  • HSV vaccine No effective HSV vaccine is available, and therefore current research is oriented towards the development of antiviral compounds limiting the primary infection and supporting further treatment.
  • the approved anti-HSV therapies involve mainly nucleoside analogues, such as acyclovir (ACV), which interfere with viral DNA synthesis reducing virus replication and shedding.
  • ACCV acyclovir
  • Low bioavailability of this drug led to the development of derivatives with better pharmacokinetic parameters, e.g., selenoaciclovir, valacyclovir, famciclovir, and ganciclovir 12"14 .
  • Most of these drugs undergo bioconversion within the cell to ACV.
  • polymers for inhibition of HSV infection was proposed before and involved synthetic polycations and polyanions, as well as peptides/proteins.
  • the examples of the polycations which showed anti-HSV activity are poly-L-lysine and poly-L-arginine derivatives were shown to inhibit early stages of HSV replication 18 , poly(amidoamines) substituted with agmatine 19 , viologen dendrimers 20 , and a copolymer of methyl methacrylate, N,N-dimethylaminoethyl methacrylate, and butyl methacrylate (Eudragit®, El 00) which destabilized virus membrane 21 .
  • polyanions which inhibit HSV were dendrimers containing sulfonic groups 22 , poly(sodium styrene sulfonate) (PSSS) 23 and its copolymer with maleic acid 24 , and telomerized co-acryloyl anionic surfactants 25 .
  • PSSS poly(sodium styrene sulfonate)
  • telomerized co-acryloyl anionic surfactants 25 telomerized co-acryloyl anionic surfactants.
  • natural polymers having anti-HSV activity were sulfated lignins 26 , sulfated cellulose nanocrystals (CNC) 27 , peptides and aptamers, which interfered with virus - cell interactions 28"30 , and cationic polysaccharides conjugated with oligoamines via reductive amination 31 .
  • dextran substituted with amine groups in particular dextran-propyl- 1,3 -diamine, effectively inhibits HSV-1 virus replication, while this compound modified by the conversion of a primary amine group to a trimethylammonium group (monoquaternary ammonium propyl-1,3- diamine, MQ-propane- 1,3 -diamine) did not show this activity 31 , in complete contrast to structurally similar cationic derivatives of dextran being the object of the present application.
  • amine groups in particular dextran-propyl- 1,3 -diamine
  • entry inhibitors include also sulfonated galactans and fucoidans, dextran sulfate, heparin, and sulfonated lignins.
  • EP0066379 describes a composition for combating HSV type I and II, comprising a dextran sulfate sodium salt with molecular weight in the range of 4100 to 25000 and containing at least 0.2 equivalent of S0 3 Na per glucopyranosyl unit.
  • WO2000024784 application discloses dextran derivatives showing biological activity towards HSV, namely dextranpolyaldehyde-resorcine.
  • the aim of the invention described in the present application is to provide a new effective drug for the treatment and prophylaxis of infections caused by herpes simplex viruses.
  • the subject of the invention is the use of dextran cationically modified by quaternary ammonium groups of structure I for the treatment and prophylaxis of diseases caused by HSV-1 virus.
  • dextran modified by glycidyltrimethylammonium chloride (GTMAC) is used.
  • GTMAC glycidyltrimethylammonium chloride
  • Cationically modified dextran has the degree of substitution in the range of 20-100%, preferably 20-60%, more preferably 25-60%.
  • the use includes polymers based on dextran in the form of ointment or drops applied topically to the skin or to the eye, orally, intraperitoneally, or intravenously in the systemic treatment.
  • Fig. 1 shows IR-ATR spectrum of cationically modified dextrans with the weight of 6 kDa
  • Fig. 2 is IR-ATR spectrum of cationically modified dextrans with the weight of 40 kDa
  • Fig. 3 is IR-ATR spectrum of cationically modified dextrans with the weight of 100 kDa
  • Fig. 4 illustrates the cytotoxicity of polymers. Cell viability is shown as % of the value for the control sample, i.e. not treated with polymers.
  • Fig. 5 shows the antiviral activity of GTMAC-modified dextrans.
  • Fig. 6 illustrates the relation between the anti-HSV-1 activity and the degree of substitution.
  • Fig. 7 presents cytotoxicity of the tested polymers.
  • Fig. 8 shows influence of DS and MW of DEXxDSy on their anti-HSV-1 activity.
  • Fig. 9 shows concentration-dependent antiviral activity of DEX100DS42 against HSV-1
  • Fig. 10 shows inhibition of HSV-1 replication cycle at different stages.
  • Fig. 11 shows UV-Vis absorption spectra of Azure A in the presence of 0.2 mg/ml of HS and different concentrations of DEX100DS42
  • Fig. 12 illustrates dependence of the relative concentration of free (unbound) HS on the weight ratio of DEX100DS42 and HS.
  • Fig. 13 presents HSV-1 attachment in the presence of DEX100DS42, as determined with flow cytometry.
  • Fig. 14 shows modified dextrans block HSV-1 attachment to susceptible cells.
  • Fig. 15 shows subcellular localization of dextran derivatives.
  • the solution was transferred to a dialysis tube with the molecular weight cutoff value of 3.5 kDa and the dialysis was carried out against water until the conductivity of the said solution decreased below 10 ⁇ 8. Obtained polymers were isolated with freeze-drying. These were denoted by acronyms in which the first number is the molecular weight in kilodaltons, while the second is the degree of substitution (DS) in percentage.
  • Dextran and its cationic derivatives were subjected to the elemental analysis in order to determine the degree of substitution (DS) of glucose units with GTMAC groups, defined as a number of GTMAC groups per 1 glucose unit in dextran. The results are reported in Table 2.
  • Zeta potential of cationic derivatives of dextran Zeta potential was measured with NanoZetasizer instrument from Malvern. The measurements were performed for samples at the concentration of 1 mg/ml in 0.015 M NaCl. The results are reported in Table 4.
  • the cytotoxicity of cationic derivatives of dextran was determined using XTT assay.
  • the assay was performed on Vero E6 cell line which is permissive for HSV-1 virus. This assay is based on the determination of mitochondrial enzymes' activity by testing the ability thereof to reduce a substrate to a colored product.
  • the cell viability was determined with measuring the absorbance at 450 nm in relation to the control which was represented by cells cultured in medium without additives.
  • Cells were cultured for 2 days in a mixture of MEM medium, with the addition of Hank's salt, and M199 medium, with the addition of Eagle's medium in 2: 1 ratio, supplemented with 3% heat-inactivated fetal bovine serum (FBS), as well as penicillin and streptomycin, with the addition of the tested polymer.
  • MEM medium with the addition of Hank's salt, and M199 medium, with the addition of Eagle's medium in 2: 1 ratio, supplemented with 3% heat-inactivated fetal bovine serum (FBS), as well as penicillin and streptomycin, with the addition of the tested polymer.
  • FBS heat-inactivated fetal bovine serum
  • Results presented in Fig. 4 show the cytotoxicity of the synthesized polymers at various concentrations.
  • HSV-1 virus replication Upon the determination of compound concentrations without toxic effect on cells, their effect on HSV-1 virus replication was tested.
  • Fig. 5 shows the antiviral activity of GTMAC-modified dextrans.
  • GTMAC- modified dextrans with various molar weight (6 , 40, and 100 kDa) were used. Suitable unmodified dextrans were used as controls (ctrl). For all compounds the concentration used was 500 ⁇ g/ml.
  • Fig. 6 shows the relation between the anti-HSV-1 activity and the degree of substitution. Three different degrees of polymer substitution were used: 23%, 37%, and 52%. The presented diagram clearly shows that the antiviral activity of the polymer correlates with the degree of substitution.
  • DEXxDSy Cationic derivatives of dextran (DEXxDSy) as effective inhibitors of HSV-1 and HSV-2 infection.
  • Dextran (DEX, MW of 6, 40 and 100 kDa, from Leuconostoc spp., Sigma), glycidyltrimethylammonium chloride (GTMAC, >90% Sigma), heparan sulfate (HS), Azure A chloride (Fluka, Fluka standard), PBS (tablet, Sigma), fluorescein 5(6)-isotiocyanate (FITC, >90% (HPLC), Sigma), sodium chloride (analytical grade, POCh), NaOH (analytical grade, POCh), HC1 (pure, POCh) were used as received. Water was distilled twice and deionized using the Millipore Simplicity system.
  • FTIR spectra were obtained on a Bruker IFS 48 spectrometer. Elemental analysis (EA) was performed on a EuroEA 3000 Elemental Analyzer. The UV/VIS absorption spectra were obtained at room temperature in 1-cm quartz cuvettes using the single beam diode array Hewlett-Packard 8452A spectrophotometer with a resolution of 2 nm in the range of 190-820 nm. Zeta potential measurements were performed using Zetasizer Nano ZS instrument (Malvern Instruments). The sample was illuminated with a 633-nm laser and the intensity of light scattered at an angle of 173° was measured with an avalanche photodiode.
  • the products are denoted as DEXxDSy where x is the MW of DEX in kDa and y is the degree of substitution defined as the number of GTMAC groups attached to DEX per 100 glucose repeating units.
  • x is the MW of DEX in kDa
  • y is the degree of substitution defined as the number of GTMAC groups attached to DEX per 100 glucose repeating units.
  • Table S2 The first polymer sample in each series was obtained by reacting 2 g of DEX dissolved in 100 ml of distilled water with initial volume of GTMAC (20 or 40 ml) for 1 or 2 h. Then, a defined volume of the reaction mixture was withdrawn to isolate a polymer sample while to the rest of the reaction mixture the defined volume of GTMAC was added and the resulting mixture was heated while mixing for the defined time period.
  • the withdrawal/GTMAC addition/heating cycles were repeated twice or thrice.
  • To isolate the polymer the reaction mixtures were neutralized to pH ⁇ 7 by the addition of 1 M HC1, cooled and transferred to dialysis tubes (MW cutoff values of 3.5, 12.8 and 14 kDa, depending of the MW of DEX). The dialysis was carried out against distilled water until the conductivity of the liquid surrounding the tube decreased below 10 ⁇ 8.
  • the polymers obtained were isolated from the solution using the freeze-drying technique.
  • the substitution of DEX with GTMAC was confirmed using EA, IR spectroscopy and zeta potential measurements.
  • the DS values of the polymers obtained covered almost the whole possible range, i.e. from 2 to 98.
  • DEX6DS41, DEX40DS37 and DEX100DS40 were fluorescently labeled as described previously 33 . Briefly, 50 mg of the respective polymer was dissolved in 2 ml of warm DMSO and 1 drop of pyridine and 3 mg of FITC in 2 ml of DMSO were added. The mixture was heated to 95°C for 2 hours under continuous stirring. Then, DMSO was removed by dialysis against water and the product was lyophilized.
  • the DS of the DEXxDSy was determined with elemental analysis (EA).
  • EA elemental analysis
  • the ratio of N/C in each polymer was determined and the DS was calculated using the following equation:
  • Vero E6 cells (Cercopithecus aethiops kidney epithelial, ATCC: CRL-1586) were routinely maintained under Dulbecco-modified Eagle's medium (DMEM, high glucose, Life Technologies) supplemented with 3% heat-inactivated fetal bovine serum (FBS, Life Technologies), penicillin (100 U/ml), and streptomycin (100 ⁇ g/ml) at 37°C in atmosphere containing 5% C0 2 .
  • DMEM Dulbecco-modified Eagle's medium
  • FBS heat-inactivated fetal bovine serum
  • penicillin 100 U/ml
  • streptomycin 100 ⁇ g/ml
  • HSV-1 strain 17+ and HSV-2 strain HG52 were obtained from Public Health England (0104151v and 0104152v, respectively).
  • Virus stocks were generated by infecting Vero E6 cells monolayers for 48 h, then lysing the cells with two freeze-thaw cycles. Collected lysates were aliquoted and stored at -80°C. Mock samples were prepared in the same manner, using mock-infected cells. Virus stocks were quantified by titration on Vero E6 cells (48 h infection, 37°C), according to Reed and Muench method 34 . Following a 48 h incubation the number of cytopathic effect (CPE)-positive wells was counted and the TCID50 was calculated.
  • CPE cytopathic effect
  • a series of assays was employed to determine the mechanism of antiviral activity of DEXxDSy.
  • the tests differ mainly in the sequence in which the cells, the virus and the polymer may interact.
  • Virus stock samples were incubated with DEXxDSy polymers (500 ⁇ g/ml) for 1 h at 22°C under constant mixing. Subsequently, the samples were diluted to decrease the polymer concentration below its active range (25 ⁇ g/ml). In negative control samples cell culture medium was added instead of the polymer solution. The samples were then titrated on fully confluent Vero E6 cells to assess viral yield. This test indicates whether the antiviral effect results from a direct interaction between the polymer and the virus.
  • Viral DNA was isolated from supernatants using Viral DNA/RNA Isolation Kit (A&A Biotechnology, Tru). Virus yield was then determined by quantitative real-time PCR (qPCR). The reaction was carried out in a CFX96 TouchTM Real-Time PCR Detection System (Bio-Rad), in 10 ⁇ reaction mixture consisting of 1 x Kapa Probe Fast qPCR Master Mix, specific probe labeled with 6-carboxyfluorescein (FAM) and Black Hole Quencher 1 (BHQ1) (sequence: 5' FAM CCG CCG AAC TGA GCA GAC ACC CGC GC BHQ1 -3 ', 100 nM), and primers (450 nM each, sequences: sense primer 5 '-CAT CAC CGA CCC GGA GAG GGA C-3', anti-sense primer 5' GGG CCA GGC GCT TGT TGG TGT A-3') and 2.5 ⁇ of viral DNA in 10 ⁇ .
  • qPCR quantitative real-time PCR
  • the temperature profile included 3 min at 95°C, followed by 37 cycles of 2 s at 95°C and 20 s at 60°C.
  • DNA standards were prepared. Briefly, fragment of DNA polymerase gene, conserved among HSV-1 and HSV-2 strains, was amplified using the primers mentioned above and cloned into pTZ57R/T (Thermo Scientific, Tru) plasmid using InsTAclone PCR cloning kit (Thermo Scientific, Poland). The plasmid was propagated in E. coli TOP 10 (Life Technologies, Poland), purified with GeneJET Plasmid Miniprep Kit (Thermo Scientific, Poland) and linearized by digestion with Kpnl restriction enzyme. Concentration of the linearized DNA was determined spectrophotometrically and the number of copies per milliliter was calculated. Six subsequent serial dilutions were then used as qPCR reaction template.
  • Vero E6 cells were seeded in 6-well plates and cultured for 2 days at 37°C in atmosphere containing 5% CO2. For analysis, cells were trypsinized and fixed with 4% PFA in 1 x PBS for 20 min. Subsequently, the cells were permeabilized by 20 min incubation in 0.1% Triton X100 in 1 x PBS and non-specific binding was blocked by overnight incubation with 5% BSA in 1 x PBS at 4°C.
  • Protein labelling was carried out by 2 h incubation with primary rabbit anti-HSV VP5 antibody (20-HR50, Fitzgerald Industries, USA) diluted 1 :500, followed by lh incubation with secondary goat anti-rabbit antibody, conjugated with AlexaFluor 488 (A11001, Invitrogen, Poland). After washing with 0.1% Tween-20 in 1 x PBS, the cells were re-suspended in 1 x PBS and analyzed with FACS Calibur (Becton Dickinson) using Cell Quest software. Confocal microscopy
  • Vero E6 cells were seeded on coverslips in 6-well plates and cultured for 2 days at 37°C with 5% C0 2 . For analysis of virus adhesion and entry, Assays II and III were carried out, respectively. Immediately after washing away unbound virions, cells were fixed with 4% PFA in 1 x PBS for 20 min. Subsequently, the cells were permeabilized by 20 min incubation in 0.1% Triton X100 in 1 x PBS and non-specific binding sites were blocked by overnight incubation in 5% BSA in 1 x PBS at 4°C.
  • virus particles were incubated for 2 h with primary rabbit anti-HSV VP5 antibody (20-HR50, Fitzgerald Industries, USA) diluted 1 :500 in 1 x PBS with 0.5% Tween 20, followed by 1 h incubation with secondary goat anti -rabbit antibody, conjugated with AlexaFluor 488 (A11001, Invitrogen, Poland). F-actin was co-stained with AlexaFluor 633 -conjugated phalloidin (Invitrogen, Poland). Nuclear DNA was labelled with 0.1 ⁇ g/ml 4',6'-diamidino-2-phenylindole (DAPI) (Sigma- Aldrich, Poland).
  • DAPI 4',6'-diamidino-2-phenylindole
  • cytotoxicity of DEXxDSy was determined using XTT assay, which relies on the ability of mitochondrial enzymes to convert the substrate (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) into colored formazan salts.
  • the assay was performed on Vero E6 cell line which is an accepted model for HSV-1 and HSV-2 infection. Briefly, cells were incubated for 48 hours in the presence of different DEXxDSy polymers. Results presented in Fig. 7 clearly show that the polymers of lower MW are characterized by lower toxicity.
  • Fig. 7 cell viability was assessed using XTT assay 2 days p.i. Background fluorescence was subtracted for all the samples. Cell viability was calculated in reference to untreated cells (control). Derivatives of dextran with MW of 6 kDa (A), 40 kDa (B) and 100 kDa (C) with various DS were tested in the range of concentrations 50 - 2000 ⁇ g/ml. Results are presented as average ⁇ SD. Similar results were obtained in at least three independent experiment.
  • HSV-1 encodes several glycoproteins responsible for cell surface binding and virus entry. Glycoproteins gB and gC are responsible for initial binding of the virus at cell surface, involving HS proteoglycans (HSPGs). Virions devoid of these glycoproteins demonstrate significantly impaired infectivity 35 . gB was also found to bind PILR-a, a particle considered to be a HSV-1 coreceptor 31 . Glycoprotein D (gD) is considered to be the main entry mediator and several receptors for gD have been identified.
  • HSPGs HS proteoglycans
  • nectin-1 and nectin-2 The most important are nectin-1 and nectin-2, herpesvirus entry mediator (HVEM) and 3-0 sulfated HSPG 38-41 .
  • HVEM herpesvirus entry mediator
  • the in vivo experiments carried out using single and double knockout mice have demonstrated the dependence of HSV-2 on nectin-1 for entry and spread 42 and HSV-1 on HSPG receptor for their entry to the cells and spread.
  • the in vitro experiments carried out using human corneal fibroblasts have shown that, unlike HSV-1, HSV- 2 does not use the HSPG receptor for cell penetration 43 .
  • Another two glycoproteins taking part in HSV-1 entry, gH and gL form a heterodimer, which dissociates upon binding to its receptor, ⁇ - or avP8-integrin. This interaction induces a change in gH conformation which induces virus entry into the cell and directs the virus to acidic endosomes 44 .
  • Fig. 8 demonstrates the decreased virus replication in the presence of both cationically modified 40 kDa DEX (DEX40DSy) and in the presence of cationically modified 100 kDa DEX (DEXlOODSy), for polymers with DS > -20-40%.
  • the anti-HSV-1 activity of both polymers was similar in this assay.
  • cationically modified 6 kDa DEX showed no significant antiviral activity in the range of DS values studied.
  • the decrease in the virus yield is presented as log removal value (LRV), showing the relative decrease in the number of virus DNA copies in the sample treated with the compound, compared to the untreated control sample.
  • LUV log removal value
  • Virus replication assay (Assay 0) was conducted as described above with varying concentrations of the polymer. Then, viral yield in samples was quantified by titration or qPCR. The results are presented in Figure 9. Calculated IC50 values were 37.07 ⁇ g/ml for HSV-1 and 48.56 ⁇ g/ml for HSV-2 as determined by virus titration and 31.71 ⁇ g/ml for HSV- 1 and 17.99 ⁇ g/ml for HSV-2 as determined by qPCR.
  • Fig. 9 viral yield in samples was quantified by titration (A, B) or qPCR (C, D).
  • A, B titration
  • C, D qPCR
  • results are presented as average ⁇ SD. Similar results were obtained in at least three independent experiment.
  • HSV-1 was quantified by titration (A) and by plaque assay (B). Obtained results were normalized to the control sample of polymer-untreated cells. Results are presented as average ⁇ SD. Similar results were obtained in at least three independent experiment.
  • HS is an anionic sulfated polysaccharide (glycosaminoglycan, GAG) which serves as an attachment factor for HSV viruses.
  • GAG glycosaminoglycan
  • 3-O-sulfo groups constitute one the three receptors used by HSV-1 to enter the cell 38 ' 40 .
  • Azure A a cationic dye.
  • Azure A in the solution of HS forms aggregates absorbing at 513 nm, while the monomeric form of the dye (prevalent in the absence of HS) absorbs at 630 nm.
  • Fig. 12 shows the dependence of the concentration of free (unbound) HS on the mass ratio of DEX100DS42 to HS present in solution. The amount of DEX100DS42 required to bind 1 mg of HS was calculated to be equal to 1.30 mg.
  • vir ctr cells not treated with the polymer and infected with HSV-1
  • mock cells not infected with HSV-1
  • vir+DEX100DS42 cells treated with the polymer and infected with HSV-1.
  • Value on y axis represents the number of cells that internalized the virus, relative to the control sample (not treated with the polymer). Results are presented as average ⁇ SD. Similar results were obtained in at least three independent experiment.
  • confocal images were obtained for cells prepared as described for Assay II. After washing off the unbound virus particles, the cells were fixed and stained for visualization of HSV and F-actin.
  • Fig. 14 demonstrates maximal projections of XY stacks. Cells pre-incubated with mock (A) or 500 ⁇ g/ml DEX100DS42 (B) were incubated with HSV-lvirions. Blue - DNA, red - f-actin, green - HSV-1. Maximal projections of collected stacks are presented. Scale bar 10 ⁇ . Similar results were obtained in at least three independent experiment.
  • Cationic derivatives of DEX with different molecular mass (MW) and degree of substitution (DS) were obtained and studied as potential anti-herpes agents. They were found to interact with HS and the efficiency of binding is related to the degree of cationic modification. The analysis of obtained results showed that evaluated polymers can efficiently inhibit HSV-1 entry to the cell.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Biotechnology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Medicinal Preparation (AREA)

Abstract

The use of dextran cationically modified by quaternary ammonium groups of structure I for the treatment and prophylaxis of diseases caused by herpes simplex viruses, wherein dextran is modified using glycidyltrimethylammonium chloride (GTMAC).

Description

The use of cationic derivative of dextran for inhibition of herpes simplex viruses
The subject of the invention is the use of dextran polymer for the prophylaxis and treatment of infections caused by human herpesvirus.
Herpes simplex viruses type 1 and 2 (HSV-1 and HSV-2, respectively) belong to the Herpesviridae family encompassing more than 100 viruses, eight of which affect humans 1. They are also widespread in human population, as it is estimated that 50-90% of the population worldwide may be infected 2. HSV-1 is mainly associated with the watery blisters in the mouth area (lips, tongue, oral mucosa), while HSV-2 mostly affects genital region 3 4. Both of them are, however, also associated with potentially fatal viral encephalitis and stromal keratitis, an ocular disease, which is a leading cause of cornea-derived blindness in developed countries 5"7.
The HSV virion is a spherical, complex structure, about 200 nm in diameter. The inner core, containing the viral DNA stabilized by polyamines, is encapsulated by an icosahedral capsid, which is further coated with an unstructured protein layer, called the tegument. The whole structure is surrounded by a lipid bilayer decorated with viral surface glycoproteins. HSV is a linear dsDNA virus, with genome of 152 and 155 kbp for HSV-1 for HSV-2, respectively. Two major genomic regions - L (long) and S (short) - both flanked with inverted repeats may be distinguished. The HSV genome encodes for at least 90 transcriptional units, which are sorted into 3 classes: a - immediate early, β - early, and γ - late genes. The a genes are located in the inverted repeats of both L and S components and encode for proteins regulating expression of early and late genes. The β and γ genes are distributed within the L and S region
8
Primary HSV infection begins with epithelial cells, usually oral or ocular for HSV-1 and genital for HSV-2. The virus is easily spread through direct contact, i.e., kissing or sexual intercourse. After a productive infection in the mucosal tissue, HSV proceeds to enter nerve endings and, employing retrograde transport, makes its way to the dorsal root neurons nuclei, where it establishes latency - a state of lifelong infection. This possibility, a hallmark of the Herpesviridae family, is facilitated by a failure of immediate early genes transcription caused by ineffective axonal transport of viral regulation factors, such as VP 16 and IFN-a activity in sensory neurons 9 10. Once the virus reaches the latent state, the infection seems to be incurable and may be reactivated, either spontaneously or by stress stimuli, such as fever, UV exposure, or hormonal changes u.
No effective HSV vaccine is available, and therefore current research is oriented towards the development of antiviral compounds limiting the primary infection and supporting further treatment. Currently, the approved anti-HSV therapies involve mainly nucleoside analogues, such as acyclovir (ACV), which interfere with viral DNA synthesis reducing virus replication and shedding. Low bioavailability of this drug led to the development of derivatives with better pharmacokinetic parameters, e.g., selenoaciclovir, valacyclovir, famciclovir, and ganciclovir 12"14. Most of these drugs undergo bioconversion within the cell to ACV. An alternative for treatment of some ACV-resistant HSV infections is foscarnet, which hampers viral polymerase activity by binding to the pyrophosphate binding site 15. All mentioned compounds are oriented on the same molecular target, and therefore some cross resistance is observed 16 1V. For these reasons, there is a need for the development of novel anti -herpes drugs with different mechanism of action. The site of entry of herpesviruses seems to be a promising target, as inhibition of early steps of the virus infection may limit infection, but also virus transmission. In the current study we made an effort to develop the non-toxic cationic polymer that could prevent virions from entering the cell via the formation of multivalent interactions either with the virion (direct inactivation) or with the cell.
Application of polymers for inhibition of HSV infection was proposed before and involved synthetic polycations and polyanions, as well as peptides/proteins. The examples of the polycations which showed anti-HSV activity are poly-L-lysine and poly-L-arginine derivatives were shown to inhibit early stages of HSV replication18, poly(amidoamines) substituted with agmatine19, viologen dendrimers20, and a copolymer of methyl methacrylate, N,N-dimethylaminoethyl methacrylate, and butyl methacrylate (Eudragit®, El 00) which destabilized virus membrane21. The examples of polyanions which inhibit HSV were dendrimers containing sulfonic groups 22, poly(sodium styrene sulfonate) (PSSS)23 and its copolymer with maleic acid24, and telomerized co-acryloyl anionic surfactants25. Among natural polymers having anti-HSV activity were sulfated lignins26, sulfated cellulose nanocrystals (CNC)27, peptides and aptamers, which interfered with virus - cell interactions 28"30, and cationic polysaccharides conjugated with oligoamines via reductive amination 31.
Amine and ammonium derivatives of natural polysaccharides (e.g., dextran, pullulan) also have been studied. It has been shown that dextran substituted with amine groups, in particular dextran-propyl- 1,3 -diamine, effectively inhibits HSV-1 virus replication, while this compound modified by the conversion of a primary amine group to a trimethylammonium group (monoquaternary ammonium propyl-1,3- diamine, MQ-propane- 1,3 -diamine) did not show this activity31, in complete contrast to structurally similar cationic derivatives of dextran being the object of the present application.
According to some studies, entry inhibitors include also sulfonated galactans and fucoidans, dextran sulfate, heparin, and sulfonated lignins.
EP0066379 describes a composition for combating HSV type I and II, comprising a dextran sulfate sodium salt with molecular weight in the range of 4100 to 25000 and containing at least 0.2 equivalent of S03Na per glucopyranosyl unit.
WO2000024784 application discloses dextran derivatives showing biological activity towards HSV, namely dextranpolyaldehyde-resorcine.
The aim of the invention described in the present application is to provide a new effective drug for the treatment and prophylaxis of infections caused by herpes simplex viruses.
Subject Matter of the Invention
The subject of the invention is the use of dextran cationically modified by quaternary ammonium groups of structure I for the treatment and prophylaxis of diseases caused by HSV-1 virus. Preferably, dextran modified by glycidyltrimethylammonium chloride (GTMAC) is used.
Figure imgf000004_0001
Structure I
Preferably, dextran with molecular weight from 6 to 100 kDa is used. Cationically modified dextran has the degree of substitution in the range of 20-100%, preferably 20-60%, more preferably 25-60%.
The use includes polymers based on dextran in the form of ointment or drops applied topically to the skin or to the eye, orally, intraperitoneally, or intravenously in the systemic treatment.
The invention has been illustrated with figures which present preferred embodiments:
Fig. 1 shows IR-ATR spectrum of cationically modified dextrans with the weight of 6 kDa, Fig. 2 is IR-ATR spectrum of cationically modified dextrans with the weight of 40 kDa, Fig. 3 is IR-ATR spectrum of cationically modified dextrans with the weight of 100 kDa, Fig. 4 illustrates the cytotoxicity of polymers. Cell viability is shown as % of the value for the control sample, i.e. not treated with polymers.
Fig. 5 shows the antiviral activity of GTMAC-modified dextrans.
Fig. 6 illustrates the relation between the anti-HSV-1 activity and the degree of substitution. Fig. 7 presents cytotoxicity of the tested polymers.
Fig. 8 shows influence of DS and MW of DEXxDSy on their anti-HSV-1 activity.
Fig. 9 shows concentration-dependent antiviral activity of DEX100DS42 against HSV-1
(A,C) and HSV-2 (B,D).
Fig. 10 shows inhibition of HSV-1 replication cycle at different stages.
Fig. 11 shows UV-Vis absorption spectra of Azure A in the presence of 0.2 mg/ml of HS and different concentrations of DEX100DS42 | g/ml].
Fig. 12 illustrates dependence of the relative concentration of free (unbound) HS on the weight ratio of DEX100DS42 and HS.
Fig. 13 presents HSV-1 attachment in the presence of DEX100DS42, as determined with flow cytometry.
Fig. 14 shows modified dextrans block HSV-1 attachment to susceptible cells.
Fig. 15 shows subcellular localization of dextran derivatives.
Embodiments
The subject of the invention has been presented in more detail in the embodiments.
Example 1
Synthesis of cationic dextrans
0.5 g of dextran with the molecular weight of 6 kDa (DEX6, Sigma-Aldrich), 40 kDa (DEX40, Sigma-Aldrich), or 100 kDa (DEXIOO, Sigma-Aldrich) was dissolved in 25 ml of distilled water, followed by the addition of suitable amount (Table 1) of glycidyltrimethylammonium chloride (GTMAC, Sigma-Aldrich). The solution was alkalized by adding 1 ml 5 M solution of NaOH. The mixture was intensively stirred with magnetic stirrer and heated under reflux for 4 hours at the temperature of 80°C. Upon synthesis, the reaction mixture was cooled to the room temperature, and neutralized with HCl solution. The solution was transferred to a dialysis tube with the molecular weight cutoff value of 3.5 kDa and the dialysis was carried out against water until the conductivity of the said solution decreased below 10 μ8. Obtained polymers were isolated with freeze-drying. These were denoted by acronyms in which the first number is the molecular weight in kilodaltons, while the second is the degree of substitution (DS) in percentage.
Figure imgf000005_0001
Example 2
Elemental analysis-based determination of the degree of substitution in cationic dextrans
Dextran and its cationic derivatives were subjected to the elemental analysis in order to determine the degree of substitution (DS) of glucose units with GTMAC groups, defined as a number of GTMAC groups per 1 glucose unit in dextran. The results are reported in Table 2.
Table 2. Results of elemental analysis of obtained cationic derivatives of dextran
Polymer C f%l H f%l N f%l N/C
DEX6DS0 40.75 6.544 0 0
DEX6DS23 42.49 6.98 1.550 0.0365
DEX6DS37 42.33 6.632 2.24 0.0529
DEX6DS41 41.23 7.214 2.32 0.0563
DEX6DS50 43.55 7.387 2.83 0.0650
DEX6DS52 43.38 7.491 2.87 0.0662
DEX6DS64 41.96 7.379 3.17 0.0754
DEX6DS78 42.62 7.681 3.64 0.0853
DEX40DS0 40.97 6.526 0 0 DEX40DS24 39.60 7.033 1.48 0.0373
DEX40DS26 40.89 7.103 1.64 0.0401
DEX40DS28 39.61 6.866 1.71 0.0432
DEX40DS29 39.56 7.058 1.72 0.0434
DEX40DS37 41.00 7.333 2.16 0.0527
DEX40DS51 41.30 7.587 2.71 0.0656
DEX40DS64 39.58 7.430 2.99 0.0757
DEX100DS0 40.97 6.526 0 0
DEX100DS21 40.30 7.039 1.35 0.0334
DEX100DS33 38.67 7.283 1.86 0.0480
DEX100DS35 41.53 7.372 2.12 0.0510
DEX100DS41 38.37 7.261 2.19 0.0571
DEX100DS42 40.13 7.323 2.32 0.0577
DEX100DS55 38.41 7.298 2.65 0.0690
DEX100DS59 41.91 7.613 3.04 0.0724
DEX100DS71 41.69 7.846 3.37 0.0808
DEX100DS74 40.61 7.726 3.36 0.0827
DEX100DS81 41.27 8.09 3.59 0.0869
DEX100DS82 40.94 7.943 3.60 0.0878
DEX100DS98 40.59 7.932 3.93 0.0967
Example 3
IR spectra for cationic derivatives of dextran
In order to confirm dextran substitutions with GTMAC, IR spectra were measured. The presence of GTMAC groups in modified dextran is demonstrated by a band at about 1475cm"1. The selected results are reported in Table 3 and Fig. 1-3).
Table 3.
Figure imgf000006_0001
Example 4
Zeta potential of cationic derivatives of dextran Zeta potential was measured with NanoZetasizer instrument from Malvern. The measurements were performed for samples at the concentration of 1 mg/ml in 0.015 M NaCl. The results are reported in Table 4.
Table 4. Zeta potential of 1 mg/ml solutions of cationic derivatives of dextran in 0.015 M
NaCl.
Polymer DS Zeta potential [11]
DEX6DS0 0 1.80 ± 1.78
DEX6DS23 23 +9.48 ± 1.80
DEX6DS37 37 +11.5 ±2.29
DEX6DS41 41 +9.69+1.73
DEX6DS50 50 +19.5 + 0.59
DEX6DS59 59 +16.4+1.31
DEX6DS64 64 +18.6+1.99
DEX6DS78 78 +8.77 + 0.90
DEX40DS0 0 -1.87 + 0.64
DEX40DS24 24 +7.06+1.23
DEX40DS26 26 +14.0+1.61
DEX40DS28 28 +17.1 ± 1.48
DEX40DS29 29 +9.06 + 2.93
DEX40DS37 37 +15.4 + 0.95
DEX40DS51 51 +17.9+1.51
DEX40DS64 64 +33.2+1.65
DEX40DS66 66 +22.4 + 2.15
DEX100DS0 0 -2.85 ± 0.94
DEX100DS21 21 +9.78 + 2.58
DEX100DS33 33 +22.8 + 3.27
DEX100DS35 35 +25.2+1.42
DEX100DS41 41 +21.5 + 2.21
DEX100DS42 42 +15.7+1.17 DEX100DS55 55 +22.9 ± 1.31
DEX100DS59 59 +22.1 ±0.56
DEX100DS71 71 +29.5 + 0.55
DEX100DS74 74 +25.5 + 0.93
DEX100DS81 81 +23.4 + 0.20
DEX100DS82 82 +35.3 + 2.00
DEX100DS98 98 +24.2 + 4.31
Example 5
Effect of cationically modified dextrans on the cell viability
The cytotoxicity of cationic derivatives of dextran was determined using XTT assay. The assay was performed on Vero E6 cell line which is permissive for HSV-1 virus. This assay is based on the determination of mitochondrial enzymes' activity by testing the ability thereof to reduce a substrate to a colored product. The cell viability was determined with measuring the absorbance at 450 nm in relation to the control which was represented by cells cultured in medium without additives. Cells were cultured for 2 days in a mixture of MEM medium, with the addition of Hank's salt, and M199 medium, with the addition of Eagle's medium in 2: 1 ratio, supplemented with 3% heat-inactivated fetal bovine serum (FBS), as well as penicillin and streptomycin, with the addition of the tested polymer.
Results presented in Fig. 4 show the cytotoxicity of the synthesized polymers at various concentrations.
Example 6
Effect of cationically modified dextrans on the replication of human herpesvirus type I (HS V- 1)
Upon the determination of compound concentrations without toxic effect on cells, their effect on HSV-1 virus replication was tested. Cells were pre-incubated with dextrans at suitable concentrations for 30 min and, subsequently, still in the presence of dextrans, were infected with the virus at TCID5o=400. After 2 hours, unbound virus particles were removed by washing 3 times with PBS, and the incubation was performed, also in the presence of dextrans, for the next 2 days. Subsequently, DNA was isolated from cell medium and used as a matrix in real-time PCR. Dextrans' ability to inhibit HSV-1 replication was determined as a decrease in virus copy number per millilitre of medium.
Fig. 5 shows the antiviral activity of GTMAC-modified dextrans. For the analysis, GTMAC- modified dextrans with various molar weight (6 , 40, and 100 kDa) were used. Suitable unmodified dextrans were used as controls (ctrl). For all compounds the concentration used was 500 μg/ml.
Based on the diagram, it can be clearly seen that GTMAC-modified dextran inhibits HSV-1 multiplication when compared with the suitable control sample. Next, the effect of the degree of GTMAC substitution on the antiviral activity at the constant molar weight of dextran (6 kDa) was determined.
Fig. 6 shows the relation between the anti-HSV-1 activity and the degree of substitution. Three different degrees of polymer substitution were used: 23%, 37%, and 52%. The presented diagram clearly shows that the antiviral activity of the polymer correlates with the degree of substitution.
Example 7
Cationic derivatives of dextran (DEXxDSy) as effective inhibitors of HSV-1 and HSV-2 infection.
Dextran (DEX, MW of 6, 40 and 100 kDa, from Leuconostoc spp., Sigma), glycidyltrimethylammonium chloride (GTMAC, >90% Sigma), heparan sulfate (HS), Azure A chloride (Fluka, Fluka standard), PBS (tablet, Sigma), fluorescein 5(6)-isotiocyanate (FITC, >90% (HPLC), Sigma), sodium chloride (analytical grade, POCh), NaOH (analytical grade, POCh), HC1 (pure, POCh) were used as received. Water was distilled twice and deionized using the Millipore Simplicity system. FTIR spectra were obtained on a Bruker IFS 48 spectrometer. Elemental analysis (EA) was performed on a EuroEA 3000 Elemental Analyzer. The UV/VIS absorption spectra were obtained at room temperature in 1-cm quartz cuvettes using the single beam diode array Hewlett-Packard 8452A spectrophotometer with a resolution of 2 nm in the range of 190-820 nm. Zeta potential measurements were performed using Zetasizer Nano ZS instrument (Malvern Instruments). The sample was illuminated with a 633-nm laser and the intensity of light scattered at an angle of 173° was measured with an avalanche photodiode.
Synthesis of cationically modified dextran (DEX)
The synthesis of cationic derivatives of dextran is presented in Scheme 1. Cationic derivatives of DEX were obtained by the substitution of hydroxyl groups with GTMAC as described previously 32 with some modifications. We have applied two synthetic protocols. In the first synthetic procedure, 0.5 g of DEX was dissolved in 25 ml of distilled water, then NaOH (2.5 mmol) was added. The solution was stirred with a magnetic stirrer and heated to 60°C or 80°C. In the next step different volumes of GTMAC were added and the mixtures were kept at 60°C or 80°C for 4 h while being stirred. The MW values of DEX, volume of GTMAC and the reaction temperature used to obtain each DEX derivative are given in Table SI (see Supporting Information). The products are denoted as DEXxDSy where x is the MW of DEX in kDa and y is the degree of substitution defined as the number of GTMAC groups attached to DEX per 100 glucose repeating units. In the second synthetic procedure both the reaction time and GTMAC volume were varied (Table S2). The first polymer sample in each series was obtained by reacting 2 g of DEX dissolved in 100 ml of distilled water with initial volume of GTMAC (20 or 40 ml) for 1 or 2 h. Then, a defined volume of the reaction mixture was withdrawn to isolate a polymer sample while to the rest of the reaction mixture the defined volume of GTMAC was added and the resulting mixture was heated while mixing for the defined time period. The withdrawal/GTMAC addition/heating cycles were repeated twice or thrice. To isolate the polymer the reaction mixtures were neutralized to pH~7 by the addition of 1 M HC1, cooled and transferred to dialysis tubes (MW cutoff values of 3.5, 12.8 and 14 kDa, depending of the MW of DEX). The dialysis was carried out against distilled water until the conductivity of the liquid surrounding the tube decreased below 10 μ8. The polymers obtained were isolated from the solution using the freeze-drying technique. The substitution of DEX with GTMAC was confirmed using EA, IR spectroscopy and zeta potential measurements. The DS values of the polymers obtained covered almost the whole possible range, i.e. from 2 to 98.
Synthesis of fluorescently labeled DEXxDSy
DEX6DS41, DEX40DS37 and DEX100DS40 were fluorescently labeled as described previously33. Briefly, 50 mg of the respective polymer was dissolved in 2 ml of warm DMSO and 1 drop of pyridine and 3 mg of FITC in 2 ml of DMSO were added. The mixture was heated to 95°C for 2 hours under continuous stirring. Then, DMSO was removed by dialysis against water and the product was lyophilized.
Determination of the degree of substitution (DS) of DEX derivatives with GTMAC
The DS of the DEXxDSy was determined with elemental analysis (EA). The ratio of N/C in each polymer was determined and the DS was calculated using the following equation:
DS [%] = 100 (457.34 (N/C)3 + 3.007 (N/C)2 + 5.5951 (N/C)) where N/C is the ratio of the mass fraction of nitrogen to carbon
Studies on the interactions between the DEXxDSy and HS using a colorimetric method The mixtures containing a constant concentration of HS (0.2 mg/ml) and 1 x PBS solutions of various DEXxDSy at different concentrations (0.06-0.27 mg/ml) were prepared. The mixtures were shaken for 10 min and centrifuged at 3000 rpm for 10 min to separate the insoluble complex of HS with DEXxDSy. Then, 0.1 ml of the supernatant was added to the mixture of 0.9 ml of 1 x PBS and 1 ml of 8 10"5 M Azure A solution and the UV-VIS spectra were measured.
Cell culture
Vero E6 cells (Cercopithecus aethiops kidney epithelial, ATCC: CRL-1586) were routinely maintained under Dulbecco-modified Eagle's medium (DMEM, high glucose, Life Technologies) supplemented with 3% heat-inactivated fetal bovine serum (FBS, Life Technologies), penicillin (100 U/ml), and streptomycin (100 μg/ml) at 37°C in atmosphere containing 5% C02.
Virus preparation and titration
HSV-1 strain 17+ and HSV-2 strain HG52 were obtained from Public Health England (0104151v and 0104152v, respectively). Virus stocks were generated by infecting Vero E6 cells monolayers for 48 h, then lysing the cells with two freeze-thaw cycles. Collected lysates were aliquoted and stored at -80°C. Mock samples were prepared in the same manner, using mock-infected cells. Virus stocks were quantified by titration on Vero E6 cells (48 h infection, 37°C), according to Reed and Muench method 34. Following a 48 h incubation the number of cytopathic effect (CPE)-positive wells was counted and the TCID50 was calculated.
Cell viability assay
Cell viability was assessed on Vero E6 cell line using XTT Cell Viability Assay Kit (Biological Industries) according to vendor's protocol. Briefly, cells were seeded in 96-well plates and cultured for 48 h. Subsequently medium was discarded and cells were overlaid with fresh medium supplemented with DEXxDSy polymers or control samples. Culture was carried out for 48 h at 37°C and after that incubation medium was refreshed again and 25 μΐ of activated XTT reagent was added to each well. Following a 2 h incubation at 37°C, media were transferred to a new 96-well plate and absorbance was measured at λ=480 nm using a spectrophotometer (Flexstation 3, Molecular Devices). The results were normalized against a control of untreated cells (100% viability).
Virus replication assay (Assay 0)
Vero E6 cells were seeded in 6-well plates and cultured for 2 days at 37°C in atmosphere supplemented with 5% of CO2. Subsequently, media was discarded and cells were overlaid with fresh media supplemented with DEXxDSy polymers or control samples. Following a 30 min incubation, cells were infected with HSV-1 or HSV-2 virus at TCID50 = 400/ml, or mock-infected. Infection was carried out for 2 h at 37°C. Subsequently, unbound virus particles were removed by triple washing with sterile 1 χ PBS, and culture media supplemented with polymers were applied. Samples were analyzed 2 days post-inoculation
(P i ).
Plaque assay
24 h prior to the infection cells were seeded in 6-well plates. On day 0 sub-confluent cells were infected with 10-fold serial dilutions of the virus and incubated for 1 h at 37°C. Subsequently, cells were washed once with 1 χ PBS and overlaid with 2 ml of media supplemented with 10% heat-inactivated fetal bovine serum (FBS) and 0.33% methylcellulose. After 72 h, media were discarded and 1 ml of 0.1% crystal violet solution in 50:50 water : ethanol mixture was added to each well. Following a 10 min incubation at room temperature, cells were washed twice with water and plaques were counted.
Mechanism of anti-HSV action of DEXxDSy
A series of assays was employed to determine the mechanism of antiviral activity of DEXxDSy. The tests differ mainly in the sequence in which the cells, the virus and the polymer may interact.
Assay I: Does the compound inactivate the virus?
Virus stock samples were incubated with DEXxDSy polymers (500 μg/ml) for 1 h at 22°C under constant mixing. Subsequently, the samples were diluted to decrease the polymer concentration below its active range (25 μg/ml). In negative control samples cell culture medium was added instead of the polymer solution. The samples were then titrated on fully confluent Vero E6 cells to assess viral yield. This test indicates whether the antiviral effect results from a direct interaction between the polymer and the virus.
Assay II: Does the compound protect the cell?
Fully confluent Vero E6 cells were overlaid with polymer solution and incubated for 1 h at 37°C. Subsequently, the polymer was removed by triple wash with 1 x PBS and virus at TCID50 = 400/ml or mock at equal volume was added. After 2 h infection at 37°C, the virus was washed away, fresh culture medium was applied, and the cells were cultured for 48 h. Samples of cell culture supernatant were collected and viral yield was assessed. As no unbound polymer is present during the exposure to virus, inhibition of infection in this test indicates the influence of the polymer on the cell (e.g., cell surface coating, induction of innate immune responses).
Assay III: Does the compound block virus - receptor interaction?
Fully confluent Vero E6 cells were chilled on ice, overlaid with an ice-cold solution of the DEXxDSy polymer (500 μg/ml) containing the virus at TCID50 = 400/ml or mock at equal volume in cell culture medium. Under these conditions, virions can attach to their receptor on the cell surface, but cannot enter the cell, as the intracellular transport is inhibited at lower temperatures. Following 1 h incubation at 4°C, the polymer solution was removed, cells were washed thrice with ice-cold 1 x PBS to remove any unbound polymers and virions, overlaid with fresh medium and cultured for 48 h at 37°C. Then, samples of cell culture supernatant were collected and viral yield was assessed. If the polymer inhibits the virus-receptor interaction, a decline in the viral yield is observed compared to the control.
Assay IV: Does the compound hamper infection at later stages?
Fully confluent Vero E6 cells were infected with virus at TCID50 = 400/ml or mock for 2 h at 4°C. Subsequently, cells were rinsed thrice with 1 x PBS to remove unbound virus particles and fresh medium supplemented with the polymer was added. Cells were cultured for 48 h at 37°C, then medium samples were collected and viral yield was assessed. Since the polymer is absent during the early stages of infection (virus adsorption/attachment and entry), a decline in the viral yield compared to the control in this test indicates that the antiviral activity of the polymer is due to inhibition of later stages of virus replication cycle (e.g., virus replication, assembly or egress). qPCR
Viral DNA was isolated from supernatants using Viral DNA/RNA Isolation Kit (A&A Biotechnology, Poland). Virus yield was then determined by quantitative real-time PCR (qPCR). The reaction was carried out in a CFX96 Touch™ Real-Time PCR Detection System (Bio-Rad), in 10 μΐ reaction mixture consisting of 1 x Kapa Probe Fast qPCR Master Mix, specific probe labeled with 6-carboxyfluorescein (FAM) and Black Hole Quencher 1 (BHQ1) (sequence: 5' FAM CCG CCG AAC TGA GCA GAC ACC CGC GC BHQ1 -3 ', 100 nM), and primers (450 nM each, sequences: sense primer 5 '-CAT CAC CGA CCC GGA GAG GGA C-3', anti-sense primer 5' GGG CCA GGC GCT TGT TGG TGT A-3') and 2.5 μΐ of viral DNA in 10 μΐ. The temperature profile included 3 min at 95°C, followed by 37 cycles of 2 s at 95°C and 20 s at 60°C. For quantification DNA standards were prepared. Briefly, fragment of DNA polymerase gene, conserved among HSV-1 and HSV-2 strains, was amplified using the primers mentioned above and cloned into pTZ57R/T (Thermo Scientific, Poland) plasmid using InsTAclone PCR cloning kit (Thermo Scientific, Poland). The plasmid was propagated in E. coli TOP 10 (Life Technologies, Poland), purified with GeneJET Plasmid Miniprep Kit (Thermo Scientific, Poland) and linearized by digestion with Kpnl restriction enzyme. Concentration of the linearized DNA was determined spectrophotometrically and the number of copies per milliliter was calculated. Six subsequent serial dilutions were then used as qPCR reaction template.
Flow cytometry
Vero E6 cells were seeded in 6-well plates and cultured for 2 days at 37°C in atmosphere containing 5% CO2. For analysis, cells were trypsinized and fixed with 4% PFA in 1 x PBS for 20 min. Subsequently, the cells were permeabilized by 20 min incubation in 0.1% Triton X100 in 1 x PBS and non-specific binding was blocked by overnight incubation with 5% BSA in 1 x PBS at 4°C. Protein labelling was carried out by 2 h incubation with primary rabbit anti-HSV VP5 antibody (20-HR50, Fitzgerald Industries, USA) diluted 1 :500, followed by lh incubation with secondary goat anti-rabbit antibody, conjugated with AlexaFluor 488 (A11001, Invitrogen, Poland). After washing with 0.1% Tween-20 in 1 x PBS, the cells were re-suspended in 1 x PBS and analyzed with FACS Calibur (Becton Dickinson) using Cell Quest software. Confocal microscopy
Vero E6 cells were seeded on coverslips in 6-well plates and cultured for 2 days at 37°C with 5% C02. For analysis of virus adhesion and entry, Assays II and III were carried out, respectively. Immediately after washing away unbound virions, cells were fixed with 4% PFA in 1 x PBS for 20 min. Subsequently, the cells were permeabilized by 20 min incubation in 0.1% Triton X100 in 1 x PBS and non-specific binding sites were blocked by overnight incubation in 5% BSA in 1 x PBS at 4°C. To visualize virus particles, cells were incubated for 2 h with primary rabbit anti-HSV VP5 antibody (20-HR50, Fitzgerald Industries, USA) diluted 1 :500 in 1 x PBS with 0.5% Tween 20, followed by 1 h incubation with secondary goat anti -rabbit antibody, conjugated with AlexaFluor 488 (A11001, Invitrogen, Poland). F-actin was co-stained with AlexaFluor 633 -conjugated phalloidin (Invitrogen, Poland). Nuclear DNA was labelled with 0.1 μg/ml 4',6'-diamidino-2-phenylindole (DAPI) (Sigma- Aldrich, Poland). Immunostained cultures were mounted on glass slides with ProLong Diamond Antifade Mountant (Life technologies, Poland). Fluorescent images were acquired under a ZEISS LSM 710 (release version 8.1) confocal microscope and acquired with ZEN 2012 SP1 (black edition, version 8.1.0.484) software. Stacks acquisition parameters were as follows: frame size 1024 x 1024, step size 0.35 μπι, pixel size 0.10 μπι. For image processing ImageJ FIJI version was used.
Statistical analysis
All the experiments were performed in triplicates and the results are expressed as mean ±SD. To determine the significance of the obtained results a comparison between groups was made using the Student's t test. P values <0.05 were considered significant.
Synthesis of DEXxDSy derivatives
In order to systematically study the subject, an extended library of polymers was synthesized, varying in the chain's length and the content/density of positively charged GTMAC residues. Cationic DEX derivatives (DEXxDSy) were synthesized by the GTMAC epoxide ring opening in basic conditions followed by the substitution of the hydrogen atom of the hydroxyl groups with GTMAC. The synthesis of cationic derivatives of DEX is presented in Scheme 1. The structure of the DEXxDSy polymers was confirmed using EAand FTIR spectroscopy. EA revealed the presence of nitrogen in the obtained polymers, which unambiguously confirms the successful substitution of DEX with GTMAC. DS values were calculated based on the elemental composition of the polymers.
Figure imgf000013_0001
OH CH3
I
N+-CH3
CH3
Scheme 1. Synthesis of the cationic derivatives of DEX.
In the FTIR spectra of all GTMAC-substituted polymers a weak band at 1477 cm"1 appeared that may be attributed to the presence of asymmetric angular binding of the methyl groups in GTMAC, thereby confirming substitution of DEX with GTMAC. The zeta potential was determined for the synthesized polymers (1 mg/ml solution in 0.015 M NaCl at 25°C). All measurements were carried out in triplicate. As expected, the DEX derivatives have a positive zeta potential resulting from the presence of the cationic quaternary amino groups of GTMAC attached to the main chain. The values of zeta potential ranged from +7 to 35 mV and they generally increased with increasing DS of the polymers studied.
Cytotoxicity
In order to evaluate the usability of developed compounds, cytotoxicity of DEXxDSy was determined using XTT assay, which relies on the ability of mitochondrial enzymes to convert the substrate (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) into colored formazan salts. The assay was performed on Vero E6 cell line which is an accepted model for HSV-1 and HSV-2 infection. Briefly, cells were incubated for 48 hours in the presence of different DEXxDSy polymers. Results presented in Fig. 7 clearly show that the polymers of lower MW are characterized by lower toxicity. Increase in the polymer toxicity with increasing MW may be explained by the fact that longer polymer chain adopts the globular conformation with high surface density resulting from large number of charged residues present. It is also very clear that the increase in charge density increases polymer antiviral activity. Thus, there is a need to optimize the system to ensure the highest antiviral activity while maintaining low toxicity. It was found that DEX100DS42 had optimal activity/toxicity properties and therefore it was chosen for mechanistic studies.
In Fig. 7 cell viability was assessed using XTT assay 2 days p.i. Background fluorescence was subtracted for all the samples. Cell viability was calculated in reference to untreated cells (control). Derivatives of dextran with MW of 6 kDa (A), 40 kDa (B) and 100 kDa (C) with various DS were tested in the range of concentrations 50 - 2000 μg/ml. Results are presented as average ± SD. Similar results were obtained in at least three independent experiment.
Antiviral activity of DEXxDSy
The entry of virus into host cells is the first and critical step in the viral pathogenesis 35'36. It is mediated by an interplay of viral and cellular proteins, which differ in the number and characteristics between viral species. HSV-1 encodes several glycoproteins responsible for cell surface binding and virus entry. Glycoproteins gB and gC are responsible for initial binding of the virus at cell surface, involving HS proteoglycans (HSPGs). Virions devoid of these glycoproteins demonstrate significantly impaired infectivity 35. gB was also found to bind PILR-a, a particle considered to be a HSV-1 coreceptor 31. Glycoprotein D (gD) is considered to be the main entry mediator and several receptors for gD have been identified. The most important are nectin-1 and nectin-2, herpesvirus entry mediator (HVEM) and 3-0 sulfated HSPG 38-41. The in vivo experiments carried out using single and double knockout mice have demonstrated the dependence of HSV-2 on nectin-1 for entry and spread 42 and HSV-1 on HSPG receptor for their entry to the cells and spread. Interestingly, the in vitro experiments carried out using human corneal fibroblasts have shown that, unlike HSV-1, HSV- 2 does not use the HSPG receptor for cell penetration 43. Another two glycoproteins taking part in HSV-1 entry, gH and gL form a heterodimer, which dissociates upon binding to its receptor, ανβό- or avP8-integrin. This interaction induces a change in gH conformation which induces virus entry into the cell and directs the virus to acidic endosomes 44.
In order to assess inhibitory activity of the tested polymers, virus replication assay was performed. Fig. 8 demonstrates the decreased virus replication in the presence of both cationically modified 40 kDa DEX (DEX40DSy) and in the presence of cationically modified 100 kDa DEX (DEXlOODSy), for polymers with DS > -20-40%. The anti-HSV-1 activity of both polymers was similar in this assay. On the other hand, cationically modified 6 kDa DEX showed no significant antiviral activity in the range of DS values studied. The decrease in the virus yield is presented as log removal value (LRV), showing the relative decrease in the number of virus DNA copies in the sample treated with the compound, compared to the untreated control sample. In Fig. 8 cationic dextrans with MW of 6, 40 and 100 kDa with different DS were present throughout the infection. Two days p.i. the amount of viral DNA in cell culture supernatant was assessed by qPCR. Difference in viral yield is presented on y-axis as log removal value (LRV), showing the relative decrease in the amount of virus in cell culture media, compared to the control. Results are presented as average ± SD. Similar results were obtained in at least three independent experiment.
Virus replication assay (Assay 0) was conducted as described above with varying concentrations of the polymer. Then, viral yield in samples was quantified by titration or qPCR. The results are presented in Figure 9. Calculated IC50 values were 37.07 μg/ml for HSV-1 and 48.56 μg/ml for HSV-2 as determined by virus titration and 31.71 μg/ml for HSV- 1 and 17.99 μg/ml for HSV-2 as determined by qPCR.
In Fig. 9 viral yield in samples was quantified by titration (A, B) or qPCR (C, D). For virus titration the virus quantity is presented as TCID50, and for qPCR as number of viral DNA copies per milliliter. Results are presented as average ± SD. Similar results were obtained in at least three independent experiment.
Mechanism of action
An effort was made to elucidate the antiviral mechanism of action of modified dextrans. For this, described above set of functional assays was used. For mechanistic studies DEX100DS42 polymer was used.
First, the influence was investigated of DEX100DS42 on HSV-1 virions (Assay I). No significant reduction of viral titer was detected. Next, the possibility that the polymer renders the cell resistant to the infection was tested (Assay II). Decrease in virus yield (-50%) was noted using both techniques. Subsequently, the ability of DEX100DS42 to block viral adhesion to the cell was examined (Assay III). A decrease in viral titer reaching -90% for virus titration and 100%> for plaque assay was observed. Finally, it was tested whether DEX100DS42 may interfere with virus replication and release (Assay IV). Decrease in virus titer (-50%)) was observed. The results of the tests carried out using the functional assays are presented in Fig. 10, where following functional assays described in the methods section HSV-1 was quantified by titration (A) and by plaque assay (B). Obtained results were normalized to the control sample of polymer-untreated cells. Results are presented as average ± SD. Similar results were obtained in at least three independent experiment.
Interaction between the cationic DEX and HSV attachment receptor
The interaction between DEXxDSy polymers and HS was studied. HS is an anionic sulfated polysaccharide (glycosaminoglycan, GAG) which serves as an attachment factor for HSV viruses. Further, a variant of HS in which 3-O-sulfo groups are present constitute one the three receptors used by HSV-1 to enter the cell 38'40. The ability of the studied polymers to bind HS was assessed with a colorimetric method using Azure A, a cationic dye. Azure A in the solution of HS forms aggregates absorbing at 513 nm, while the monomeric form of the dye (prevalent in the absence of HS) absorbs at 630 nm. Addition of the DEXxDSy polymer caused the disruption of dye aggregates resulting in the increase in intensity of the absorption at 630 nm (monomelic band) and in the decrease of absorption at 513 nm (aggregate absorption band). The plot showing changes in the absorption spectra of Azure A upon addition of HS is shown in Fig. 11.
Based on the results of these experiments one can conclude that the cationic DEX derivatives form complexes with HS in 1 x PBS buffer (pH=7.4) what is consistent with our previous observation that cationic dextran derivatives reverse the anticoagulant activity of heparin in vitro and in vivo due to its complexation13. It was also possible to quantify the complexation process. Fig. 12 shows the dependence of the concentration of free (unbound) HS on the mass ratio of DEX100DS42 to HS present in solution. The amount of DEX100DS42 required to bind 1 mg of HS was calculated to be equal to 1.30 mg.
Obtained data demonstrated clearly that HS is complexed with all DEXxDSy polymers obtained. The general trend was that cationic DEXxDSy of a given MW bound HS stronger when their DS was higher. Among DEXxDSy of comparable DS the derivatives of DEX with MW of 40 kDa showed the strongest HS binding properties.
To confirm whether the interactions between cationic dextrans and HS present on the cell surface play a role in the inhibition of virus entry, flow cytometry analysis was carried out, as described in Methods section. Data was normalized against a control of cells infected without pre-incubation with dextran. The signal recorded from the infected sample treated with DEX100DS42 was vastly limited, reaching the intensity observed for mock-treated sample. The results are presented in Fig. 13 and suggest that interaction between modified dextrans and HS present at the cell surface hinders the viral adhesion to the host's cell. In Fig. 13 cells were pre-incubated with the polymer for 1 h, then infected with HSV-1 or mock sample for 2 h in the absence of DEX100DS42. vir ctr: cells not treated with the polymer and infected with HSV-1, mock: cells not infected with HSV-1, vir+DEX100DS42: cells treated with the polymer and infected with HSV-1. Value on y axis represents the number of cells that internalized the virus, relative to the control sample (not treated with the polymer). Results are presented as average ± SD. Similar results were obtained in at least three independent experiment.
To additionally verify the ability of cationic dextrans to inhibit HSV attachment to cell surface, confocal images were obtained for cells prepared as described for Assay II. After washing off the unbound virus particles, the cells were fixed and stained for visualization of HSV and F-actin. Fig. 14 demonstrates maximal projections of XY stacks. Cells pre-incubated with mock (A) or 500 μg/ml DEX100DS42 (B) were incubated with HSV-lvirions. Blue - DNA, red - f-actin, green - HSV-1. Maximal projections of collected stacks are presented. Scale bar 10 μπι. Similar results were obtained in at least three independent experiment.
Even though our results clearly show that developed polymers should interfere with early steps of virus replication, inhibition of virus replication was also observed. One may assume that observed inhibition of virus replication at the later stages of infection may result from multi-cycle replication and blockade of subsequent transmission events, however, an additional mechanism involving interruption of virus assembly, packing or releasing cannot be ruled out. Mechanistic studies indicated that the GTMAC-substituted dextrans hamper virus infection by interference with virus binding to the cell surface. Such decrease in virus density on susceptible cells will most likely limit virus entry and virus infectivity, as observed in vitro. One may also assume polymers may hamper entry of HSV-1 via HS receptor during ocular infections. We believe that the developed polymers are able to interfere with HSV-1 (and possibly also HSV-2) transmission and limit virus spread between cells and as such may be used for prophylaxis or in combination with replication inhibitors.
Subcellular localization of dextran derivatives
To verify, whether polymers may enter the cell fluorescent derivatives of DEXxDSy were used Briefly, cells were incubated with 250 μg/ml of fluorescently labeled DEXxDSy for 1 h at 37°C, then cells were rinsed with 1 χ PBS, fixed and stained with phalloidin and DAPI as described above. Even though all tested polymers (DEX6DS41, DEX40DS37, and DEX100DS40) were present inside the cell, the intracellular presence of DEX100DS40 seemed to be limited, compared to the polymers of lower MW. However, detailed study should be carried out to quantify the polymer internalization. The results are presented in Fig.15. Fluorescent derivatives of DEX6DS4i (A), DEX40DS3v (B) and DEX100DS40 (C). Cells were incubated with the compounds for 1 h at 37°C. Blue - DNA, red - f-actin, green - dextran. Images present single slices of XY stacks. Scale bar 10 μιη. Similar results were obtained in at least three independent experiment.
Cationic derivatives of DEX with different molecular mass (MW) and degree of substitution (DS) were obtained and studied as potential anti-herpes agents. They were found to interact with HS and the efficiency of binding is related to the degree of cationic modification. The analysis of obtained results showed that evaluated polymers can efficiently inhibit HSV-1 entry to the cell.
References:
(1) Fatahzadeh, M; Schwartz, R. A. Human Herpes Simplex Virus Infections: Epidemiology, Pathogenesis, Symptomatology, Diagnosis, and Management. J. Am. Acad. Dermatol. 2007, 57 (5), 737-763.
(2) Strick, L. B.; Wald, A.; Celum, C. Management of Herpes Simplex Virus Type 2 Infection in HIV Type 1 -Infected Persons. Clin. Infect. Dis. 2006, 43 (3).
(3) Szczubialka, K.; Pyre, K.; Nowakowska, M. In Search for Effective and Definitive Treatment of Herpes Simplex Virus Type 1 (HSV-1) Infections. RSCAdv. 2016, 6 (2), 1058-1075.
(4) Smith, J. S.; Robinson, N. J. Age-Specific Prevalence of Infection with Herpes Simplex Virus Types 2 and 1 : A Global Review. J Infect Dis 2002, 186 Suppl, S3-28.
(5) Kropp, R. Y.; Wong, T.; Cormier, L.; Ringrose, A.; Burton, S.; Embree, J. E.; Steben, M. Neonatal Herpes Simplex Virus Infections in Canada: Results of a 3 -Year National Prospective Study. Pediatrics 2006, 117 (6), 1955-1962.
(6) Farooq, A. V; Shukla, D. Herpes Simplex Epithelial and Stromal Keratitis: An Epidemiologic Update.
Surv Ophthalmol 2012, 57 (5), 448-462.
(7) Moerdyk-Schauwecker, M.; Stein, D. A.; Eide, K.; Blouch, R. E.; Bildfell, R.; Iversen, P.; Jin, L.
Inhibition of HSV-1 Ocular Infection with Morpholino Oligomers Targeting ICP0 and ICP27. Antiviral Res. 2009, 84 (2), 131-141.
(8) Knipe, D. M.; Howley, P. M. Fields Virology, 6th ed.; Wolters Kluwer/Lippincott Williams & Wilkins Health: Philadelphia, PA, 2013.
(9) Wilson, A. C; Mohr, I. A Cultured Affair: HSV Latency and Reactivation in Neurons. Trends Microbiol. 2012, 20 (12), 604-611.
(10) De Regge, N.; Van Opdenbosch, N.; Nauwynck, H. J.; Efstathiou, S.; Favoreel, H. W. Interferon Alpha Induces Establishment of Alphaherpesvirus Latency in Sensory Neurons in Vitro. PLoS One 2010, 5 (9).
(11) Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis; Arvin, A., Campadelli-Fiume, G., Mocarski, E., Moore, P. S., Roizman, B., Whitley, R., Yamanishi, K., Eds.; Cambridge, 2007.
(12) Cirelli, R.; Heme, K.; McCrary, M.; Lee, P.; Tyring, S. K. Famciclovir: Review of Clinical Efficacy and Safety. Antiviral Res. 1996, 29 (2-3), 141-151.
(13) Sacks, S. L.; Griffiths, P. D.; Corey, L.; Cohen, C; Cunningham, A.; Dusheiko, G. M.; Self, S.;
Spruance, S.; Stanberry, L. R.; Wald, A.; Whitley, R. J. HSV Shedding. Antiviral Res. 2004, 63 (SUPPL. 1), S19-S26.
(14) Sahu, P. K.; Umme, T.; Yu, I; Nayak, A.; Kim, G.; Noh, M. ; Lee, J.-Y.; Kim, D.-D.; Jeong, L. S.
Selenoacyclovir and Selenoganciclovir: Discovery of a New Template for Antiviral Agents. J. Med. Chem. 2015, 58 (21), 8734-8738.
(15) Sauerbrei, A. Optimal Management of Genital Herpes: Current Perspectives. Infect Drug Resist 2016, 9, 129-141.
(16) Morfin, F.; Thouvenot, D. Herpes Simplex Virus Resistance to Antiviral Drugs. J. Clin. Virol. 2003, 26 (1), 29-37.
(17) Blot, N.; Schneider, P.; Young, P.; Janvresse, C; Dehesdin, D.; Tron, P.; Vannier, J. P. Treatment of an Acyclovir and Foscarnet-Resistant Herpes Simplex Virus Infection with Cidofovir in a Child after an Unrelated Bone Marrow Transplant. Bone Marrow Transplant. 2000, 26 (8), 903-905.
(18) Langeland, N.; Moore, L. J.; Holmsen, H.; Haarr, L. Interaction of Polylysine with the Cellular Receptor for Herpes Simplex Virus Type 1. J Gen Virol 1988, 69 (Pt 6), 1137-1145.
(19) Donalisio, M.; Ranucci, E.; Cagno, V.; Civra, A.; Manfredi, A.; Cavalli, R.; Ferruti, P.; Lembo, D.
Agmatine-Containing Poly(amidoamine)s as a Novel Class of Antiviral Macromolecules: Structural Properties and in Vitro Evaluation of Infectivity Inhibition. Antimicrob. Agents Chemother. 2014, 58 (10), 6315-6319.
(20) Asaftei, S.; De Clercq, E. "Viologen"dendrimers as Antiviral Agents: The Effect of Charge Number and Distance. J. Med. Chem. 2010, 53 (9), 3480-3488.
(21) Alasino, R. V; Ausar, S. F.; Bianco, I. D.; Castagna, L. F.; Contigiani, M.; Beltramo, D. M. Amphipathic and Membrane-Destabilizing Properties of the Cationic Acrylate Polymer Eudragit® E100. Macromol. Biosci. 2005, 5 (3), 207-213.
(22) Gong, Y.; Matthews, B.; Cheung, D.; Tarn, T.; Gadawski, I.; Leung, D.; Holan, G.; Raff, J.; Sacks, S.
Evidence of Dual Sites of Action of Dendrimers: SPL-2999 Inhibits Both Virus Entry and Late Stages of Herpes Simplex Virus Replication. Antiviral Res. 2002, 55 (2), 319-329.
(23) Zeitlin, L.; Whaley, K. J.; Hegarty, T. A.; Moench, T. R.; Cone, R. A. Tests of Vaginal Microbicides in the Mouse Genital Herpes Model. Contraception 1997, 56 (5), 329-335.
(24) Qiu, M.; Chen, Y.; Song, S.; Song, H.; Chu, Y.; Yuan, Z.; Cheng, L.; Zheng, D.; Chen, Z.; Wu, Z. Poly (4-Styrenesulfonic Acid-Co-Maleic Acid) Is an Entry Inhibitor against Both HIV-1 and HSV Infections - Potential as a Dual Functional Microbicide. Antiviral Res. 2012, 96 (2), 138-147.
(25) Leydet, A.; Barragan, V.; Boyer, B.; Montero, J. L.; Roque, J. P.; Witvrouw, M.; Este, J.; Snoeck, R.;
Andrei, G.; De Clercq, E. Polyanion Inhibitors of Human Immunodeficiency Virus and Other Viruses. 5. Telomerized Anionic Surfactants Derived from Amino Acids. J. Med. Chem. 1997, 40 (3), 342-349.
(26) Raghuraman, A.; Tiwari, V.; Zhao, Q.; Shukla, D.; Debnath, A. K.; Desai, U. R. Viral Inhibition Studies on Sulfated Lignin, a Chemically Modified Biopolymer and a Potential Mimic Heparan Sulfate. Biomacromolecules 2007, 8 (5), 1759-1763.
(27) Zoppe, J. O.; Ruottinen, V.; Ruotsalainen, J.; Ronkko, S.; Johansson, L.-S.; Hinkkanen, A.; Jarvinen, K.;
Seppala, J. Synthesis of Cellulose Nanocrystals Carrying Tyrosine Sulfate Mimetic Ligands and Inhibition of Alphavirus Infection. Biomacromolecules 2014, 15 (4), 1534-1542.
(28) Akkarawongsa, R.; Pocaro, N. E.; Case, G.; Kolb, A. W.; Brandt, C. R. Multiple Peptides Homologous to Herpes Simplex Virus Type 1 Glycoprotein B Inhibit Viral Infection Antimicrob Agents Chemother 2009, 53 (3), 987-996.
(29) Shogan, B.; Kruse, L.; Mulamba, G. B.; Hu, A.; Coen, D. M. Virucidal Activity of a GT-Rich Oligonucleotide against Herpes Simplex Virus Mediated by Glycoprotein B. J Virol 2006, 80 (10), 4740-4747.
(30) Moss, N.; Beaulieu, P.; Duceppe, J.-S.; Ferland, J.-M.; Garneau, M.; Gauthier, I; Ghiro, E.; Goulet, S.;
Guse, I.; Jaramillo, J.; Llinas-Brunet, M.; Malenfant, E.; Plante, R.; Poirier, M.; Soucy, F.; Wernic, D.; Yoakim, C; Deziel, R. Peptidomimetic Inhibitors of Herpes Simplex Virus Ribonucleotide Reductase with Improved in Vivo Antiviral Activity. J. Med. Chem. 1996, 39 (21), 4173-4180.
(31) Yudovin-Farber, I.; Gurt, I.; Hope, R.; Domb, A. J.; Katz, E. Inhibition of Herpes Simplex Virus by Polyamines. Antivir. Chem. Chemother. 2009, 20 (2), 87-98.
(32) Kaminski, K.; Plonka, M.; Ciejka, J.; Szczubialka, K.; Nowakowska, M.; Lorkowska, B.; Korbut, R.;
Lach, R. Cationic Derivatives of Dextran and Hydroxypropylcellulose as Novel Potential Heparin Antagonists. J. Med. Chem. 2011, 54 (19), 6586-6596.
(33) Sokolowska, E.; Kalaska, B.; Kaminski, K.; Lewandowska, A.; Blazejczyk, A.; Wietrzyk, J.; Kasacka, I.; Szczubialka, K.; Pawlak, D.; Nowakowska, M.; Mogielnicki, A. The Toxicokinetic Profile of Dex40- GTMAC3-A Novel Polysaccharide Candidate for Reversal of Unfractionated Heparin. Front. Pharmacol. 2016, 7 (MAR), 60.
(34) Reed, L. J.; Muench, H. A Simple Method of Estimating Fifty per Cent Endpoints. Am. J. Epidemiol.
1938, 27 (3), 493-497.
(35) Shukla, D.; Spear, P. G. Herpesviruses and Heparan Sulfate: An Intimate Relationship in Aid of Viral Entry. J. Clin. Invest. 2001, 108 (4), 503-510.
(36) Oh, M.-J.; Akhtar, J.; Desai, P.; Shukla, D. A Role for Heparan Sulfate in Viral Surfing. Biochem.
Biophys. Res. Commun. 2010, 391 (1), 176-181.
(37) Satoh, T.; Arii, J.; Suenaga, T.; Wang, J.; Kogure, A.; Uehori, J.; Arase, N.; Shiratori, I.; Tanaka, S.;
Kawaguchi, Y.; Spear, P. G.; Lanier, L. L.; Arase, H. PILRa Is a Herpes Simplex Virus-1 Entry Coreceptor That Associates with Glycoprotein B. Cell 2008, 132 (6), 935-944.
(38) Spear, P. G. Herpes Simplex Virus: Receptors and Ligands for Cell Entry. Cell. Microbiol. 2004, 6 (5), 401-410.
(39) Heldwein, E. E.; Lou, H.; Bender, F. C; Cohen, G. H.; Eisenberg, R. J.; Harrison, S. C. Crystal Structure of Glycoprotein B from Herpes Simplex Virus 1. Science (80-. ). 2006, 313 (5784), 217-220.
(40) Shukla, D.; Liu, J.; Blaiklock, P.; Shworak, N. W.; Bai, X.; Esko, J. D.; Cohen, G. H.; Eisenberg, R. I;
Rosenberg, R. D.; Spear, P. G. A Novel Role for 3 -O-Sulfated Heparan Sulfate in Herpes Simplex Virus 1 Entry. Cell 1999, 99 (1), 13-22.
(41) Atanasiu, D.; Whitbeck, J. C; Cairns, T. M.; Reilly, B.; Cohen, G. H.; Eisenberg, R. J. Bimolecular Complementation Reveals That Glycoproteins gB and gH/gL of Herpes Simplex Virus Interact with Each Other during Cell Fusion. Proc. Natl. Acad. Sci. U. S. A. 2007, 104 (47), 18718-18723.
(42) Taylor, J. M.; Lin, E.; Susmarski, N.; Yoon, M.; Zago, A.; Ware, C. F.; Pfeffer, K.; Miyoshi, J.; Takai, Y.; Spear, P. G. Alternative Entry Receptors for Herpes Simplex Virus and Their Roles in Disease. Cell Host Microbe 2007, 2 (1), 19-28.
(43) Tiwari, V.; Clement, C; Xu, D.; Valyi-Nagy, T.; Yue, B. Y. J. T.; Liu, J.; Shukla, D. Role for 3-0- Sulfated Heparan Sulfate as the Receptor for Herpes Simplex Virus Type 1 Entry into Primary Human Corneal Fibroblasts. J. Virol. 2006, 80 (18), 8970-8980.
(44) Gianni, T.; Salvioli, S.; Chesnokova, L. S.; Hutt-Fletcher, L. M.; Campadelli-Fiume, G. ανβ6- and ανβ8- Integrins Serve As Interchangeable Receptors for HSV gH/gL to Promote Endocytosis and Activation of Membrane Fusion. PLoSPathog. 2013, 9 (12), 1-14.

Claims

Claims
1. The use of dextran cationically modified by quaternary ammonium groups of structure I for the treatment and prophylaxis of diseases caused by herpes simplex viruses.
Figure imgf000020_0001
Structure I
2. The use according to claim 1, wherein dextran is modified using glycidyltrimethylammonium chloride (GTMAC).
3. The use according to claim 1 or 2, wherein dextran with molecular weight from 6 to 100 kDa is used.
4. The use according to claims 1-3, wherein cationically modified dextran has the degree of substitution in the range of 20-100%, preferably 20-60%, more preferably 25-60%).
5. The use according to claims 1-4, wherein cationically modified dextran is used in the form of ointment or drops applied topically to the skin or to the eye, orally, intraperitoneally, or intravenously in the systemic treatment.
PCT/IB2017/051918 2016-04-04 2017-04-04 The use of cationic derivative of dextran for inhibition of herpes simplex viruses WO2017175132A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PLP.416749 2016-04-04
PL416749A PL235490B1 (en) 2016-04-04 2016-04-04 Application of cationic derivative of dextrane for retardation of HSV-1 viral replication

Publications (1)

Publication Number Publication Date
WO2017175132A1 true WO2017175132A1 (en) 2017-10-12

Family

ID=58668929

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2017/051918 WO2017175132A1 (en) 2016-04-04 2017-04-04 The use of cationic derivative of dextran for inhibition of herpes simplex viruses

Country Status (2)

Country Link
PL (1) PL235490B1 (en)
WO (1) WO2017175132A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019177475A1 (en) * 2018-03-13 2019-09-19 Uniwersytet Jagielloński The use of copolymers for the preparation of a formulation for the treatment and prophylaxis of diseases caused by the hsv-1 virus

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0066379A2 (en) 1981-05-15 1982-12-08 Riker Laboratories, Inc. Composition for combating herpes virus
WO2000024784A1 (en) 1998-10-26 2000-05-04 Elena Alexandrovna Izaxon Dextran-containing biologically active substance
WO2006090365A2 (en) * 2005-02-22 2006-08-31 Yissum Research Development Company Polysaccharide-oligoamine conjugates as anti-amyloid and anti-viral agents
WO2011133052A2 (en) * 2010-04-22 2011-10-27 Uniwersytet Jagielloński Use of the modified polysaccharides for heparin neutralization

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0066379A2 (en) 1981-05-15 1982-12-08 Riker Laboratories, Inc. Composition for combating herpes virus
WO2000024784A1 (en) 1998-10-26 2000-05-04 Elena Alexandrovna Izaxon Dextran-containing biologically active substance
WO2006090365A2 (en) * 2005-02-22 2006-08-31 Yissum Research Development Company Polysaccharide-oligoamine conjugates as anti-amyloid and anti-viral agents
WO2011133052A2 (en) * 2010-04-22 2011-10-27 Uniwersytet Jagielloński Use of the modified polysaccharides for heparin neutralization

Non-Patent Citations (46)

* Cited by examiner, † Cited by third party
Title
"Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis", 2007, CAMBRIDGE
AKKARAWONGSA, R.; POCARO, N. E.; CASE, G.; KOLB, A. W.; BRANDT, C. R.: "Multiple Peptides Homologous to Herpes Simplex Virus Type 1 Glycoprotein B Inhibit Viral Infection", ANTIMICROB AGENTS CHEMOTHER, vol. 53, no. 3, 2009, pages 987 - 996, XP055080222, DOI: doi:10.1128/AAC.00793-08
ALASINO, R. V; AUSAR, S. F.; BIANCO, I. D.; CASTAGNA, L. F.; CONTIGIANI, M.; BELTRAMO, D. M: "Amphipathic and Membrane-Destabilizing Properties of the Cationic Acrylate Polymer Eudragit(t E100", MACROMOL. BIOSCI., vol. 5, no. 3, 2005, pages 207 - 213
ASAFTEI, S.; DE CLERCQ, E.: "Viologen''dendrimers as Antiviral Agents: The Effect of Charge Number and Distance", J. MED. CHEM., vol. 53, no. 9, 2010, pages 3480 - 3488, XP055087641, DOI: doi:10.1021/jm100093p
ATANASIU, D.; WHITBECK, J. C.; CAIRNS, T. M.; REILLY, B.; COHEN, G. H.; EISENBERG, R. J.: "Bimolecular Complementation Reveals That Glycoproteins gB and gH/gL of Herpes Simplex Virus Interact with Each Other during Cell Fusion", PROC. NATL. ACAD. SCI. U. S. A., vol. 104, no. 47, 2007, pages 18718 - 18723
BLOT, N.; SCHNEIDER, P.; YOUNG, P.; JANVRESSE, C.; DEHESDIN, D.; TRON, P.; VANNIER, J. P: "Treatment of an Acyclovir and Foscarnet-Resistant Herpes Simplex Virus Infection with Cidofovir in a Child after an Unrelated Bone Marrow Transplant", BONE MARROW TRANSPLANT., vol. 26, no. 8, 2000, pages 903 - 905
CIRELLI, R.; HEME, K.; MCCRARY, M.; LEE, P.; TYRING, S. K.: "Famciclovir: Review of Clinical Efficacy and Safety", ANTIVIRAL RES., vol. 29, no. 2-3, 1996, pages 141 - 151
DE REGGE, N.; VAN OPDENBOSCH, N.; NAUWYNCK, H. J.; EFSTATHIOU, S.; FAVOREEL, H. W.: "Interferon Alpha Induces Establishment of Alphaherpesvirus Latency in Sensory Neurons in Vitro", PLOS ONE, vol. 5, no. 9, 2010
DONALISIO, M.; RANUCCI, E.; CAGNO, V.; CIVRA, A.; MANFREDI, A.; CAVALLI, R.; FERRUTI, P.; LEMBO, D.: "Agmatine-Containing Poly(amidoamine)s as a Novel Class of Antiviral Macromolecules: Structural Properties and in Vitro Evaluation of Infectivity Inhibition", ANTIMICROB. AGENTS CHEMOTHER., vol. 58, no. 10, 2014, pages 6315 - 6319
FAROOQ, A. V; SHUKLA, D: "Herpes Simplex Epithelial and Stromal Keratitis: An Epidemiologic Update", SURV OPHTHALMOL, vol. 57, no. 5, 2012, pages 448 - 462
FATAHZADEH, M.; SCHWARTZ, R. A.: "Human Herpes Simplex Virus Infections: Epidemiology, Pathogenesis, Symptomatology, Diagnosis, and Management", J. AM. ACAD. DERMATOL., vol. 57, no. 5, 2007, pages 737 - 763, XP022380227, DOI: doi:10.1016/j.jaad.2007.06.027
GIANNI, T.; SALVIOLI, S.; CHESNOKOVA, L. S.; HUTT-FLETCHER, L. M.; CAMPADELLI-FIUME, G: "av|36- and av|38-Integrins Serve As Interchangeable Receptors for HSV gH/gL to Promote Endocytosis and Activation of Membrane Fusion", PLOSPATHOG., vol. 9, no. 12, 2013, pages 1 - 14
GONG, Y.; MATTHEWS, B.; CHEUNG, D.; TAM, T.; GADAWSKI, I.; LEUNG, D.; HOLAN, G.; RAFF, J.; SACKS, S: "Evidence of Dual Sites of Action of Dendrimers: SPL-2999 Inhibits Both Virus Entry and Late Stages of Herpes Simplex Virus Replication", ANTIVIRAL RES., vol. 55, no. 2, 2002, pages 319 - 329, XP001182382, DOI: doi:10.1016/S0166-3542(02)00054-2
HELDWEIN, E. E.; LOU, H.; BENDER, F. C.; COHEN, G. H.; EISENBERG, R. J.; HARRISON, S. C.: "Crystal Structure of Glycoprotein B from Herpes Simplex Virus 1", SCIENCE, vol. 313, no. 5784, 2006, pages 217 - 220, XP002571810, DOI: doi:10.1126/science.1126548
IRA YUDOVIN-FARBER ET AL: "Inhibition of Herpes Simplex Virus by Polyamines", ANTIVIRAL CHEMISTRY AND CHEMOTHERAPY, 1 October 2009 (2009-10-01), London, England, pages 87 - 98, XP055390002, Retrieved from the Internet <URL:http://journals.sagepub.com/doi/pdf/10.3851/IMP1401> DOI: 10.3851/IMP1401 *
KAMIRISKI, K.; PLONKA, M.; CIEJKA, J.; SZCZUBIALKA, K.; NOWAKOWSKA, M.; LORKOWSKA, B.; KORBUT, R.; LACH, R: "Cationic Derivatives of Dextran and Hydroxypropylcellulose as Novel Potential Heparin Antagonists", J. MED. CHEM., vol. 54, no. 19, 2011, pages 6586 - 6596, XP055067512, DOI: doi:10.1021/jm200380w
KNIPE, D. M.; HOWLEY, P. M: "Fields Virology", 2013, WOLTERS KLUWER/LIPPINCOTT WILLIAMS & WILKINS
KROPP, R. Y.; WONG, T.; CORMIER, L.; RINGROSE, A.; BURTON, S.; EMBREE, J. E.; STEBEN, M.: "Neonatal Herpes Simplex Virus Infections in Canada: Results of a 3-Year National Prospective Study", PEDIATRICS, vol. 117, no. 6, 2006, pages 1955 - 1962
LANGELAND, N.; MOORE, L. J.; HOLMSEN, H.; HAARR, L.: "Interaction of Polylysine with the Cellular Receptor for Herpes Simplex Virus Type 1", J GEN VIROL, vol. 69, 1988, pages 1137 - 1145
LEYDET, A.; BARRAGAN, V.; BOYER, B.; MONTERO, J. L.; ROQUE, J. P.; WITVROUW, M.; ESTE, J.; SNOECK, R.; ANDREI, G.; DE CLERCQ, E.: "Polyanion Inhibitors of Human Immunodeficiency Virus and Other Viruses. 5. Telomerized Anionic Surfactants Derived from Amino Acids", J. MED. CHEM., vol. 40, no. 3, 1997, pages 342 - 349
MOERDYK-SCHAUWECKER, M.; STEIN, D. A.; EIDE, K.; BLOUCH, R. E.; BILDFELL, R.; IVERSEN, P.; JIN, L: "Inhibition of HSV-1 Ocular Infection with Morpholino Oligomers Targeting ICPO and ICP27", ANTIVIRAL RES., vol. 84, no. 2, 2009, pages 131 - 141, XP026666292, DOI: doi:10.1016/j.antiviral.2009.07.020
MORFIN, F.; THOUVENOT, D: "Herpes Simplex Virus Resistance to Antiviral Drugs", J. CLIN. VIROL., vol. 26, no. 1, 2003, pages 29 - 37
MOSS, N.; BEAULIEU, P.; DUCEPPE, J.-S.; FERLAND, J.-M.; GARNEAU, M.; GAUTHIER, J.; GHIRO, E.; GOULET, S.; GUSE, I.; JARAMILLO, J.: "Peptidomimetic Inhibitors of Herpes Simplex Virus Ribonucleotide Reductase with Improved in Vivo Antiviral Activity", J. MED. CHEM., vol. 39, no. 21, 1996, pages 4173 - 4180, XP002072015, DOI: doi:10.1021/jm960324r
OH, M.-J.; AKHTAR, J.; DESAI, P.; SHUKLA, D.: "A Role for Heparan Sulfate in Viral Surfing", BIOCHEM. BIOPHYS. RES. COMMUN, vol. 391, no. 1, 2010, pages 176 - 181, XP026907877, DOI: doi:10.1016/j.bbrc.2009.11.027
QIU, M.; CHEN, Y.; SONG, S.; SONG, H.; CHU, Y.; YUAN, Z.; CHENG, L.; ZHENG, D.; CHEN, Z.; WU, Z.: "Poly (4-Styrenesulfonic Acid-Co-Maleic Acid) Is an Entry Inhibitor against Both HIV-1 and HSV Infections - Potential as a Dual Functional Microbicide", ANTIVIRAL RES., vol. 96, no. 2, 2012, pages 138 - 147
RAGHURAMAN, A.; TIWARI, V.; ZHAO, Q.; SHUKLA, D.; DEBNATH, A. K.; DESAI, U. R.: "Viral Inhibition Studies on Sulfated Lignin, a Chemically Modified Biopolymer and a Potential Mimic Heparan Sulfate", BIOMACROMOLECULES, vol. 8, no. 5, 2007, pages 1759 - 1763
REED, L. J.; MUENCH, H: "A Simple Method of Estimating Fifty per Cent Endpoints", AM. J. EPIDEMIOL., vol. 27, no. 3, 1938, pages 493 - 497
SACKS, S. L.; GRIFFITHS, P. D.; COREY, L.; COHEN, C.; CUNNINGHAM, A.; DUSHEIKO, G. M.; SELF, S.; SPRUANCE, S.; STANBERRY, L. R.; W: "HSV Shedding", ANTIVIRAL RES., vol. 63, no. 1, 2004, pages S19 - S26, XP004535002, DOI: doi:10.1016/j.antiviral.2004.06.004
SAHU, P. K.; UMME, T.; YU, J.; NAYAK, A.; KIM, G.; NOH, M.; LEE, J.-Y.; KIM, D.-D.; JEONG, L. S.: "Selenoacyclovir and Selenoganciclovir: Discovery of a New Template for Antiviral Agents", J. MED. CHEM., vol. 58, no. 21, 2015, pages 8734 - 8738
SATOH, T.; ARII, J.; SUENAGA, T.; WANG, J.; KOGURE, A.; UEHORI, J.; ARASE, N.; SHIRATORI, I.; TANAKA, S.; KAWAGUCHI, Y.: "PILRa Is a Herpes Simplex Virus-1 Entry Coreceptor That Associates with Glycoprotein B", CELL, vol. 132, no. 6, 2008, pages 935 - 944
SAUERBREI, A: "Optimal Management of Genital Herpes: Current Perspectives", INFECT DRUG RESIST, vol. 9, 2016, pages 129 - 141
SHOGAN, B.; KRUSE, L.; MULAMBA, G. B.; HU, A.; COEN, D. M.: "Virucidal Activity of a GT-Rich Oligonucleotide against Herpes Simplex Virus Mediated by Glycoprotein B", J VIROL, vol. 80, no. 10, 2006, pages 4740 - 4747
SHUKLA D ET AL: "Herpesviruses and heparan sulfate: an intimate relationship in aid of viral entry", JOURNAL OF CLINICAL INVESTIGATION, AMERICAN SOCIETY FOR CLINICAL INVESTIGATION, US, vol. 108, no. 4, 1 August 2001 (2001-08-01), pages 503 - 510, XP002493479, ISSN: 0021-9738 *
SHUKLA, D.; LIU, J.; BLAIKLOCK, P.; SHWORAK, N. W.; BAI, X.; ESKO, J. D.; COHEN, G. H.; EISENBERG, R. J.; ROSENBERG, R. D.; SPEAR,: "A Novel Role for 3-O-Sulfated Heparan Sulfate in Herpes Simplex Virus 1 Entry", CELL, vol. 99, no. 1, 1999, pages 13 - 22
SHUKLA, D.; SPEAR, P. G.: "Herpesviruses and Heparan Sulfate: An Intimate Relationship in Aid of Viral Entry", J. CLIN. INVEST., vol. 108, no. 4, 2001, pages 503 - 510, XP002493479
SMITH, J. S.; ROBINSON, N. J: "Age-Specific Prevalence of Infection with Herpes Simplex Virus Types 2 and 1: A Global Review", J INFECT DIS, vol. 186, 2002, pages 3 - 28
SOKOLOWSKA, E.; KALASKA, B.; KAMINSKI, K.; LEWANDOWSKA, A.; BLAZEJCZYK, A.; WIETRZYK, J.; KASACKA, I.; SZCZUBIALKA, K.; PAWLAK, D.: "The Toxicokinetic Profile of Dex40-GTMAC3-A Novel Polysaccharide Candidate for Reversal of Unfractionated Heparin", FRONT. PHARMACOL., vol. 7, 2016, pages 60
SPEAR, P. G: "Herpes Simplex Virus: Receptors and Ligands for Cell Entry", CELL. MICROBIOL., vol. 6, no. 5, 2004, pages 401 - 410
STRICK, L. B.; WALD, A.; CELUM, C.: "Management of Herpes Simplex Virus Type 2 Infection in HIV Type 1-Infected Persons", CLIN. INFECT. DIS., vol. 43, no. 3, 2006, XP002509747, DOI: doi:10.1086/505496
SZCZUBIALKA, K.; PYRE, K.; NOWAKOWSKA, M.: "Search for Effective and Definitive Treatment of Herpes Simplex Virus Type 1 (HSV-1) Infections", RSCADV., vol. 6, no. 2, 2016, pages 1058 - 1075
TAYLOR, J. M.; LIN, E.; SUSMARSKI, N; YOON, M.; ZAGO, A.; WARE, C. F.; PFEFFER, K.; MIYOSHI, J.; TAKAI, Y.; SPEAR, P. G: "Alternative Entry Receptors for Herpes Simplex Virus and Their Roles in Disease", CELL HOST MICROBE, vol. 2, no. 1, 2007, pages 19 - 28
TIWARI, V.; CLEMENT, C.; XU, D.; VALYI-NAGY, T.; YUE, B. Y. J. T.; LIU, J.; SHUKLA, D.: "Role for 3-0-Sulfated Heparan Sulfate as the Receptor for Herpes Simplex Virus Type 1 Entry into Primary Human Corneal Fibroblasts", J. VIROL., vol. 80, no. 18, 2006, pages 8970 - 8980
WILSON, A. C.; MOHR, I.: "A Cultured Affair: HSV Latency and Reactivation in Neurons", TRENDS MICROBIOL., vol. 20, no. 12, 2012, pages 604 - 611
YUDOVIN-FARBER, I.; GURT, I.; HOPE, R.; DOMB, A. J.; KATZ, E: "Inhibition of Herpes Simplex Virus by Polyamines", ANTIVIR. CHEM. CHEMOTHER., vol. 20, no. 2, 2009, pages 87 - 98, XP055390002, DOI: doi:10.3851/IMP1401
ZEITLIN, L.; WHALEY, K. J.; HEGARTY, T. A.; MOENCH, T. R.; CONE, R. A.: "Tests of Vaginal Microbicides in the Mouse Genital Herpes Model", CONTRACEPTION, vol. 56, no. 5, 1997, pages 329 - 335
ZOPPE, J. O.; RUOTTINEN, V.; RUOTSALAINEN, J.; RONKKO, S.; JOHANSSON, L.-S.; HINKKANEN, A.; JARVINEN, K.; SEPPALA, J.: "Synthesis of Cellulose Nanocrystals Carrying Tyrosine Sulfate Mimetic Ligands and Inhibition of Alphavirus Infection", BIOMACROMOLECULES, vol. 15, no. 4, 2014, pages 1534 - 1542

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019177475A1 (en) * 2018-03-13 2019-09-19 Uniwersytet Jagielloński The use of copolymers for the preparation of a formulation for the treatment and prophylaxis of diseases caused by the hsv-1 virus

Also Published As

Publication number Publication date
PL416749A1 (en) 2017-10-09
PL235490B1 (en) 2020-08-24

Similar Documents

Publication Publication Date Title
Milewska et al. HTCC as a Polymeric Inhibitor of SARS-CoV-2 and MERS-CoV
Li et al. Inhibition of herpes simplex virus by myricetin through targeting viral gD protein and cellular EGFR/PI3K/Akt pathway
Akula et al. Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) infection of human fibroblast cells occurs through endocytosis
Milewska et al. Novel polymeric inhibitors of HCoV-NL63
Gao et al. The inhibitory effects and mechanisms of 3, 6-O-sulfated chitosan against human papillomavirus infection
Gong et al. Evidence of dual sites of action of dendrimers: SPL-2999 inhibits both virus entry and late stages of herpes simplex virus replication
Lin et al. Hydrolyzable tannins (chebulagic acid and punicalagin) target viral glycoprotein-glycosaminoglycan interactions to inhibit herpes simplex virus 1 entry and cell-to-cell spread
Spoden et al. Polyethylenimine is a strong inhibitor of human papillomavirus and cytomegalovirus infection
Mercorelli et al. Early inhibitors of human cytomegalovirus: state-of-art and therapeutic perspectives
Pachota et al. Inhibition of herpes simplex viruses by cationic dextran derivatives
Donalisio et al. The AGMA1 poly (amidoamine) inhibits the infectivity of herpes simplex virus in cell lines, in human cervicovaginal histocultures, and in vaginally infected mice
Szczubiałka et al. In search for effective and definitive treatment of herpes simplex virus type 1 (HSV-1) infections
Loutfy et al. Antiviral activity of chitosan nanoparticles encapsulating silymarin (Sil–CNPs) against SARS-CoV-2 (in silico and in vitro study)
Shoup et al. Substitution-inert polynuclear platinum compounds inhibit human cytomegalovirus attachment and entry
Synowiec et al. Cat flu: Broad spectrum polymeric antivirals
US20240156966A1 (en) A sirna drug, a pharmaceutical composition, a sirna-small molecule drug conjugate, and the application thereof
Wild et al. The trimeric artesunate derivative TF27 exerts strong anti-cytomegaloviral efficacy: Focus on prophylactic efficacy and oral treatment of immunocompetent mice
Wang et al. Sulphated glucuronomannan tetramer and hexamer from Sargassum thunbergii exhibit anti-human cytomegalovirus activity by blocking viral entry
Pachota et al. Highly effective and safe polymeric inhibitors of herpes simplex virus in vitro and in vivo
WO2017175132A1 (en) The use of cationic derivative of dextran for inhibition of herpes simplex viruses
EP2616478A2 (en) Anti-heparan sulfate peptides that block herpes simplex virus infection in vivo
ITTO20100415A1 (en) ANTI-VIRAL AGENTS AND RELATED COMPOSITIONS
CN101864421B (en) Structure and application of target programmed cell death protein 5 (PDCD5) anti-influenza virus oligonucleotide
Liu et al. Emerging drug design strategies in anti-influenza drug discovery
US9464113B2 (en) Anti-heparan sulfate peptides that block herpes simplex virus infection in vivo

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17721209

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17721209

Country of ref document: EP

Kind code of ref document: A1