WO2015116531A1 - Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques - Google Patents

Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques Download PDF

Info

Publication number
WO2015116531A1
WO2015116531A1 PCT/US2015/012894 US2015012894W WO2015116531A1 WO 2015116531 A1 WO2015116531 A1 WO 2015116531A1 US 2015012894 W US2015012894 W US 2015012894W WO 2015116531 A1 WO2015116531 A1 WO 2015116531A1
Authority
WO
WIPO (PCT)
Prior art keywords
phage
phage particle
therapeutic
nucleic acid
particle according
Prior art date
Application number
PCT/US2015/012894
Other languages
English (en)
Inventor
Todd Bernard PARSLEY
Christian Furlan FREGUIA
Original Assignee
Synphagen Llc.
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 Synphagen Llc. filed Critical Synphagen Llc.
Priority to JP2016548341A priority Critical patent/JP6755801B2/ja
Priority to US15/114,634 priority patent/US10351452B2/en
Priority to EP15742945.7A priority patent/EP3099173A4/fr
Priority claimed from US14/605,320 external-priority patent/US9676641B2/en
Publication of WO2015116531A1 publication Critical patent/WO2015116531A1/fr
Priority to US18/198,519 priority patent/US20230285508A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)

Definitions

  • the present invention relates to systems, compositions and methods to produce therapeutic bacteria phages capable of delivering nucleic acids to bacteria, modified phages and the use of the modified phages to deliver nucleic acids to bacteria.
  • the ability to deliver genetic information to cells and program the cells for the production of a therapeutic agent is a powerful tool amenable to several applications including human health, industrial production, agricultural production, biotechnology, and cosmetics.
  • Bacteria are present in the gut, mucosal tissues, and skin, as well as other environments in the body. Alterations of the commensal micro-organisms have been associated with several diseases, such as diabetes, irritable bowel syndrome, obesity, and cancer. In ruminants, complex microbiomes are essential to convert plant cell wall biomass into proteins and fatty acids, and companion animals display a highly complex microbial gastrointestinal ecosystem which influences disease states. Similarly, plants exhibit a broad range of relationships with symbiotic microorganisms that result in nutrient exchanges. [0004] Over the past decade there has been considerable research directed towards understanding the relationship between the human body and the vast number of microbes that cohabitate it (i.e., the human microbiome).
  • the human intestine harbors an enormously complex, diverse, and vast microbial community, referred to as the gut microbiota (or microbiome).
  • the human gut microbiota is estimated to consist of 10 14 bacteria and archaea. In its entirety, the gut microbiota is estimated to contain 150-fold more genes than human host genomes.
  • this unique and independent ecosystem has enormous potential for physiological and pathological interactions with the host, for example, as a target for the phage gene therapy embodiments described in the present invention.
  • Phages in their most basic definition are viruses that infect bacteria.
  • the use of phages for the treatment of bacterial infections (known as phage therapy) is known.
  • phages have been used in antibacterial therapy and biotechnology as antimicrobial targeting infectious agents for both medical and industrial purposes as well as for research in gene discovery and protein expression.
  • phage therapy is the therapeutic use of bacteriophages to treat pathogenic bacterial infections.
  • Such conventional phages have been used therapeutically to treat bacterial infections that do not respond to conventional antibiotic drugs.
  • This treatment involves the infection of a pathogenic or targeted bacteria by the phage and destruction of the bacteria via the lytic cycle of the phage replication pathway, thus eliminating the bacteria.
  • Conventional methods for creating and utilizing such bacteria phages for antimicrobial purposes have been developed and used primarily in Russia and Europe.
  • Phages have also been utilized for research of various prokaryotic and eukaryotic systems and many of the basic concepts of modern molecular biology are a result of studying the genetics of phages. Because phages can accommodate the insertion of large amounts of heterologous nucleic acids, the phage is an ideal vehicle for the cloning and expression of transgenic material. Indeed, several industrial and biotechnical applications of phage are known. Primary applications in biotechnology include the use of bacteria phage for nucleic acid or genetic "library” screening, the generation of single stranded DNA for sequencing (a utility which has become obsolete with advances in DNA sequencing technologies) and phage display. Such conventional technologies rely on the ability of the recombinant phage to replicate and form infectious particles that can be amplified either on their own or with the assistance of a helper phage.
  • phage display is a laboratory technique for the study of protein- protein, protein-peptide, and protein-DNA interactions that uses bacteriophages to connect proteins with the genetic information that encodes them.
  • a gene encoding a protein of interest is inserted into a phage coat protein gene, causing the phage to "display" the protein on its outside while containing the gene for the protein on its inside, resulting in a connection between genotype and phenotype.
  • These displaying phages can then be screened against other proteins, peptides or DNA sequences, in order to detect possible interaction between the displayed protein and those other molecules. In this way, large libraries of proteins can be screened and amplified in a process called in vitro selection.
  • Such displaying phages are not designed to target particular bacteria but only to determine possible interaction with an array of proteins.
  • applications of phage display technology include determination of interaction partners of a protein (which would be used as the immobilised phage "bait" with a DNA library consisting of all coding sequences of a cell, tissue or organism) so that the function or the mechanism of the function of that protein may be determined.
  • nucleic acid therapy there are major limitations to effective delivery and clinical utilization related to stability, pharmacokinetics, intracellular target accessibility, and specificity of target tissue. Many different approaches have been taken to overcome these limitations, such as different nucleic acid encapsulation strategies, mechanical and electrical techniques for introduction of nucleic acids into cells, and viral-based delivery systems.
  • phages bacteriophages
  • the use of phages as described in the various embodiments of the present invention is analogous to a mammalian virus- based gene therapy vector such as adenovirus and lentiviral vectors used for the targeted delivery and expression of genes in eukaryotic cells; however, the present invention relates to the expression of genes and gene products in prokaryotic cells.
  • aspects of the present invention take advantage of the commensal relationship between the human host and the microbiome for the targeted delivery of nucleic acid therapies.
  • a novel platform technology is disclosed to effectively deliver nucleic acids to program bacteria for expression of therapeutic proteins and RNA molecules in vivo at sites of greatest significance for a particular disease. This approach has a higher local concentration of the therapeutic and reduces/minimizes systemic/off-target effects than conventional means. Bacteria associated with mucosal surfaces can also be exploited for the generation of novel vaccines that are more efficacious, safer and less expensive to produce than current vaccines.
  • Another embodiment of the present invention relates to biological particles based on a filamentous bacteriophage platform engineered to target specific bacteria within the microbiome of an organism for delivery of nucleic acid therapies and expression of therapeutic genes.
  • the bacteriophage-derived nanoparticles (BNPs) target specific bacteria in vivo.
  • the BNPs carry the nucleic acids encoding the therapeutic gene(s) of interest which, once delivered, will be expressed in the target bacteria.
  • This embodiment differs from conventional approaches for nucleic acid delivery to eukaryotic cells and uses 'microbial gene therapy' as a method for nucleic acid delivery for the treatment of human disease.
  • This embodiment also differs from conventional phage therapy which uses lytic phage for antibacterial purposes.
  • the inventive delivery platform does not kill the bacteria; rather it takes advantage of live bacteria for expression of therapeutic nucleic acids in vivo, making the commensal organism a site-specific therapeutic "factory". This has the advantages of delivering therapeutic nucleic acids at the biological site of greatest significance for a specific therapy, thus increasing the local concentration of the wanted therapeutics and diminishing the systemic effects.
  • BNPs programmed with a luciferase reporter gene can be constructed and characterized in vitro and in vivo as a model for delivery of nucleic acids encoding peptide therapeutics.
  • an inventive delivery platform is tunable and capable of encoding single or multiple genes of various functions that may be placed under different regulatory control mechanisms and can be modified to deliver its payload to different commensal bacterial species.
  • the delivery platform can be programmed for delivery of therapeutic DNA and RNA and has broad-based applications for expression of therapeutic proteins, vaccination strategies and modulation of bacterial biological pathways linked to human's health and disease.
  • Another aspect of the present invention uses the fact the BNPs are stable, amenable to many formulations, have no payload constraint in terms of nucleic acid sequence and no immunogenicity issues.
  • the inherent high stability of phage particles, their ease of production and the modular nature of this delivery platform will allow the targeted delivery of nucleic acid therapeutics to strategic areas of the host.
  • Another aspect of the present invention relates to methods for the creation of therapeutic phage particles by modified bacteria containing helper phage sequences and by specific phagemids. Bacteria alone with or without the helper phage sequences cannot generate therapeutic phage particles. Likewise, phagemids encoding the therapeutic phage alone cannot generate the therapeutic phages. Specific therapeutic phage particles are generated only when bacteria are modified with both the modified helper phage sequences and the phagemids. In one aspect, embodiments of the present invention differ from that of phage display in that the therapeutic phage lacks specific phage genes such that therapeutic phage particles may only be formed in the context of the packaging cell line.
  • inventions of the present invention also differ from that of phage display technology in that the therapeutic gene sequence is inserted into the phagemid as an autologous gene cassette and not in frame with the pill protein coding sequence for display on the phage surface.
  • the therapeutic phage particles are used as delivery vehicles for the transduction of nucleic acid sequence into specific target bacteria in vivo (the host organism) or to bacteria in the environment.
  • Yet another embodiment of the present invention relates to a stable bacterial host strain that contains a modified helper phage genome, such as, but not restricted to, filamentous M13 helper phage of Escherichia coli (E. coli), integrated into a bacterial host genome, although application of the technology is not dependent on integration of the sequences into the host genome.
  • a helper phage by definition is a phage that is able to supply packaging functions in trans to a filamentous phage that itself does not encode all the necessary genes for replication and packaging, but can be packaged into an infectious phage particle if introduced into a bacterial strain harboring the helper phage.
  • the bacterial host strain is generated by transformation of a bacterial strain with a plasmid encoding modified helper phage genes.
  • the bacterial strain may be any in which the modified helper phage and phagemid can function together to produce the specific therapeutic phage particle.
  • E. coli is used as an example bacterial strain.
  • the modified helper phage encoded within the plasmid has the following attributes, 1 ) it is non-lytic; 2) encodes the phage enzymes and nucleic acid sequences necessary for replication and packaging of a heterologous phage supplied in trans by therapeutic phagemid sequences; 3) is devoid of a packaging signal, and 4) may have a non- antibiotic selectable marker.
  • a phagemid by definition is a DNA plasmid that is capable of replication in bacteria as a plasmid and also encodes phage sequences, including a phage origin of replication and packaging signals that allow for replication and packaging of the encoded sequences into an infectious phage particle when present in a bacterial cell harboring a helper phage.
  • the phagemid genes encode a genetically engineered non-replicating, non-lytic phage with the following attributes: 1 ) the phagemid encodes the therapeutic gene sequence(s) under the regulatory control of a bacterial or phage promoter; 2) an origin of replication (o ) for replication in the host packaging strain; 3) the phage structural genes encoding elements necessary for recognition of a target bacterial strain (phage receptor binding protein (RBP)), attachment and entry into the targeted bacterial host; 4) signal sequences necessary for amplification and packaging by the helper phage functions; 5) may have a non-antibiotic selectable marker; multiple cloning sites flanking the therapeutic gene sequence(s), phage attachment and regulatory elements to allow for modular combinations of gene sequences; and/or 6) may contain sequence elements necessary for the integration of the phage into the target host genome.
  • the combination of the host packaging cell line and the therapeutic phagemid by transformation of the packaging cell line with the phagemid sequences results in the synthesis and packaging of the phagemid DNA into a bacteriophage particle that can act as a delivery vehicle for a specific therapeutic gene.
  • the therapeutic phage generated by combination of the (modified) host packaging bacteria cell line and the phagemid present includes one or more key features such as 1 - replication deficiency; 2- non-lytic; 3- carrying exogenous genetic material.
  • This therapeutic bacteriophage particle may then be delivered to the site of therapy in which the target bacteria resides as specified by the RBP encoded in the therapeutic phagemid, and may be delivered according to any of the modes of therapeutic application as needed, described below.
  • Another embodiment of the present invention is directed to a method for the generation of a therapeutic phage including the steps of modifying a bacteria to contain a helper phage sequence, using a phagemid including a nucleic acid sequence and generating the therapeutic phage when the modified bacteria and the phagemid are together.
  • Fig. 1 shows a composition of a helper phage plasmid 1 .
  • Fig. 2 shows an example of a packaging cell line (or a modified bacteria) 2.
  • Fig. 3 shows a composition of a phagemid 3 .
  • Fig. 4 shows a composition of a therapeutic phage particle 4.
  • the modified bacteria 2 contains a (modified) helper phage sequences 1 a.
  • modified bacterium 2 can be generated several ways, e.g. utilizing molecular biology techniques and a bacterial transposon system.
  • a specific phagemid 3 is constructed to encode phage infectivity sequence (pill), phage packaging signal and the therapeutic gene (or genes) of interest. The expression of any sequences can be under the regulatory control of inducible promoters.
  • the helper phage plasmid 1 includes sequences of a helper phage (Cassette I) containing genes that encode the packaging and replication functions for bacteriophage, but lacks a packaging signal and may lack a competent phage receptor binding protein (RBP), coding sequences that are determinants for packaging of a helper phage genome (packaging signal sequence) and specificity in infection by the phage (RBP).
  • the helper phage plasmid 1 may be sequences of the M13 filamentous helper phage, or other phage sequences or any combination thereof, necessary to replicate and package heterologous phage sequences in trans.
  • helper phage genes may be under the regulatory control of an inducible promoter, such that the phage proteins are only expressed upon activation by an added stimulus.
  • transcription of the helper phage genes may only be activated by protein(s) or peptide(s) encoded in the therapeutic phagemid 3 (Fig. 3). In this case, transformation of the host packaging strain with the therapeutic phagemid 3 would result in expression of the helper phage genes necessary to replicate and package the phagemid sequences resulting in coordinated production of the therapeutic phage particle 4 (Fig. 4).
  • the helper phage plasmid shown in Fig. 1 also encodes a non-antibiotic selectable marker (Cassette II) that allows for selection of the transformed host bacterial strain, which will harbor the helper phage elements.
  • the selectable marker may encode a specific antitoxin necessary for replication of the transformed bacteria on growth medium containing the toxin, or may encode a metabolic function that allows the transformed bacteria to grow on medium deficient in an essential nutrient.
  • the plasmid encoding the helper phage sequences would be transformed into an auxotrophic host strain for example, an E. coli strain that contains a deletion in the glyA gene.
  • the glyA gene encodes serine hydroxymethyl transferase, an enzyme involved in the biosynthetic pathway for the amino acid glycine. This strain can grow only if glycine is added to the culture medium or if it transformed with a plasmid expressing a functional glyA gene.
  • a third component may also be present in the helper phage plasmid 1 that encodes sequences necessary for the stable integration of the plasmid into the bacterial host genome, although application of the technology is not dependent on integration of the sequences into the host genome. These sequences may be those of a bacterial transposon, or may be any other genetic element that facilitates stable integration into the host genome.
  • Cassette IV in the helper phage plasmid 1 construct contains elements necessary for propagation and amplification of the plasmid sequences in host bacteria such as an E. coli origin of replication (Ori) and a selectable marker.
  • the selectable marker may confer antibiotic resistance.
  • These sequences may be those of a common commercially available bacterial plasmid. Implied within the sequences are engineered and endogenous endonuclease restriction sites necessary for cloning and insertion of phage and associated gene modules, and transcriptional promoters and terminators necessary for regulation of bacterial and phage gene expression.
  • Fig. 2 shows an example of the packaging cell line 2 (i.e., a phage packaging strain or a bacterial strain) produced by transformation with the helper phage plasmid 1 .
  • the bacterial strain has the helper phage plasmid integrated into the bacterial chromosome.
  • the bacterial strain may be any in which the modified helper phage sequence 1 a and the specific phagemid 3 (Fig. 3) can function to produce the therapeutic phage particle 4 (Fig. 4).
  • the bacterial strain is selected and the genotype maintained by culture on selective growth medium.
  • the bacterial strain itself is incapable of producing infectious phage particles and the helper phage sequence 1 a are incapable of transmission due to the lack of packaging signals in the helper phage sequence 1 a.
  • the stable integration of the helper phage sequences 1 a into the host bacteria results in the insertion of Cassettes I and II into the host chromosome. Recombination necessary for insertion of Cassettes I and II would result in the loss of Cassette IV.
  • Fig. 3 shows an example of the phagemid 3 construct.
  • the phagemid 3 includes phage sequences necessary for the synthesis and packaging of the encoded genome (Cassette I) in the presence of helper phage plasmid 1 .
  • the encoded genome encodes a therapeutic function.
  • the therapeutic function may be a known therapeutic value or an experimental therapeutic.
  • a gene that encodes an enzyme that breaks down gluten (glutenase) for the treatment of gluten intolerance e.g., celiac spruce disease
  • the therapeutic phage particle 4 (FIG. 4) would be one that targets a bacteria in the gut.
  • the therapeutic phage particle 4 containing the glutenase gene When introduced into the gut, the therapeutic phage particle 4 containing the glutenase gene would infect the target bacteria in the gut, thereby causing the target bacteria to make and excrete the enzyme at the site in the body where it was needed. Modifications could be made to any of the therapeutic genes to regulate the level of expression or excretion from the host bacteria, or to help the therapeutic product to cross a biological barrier (such as the gut lumen) once it is expressed and excreted. [0034]
  • the applications are not limited to the gut, as there are commensal microbes associated with the oral cavity, nasal cavities, skin, etc that could be targeted with the therapeutic phage particles 4 of the present invention.
  • the human scalp harbors a beautiful array of commensal bacteria (the microbiome) which form a continuous layer on the epidermis of the scalp. These commensal bacteria are also found in direct association with the hair follicle and in the subdermal tissues.
  • the bacteria comprising the dermal microbiome occupy prime real estate for treatment of dermatological maladies and are an ideal target for in vivo gene therapy.
  • the bacteria in the hair follicles can be targeted with a specific therapeutic phage particle 4 that encodes a protein that promotes hair growth.
  • the phage sequences in the phagemid 3 may also include genes that help maintain the stability of the phage in target bacteria.
  • An example of maintenance genes include the Defense against Restriction genes darA and darB of P1 phage to assist in the stability of the transduced DNA.
  • the P1 phage genome is greatly protected from type I restriction and modification systems in target bacteria, even though P1 phage DNA is a good substrate for type I restriction enzymes in vitro. This protection is due to the presence of darA and darB gene products found in the phage head and injected into recipient cells along with the DNA.
  • the therapeutic sequence(s) are encoded in Cassette II of the phagemid 3 construct and are expressed under the regulatory control of a bacterial or phage promoter (P2) that is functional in vivo in the target bacteria.
  • the promoter may be constitutive in nature or may be regulated by environmental stimuli, such that the therapeutic gene(s) would be expressed at a steady rate, or only within the context of a specific environmental stimuli, respectively.
  • the therapeutic sequence(s) may encode a single gene, multiple genes, chimeric proteins, DNA sequences or regulatory RNA such as small interfering RNA (siRNA), non-coding RNA or microRNAs (miRNA), or any precursor of such regulatory RNA molecules.
  • Encoded proteins may include signal peptides to aid in the excretion of the gene product(s) and/or other specific sequences to aid in the delivery, stability and activity of the gene product, depending on the therapeutic application.
  • Cassette III of the phagemid 3 encodes phage sequences including, but not limited to those which encode the receptor binding protein and determines the specificity and range of bacteria targeted for infection with the therapeutic phage particle 4.
  • the phagemid 3 may also contain DNA elements that facilitate integration into the genome of the targeted bacteria.
  • the g3p of the M13 bacteriophages consists of three globular domains: two N-terminal domains function in penetration and adsorption of the phage and the C-terminal domain anchors the g3p to the virion.
  • This structure/function relationship of g3p has been used in the development and application of conventional phage display.
  • BNPs can be created that are capable of delivering nucleic acids to those bacteria at biologically relevant sites in vivo.
  • Fig. 4 shows the production of the therapeutic phage particle 4 by introduction of the phagemid 3 into the packaging cell line 2 (i.e., the bacterial strain). Transformation of the packaging cell line 2 with the phagemid 3 construct encoding the therapeutic gene sequence(s) and the receptor binding protein results in the production of the therapeutic phage particles 4.
  • the therapeutic phage particles 4 may be delivered in vivo by a variety of routes (i.e.
  • the therapeutic phage particle 4 may also be applied to the environment (directly or indirectly) to an insect vector capable of transmission of a pathogen. This application, for example, includes the use of the therapeutic phage particle 4 containing one or more genes encoding a product that would disrupt the replication cycle of malaria or dengue virus within a mosquito host.
  • the therapeutic phage particle 4 may have several features such as being non-lytic and incapable of sustained independent replication.
  • the lytic feature may be abrogated by mutations or deletion of the gene(s) responsible for it.
  • gene(s) that sustain phage replication in bacteria are silenced by deletion of the genetic material or by mutations.
  • the therapeutic phage particles 4 may be used in vivo.
  • the therapeutic phage particles 4 may be specific for any species of bacteria or may infect a range of bacteria and the specificity will determine the site of delivery, i.e. phage specific for dermal microbes, microbes in hair follicles, microbes in the upper intestinal tract, in the lower intestinal tract, the duodenum, vaginal environment or any other specific site in humans or animals.
  • the therapeutic phage particles 4 are used to infect specific bacteria within the microbiome of a host organism (human, animal, or plant) or within the environment (e.g., soil).
  • application of the therapeutic phage particle 4 is coupled with consumption of a target bacteria in the form of a probiotic preparation, a topical application, or other appropriate means of application.
  • laboratory data supporting topical application has been demonstrated by a topical application of the therapeutic phage 4 and targeted expression of a report gene on mouse skin. This laboratory data was gathered by constructing a 2-plasmids system to generate therapeutic phage particles 4 that specifically contain the green fluorescent protein (GFP) sequence.
  • GFP green fluorescent protein
  • the bacteriophage nanoparticle (BNP) platform is composed of a therapeutic phagemid and a filamentous phage packaging plasmid.
  • the therapeutic phagemid is a modular shuttle plasmid capable of replication in both the target bacterium and E.coli, used for production, and carries the therapeutic nucleic acids.
  • the therapeutic plasgemid contains a filamentous phage origin of replication, a chimeric M13 phage g3p protein, for targeting of specific bacteria, and the packaging signal sequence, necessary for replication and incorporation of the phagemid ssDNA into the BNP.
  • the packaging plasmid encodes sequences necessary for replication and assembly of the bacteriophage particle, but is devoid of the phage origin of replication, packaging signal, and g3p gene. The combination of the two plasmids results in the production of BNPs that contain only the phagemid DNA sequences and not the packaging plasmid.
  • the laboratory data demonstrated the delivery of a reporter gene to E.coli in vitro and in vivo.
  • the therapeutic phagemid was engineered based on the M13 bacteriophage to carry the GFP cDNA, ORI, g3p and packaging signal sequences.
  • the packaging plasmid encodes sequences necessary for replication and assembly of the M13 bacteriophage.
  • BNPs were generated by co-transfection of the two plasmids into competent DH5a cells and purified by PEG precipitation. Individual preparations of the GFP-programed BNPs were prepared and used to transduce E. coli. The individual preparations were first assayed to assure selective packaging of the phagemid sequences into the BNP by PCR.
  • E.coli K12 cells were then transduced with the six GFP-programed BNPs and plated onto selective medium (kanamycin for the therapeutic phagemid selection) resulting in growth of bacteria transduced with BNPs only. When analyzed by flow cytometry, the transduced bacteria showed intense green fluorescent signal, demonstrating delivery and expression of the packaged genetic informatio.
  • a skin-abrasion model on Balb/C mice was employed to test the ability of BNPs to deliver the nucleic acid cargo in vivo. E. coli bacteria were applied to the skin after mild abrasion and BNPs or vehicle alone (TBS) were topically added. E. coli transduced in vitro with the GFP- programed BNPs were applied to the skin of mice as positive control.
  • the therapeutic phage particle 4 includes therapeutic gene(s) sequences 4a that may encode a single or multi-functional protein, peptide, nucleic acid such as miRNA, shRNA or siRNA, or any other envisioned molecule of therapeutic value.
  • the therapeutic gene (or genes) 4a can encode for proteins, peptides, decoys, antibodies and any other therapeutically relevant molecules (called products).
  • the encoded therapeutic product may be designed to be secreted from the infected bacterial host, or may be designed to be expressed on the surface of the infected host or may be designed to affect specific biological pathways in the targeted bacterial host.
  • the therapeutic products can be secreted and have phenotypical effects on eukaryotic and/ or prokaryotic target cells.
  • the phenotypic changes are intended to be any modifications that lead to a biological effect or multiple effects.
  • the therapeutic products can also affect internal biological pathways of the host bacteria cells or the eukaryotic cells.
  • the therapeutic products can be exposed on the host cell membrane and non-secreted.
  • the therapeutic products can be na ' fve or recombinant derived from molecular biology techniques of the nucleic acid material such as cloning, mutagenesis, recombination, or shuffling.
  • the therapeutic products can also be non-therapeutic and produce phenotypic changes in prokaryotic and eukaryotic cells, such as skin tanning, teeth whitening or suppression of odor (sensory or creation).
  • the therapeutic products can also affect the immune system by inducing an immune response or by creating immune tolerance.
  • the therapeutic phage particles 4 can target a specific bacteria strain and/or the therapeutic phage particles 4 can have a broad spectrum of bacteria targets.
  • the specificity is dictated by the capsid and recombinant pill , or tail fiber proteins that can be derived from one phage strain or can be a hybrid combination from two or more phage strains, or can be a hybrid of phage and any peptide or protein that facilitates attachment and entry of the particle to the target organism.
  • the therapeutic phage particles 4 can be delivered using any appropriate pharmaceutical formulation, e.g., ointments, gels, patches, lotion, shampoo, beverage, or freeze dried phage, using one or more delivery routes, e.g., oral, topical, parenteral, mucosal, and may be formulated for time-release delivery.
  • the pharmaceutical formulation can contain one strain of phages with proper bacteria specificity or two or more phage strains to target multiple bacteria strains.
  • the therapeutic phage particles 4 can be used for treatment of metabolic syndromes, oral hygiene, cosmetic products, vaccination, immunotolerance, protein replacements, agriculture, and industrial products, or any other envisioned appropriate therapeutic or cosmetic application.
  • the therapeutic phage particles 4 can be applied to the environment (soil, or water) to eliminate a toxin or environmental contamination, such as in an industrial chemical spill or waste product.
  • the therapeutic phage particles 4 can also be applied to waste water or industrial waste or byproduct to decontaminate or detoxify the waste.
  • the therapeutic phage particles 4 may be co-administered with the target bacteria.
  • the therapeutic phage particles 4 can applied to industrial or environmental material such as but not limited to agricultural or food production waste to produce or improve on the production a metabolic product.
  • mucosal vaccines such as dilution in mucosal secretions, entrapment in mucus gels, inactivation by proteases and nucleases, and exclusion by epithelial barriers. This means that relatively large doses of vaccine may be required for mucosal administration.
  • the therapeutic phage particles 4 can be used to improve delivery of vaccines.
  • the therapeutic phage particles 4 are used to target bacteria within the upper respiratory tract, lung or gut and deliver genes programmed to express appropriate antigens and/or immunomodulators, which results in T and/or B cell response.
  • the target bacteria will express and excrete the antigenic protein and/or immunomodulator that will be recognized by neighboring immune cells, eliciting an immune response. This results in a stronger immune response due to the close relationship of commensal bacteria with lymphocytes.
  • This approach has the advantage of enacting both the humoral and cellular arms of the immune system.
  • E. coli DH5a T1 r cells transformed with reporter phagemid and M13 packaging plasmid can be cultured in 2XTY medium and particles will be PEG precipitated from culture medium.
  • bacteriophage nanoparticles specific for Pseudomonas aeruginosa in vitro and in vivo.
  • Two therapeutic shuttle phagemids encoding g3p minor coat protein chimeras consisting of the N-terminal sequence from Pseudomonas filamentous phage pf1 (ORF437) or pf3 (ORF483) and the C-terminal domain of M13 can be engineered for construction of phage particles specific P. aeruginosa strain PAK through interaction with the PAK pili, or PAO1 through interaction with the RP4 pili, respectively.
  • the therapeutic phagemids also contain the Ori1600 and Rep protein necessary for replication and maintenance of the plasmid in P. aeruginosa along with elements necessary for production of the phage particles in E. coli.
  • Expression of luciferase is placed under the control of the E. coli constitutive ⁇ 70 promoter, which along with the promoter driving kanamycin, is active in pseudomonas.
  • the following will detail the construction and characterization of bacteriophage nanoparticle specific for Staphylococcus aureus in vitro and in vivo, as a model Gram positive organism.
  • the therapeutic phage particles 4 are modified for replication in Staphylococcus and chimeric g3p tuned for infection of Staphylococcus aureus.
  • Staphylococcus is chosen as a model Gram positive organism for POC studies due to is wide distribution on the skin, in the nares and upper respiratory tract.
  • the g3p sequences of the therapeutic phagemid are tuned to bind Staphylococcus aureus using phage display.
  • Sequences for phage display screening are based on phage tail fiber regions of published Staphylococcus phages and Staphylococcus outer membrane binding domains of lysin molecules. Once identified, chimeric g3p sequences are subcloned into the therapeutic phage particles 4 containing the Staphylococcus replication elements and a Staphylococcus constitutive promoter driving the transcription of codon optimized luciferase.
  • the therapeutic phage particles 4 can be amplified and BNPs are produced in SA80B E. coli cells (Lucigen) to circumvent the restriction properties of shuttle plasmids between E. coli and Staphylococcus.
  • the therapeutic phage particles 4 can be used to develop diagnostics kits for the detection of microbiome associated diseases.
  • phages can detect changes in quality and number of specific bacteria associated with the microbiome alteration during diseases.
  • phage diagnostic kits may use body fluids as well as tissues.
  • the diagnostic function is achieved by using phages that carry genetic information encoding proteins suitable for imaging such as, for example, fluorescent proteins.
  • the therapeutic phage particles 4 can be directly used in vivo for imaging purposes.
  • Such phages are administered in vivo via oral, topical, aerosol, parental and other appropiate ways. The expression on imaging protein from the bacteria targeted by such phages will allow in vivo imaging.
  • the therapeutic phage particles 4 can also be used for in vivo for delivery of nucleic acids encoding immunoregulatory proteins. In this regard, P. aeruginosa is a significant opportunistic pathogen.
  • CF cytstic fibrosis
  • CF patients whose abnormal airway epithelia allow long-term bacterial colonization of the lungs.
  • agents to suppress inflammation such as systemic corticosteroids, however with significant adverse consequences of such therapy.
  • Interleukin (IL)-10 is an important immunoregulatory cytokine whose expression is diminished in CF [47].
  • IL-10 limits and terminates inflammatory responses and regulates the differentiation and proliferation of several immune cells such as T cells, B cells, natural killer cells, antigen-presenting cells, mast cells, and granulocytes.
  • IL-10 has been shown to mediate immunostimulatory properties that help to eliminate infectious and noninfectious particles with limited inflammation.
  • IL-10/IL-10 receptor system is now seen as a new therapeutic target and recombinant human IL-10 is currently being tested in clinical trials for many indications such rheumatoid arthritis, inflammatory bowel disease, psoriasis, organ transplantation, and chronic hepatitis C.
  • Local delivery to the site of inflammation has advantages over systemic targeting of this pathway. Therefore, a method for in vivo delivery of nucleic acids for site-specific expression of IL-10 would have broad range therapeutic benefit.
  • the therapeutic phage particles 4 e.g., pf3 pseudomonas phagemid
  • the therapeutic phage particles 4 can also be used for in vivo for delivery of of RNA-based nucleic acid therapies.
  • BNPs can be developed for the delivery and expression of genes encoding siRNA to regulate bacterial gene expression in Pseudomonas and to program E. coli for the delivery of shRNA to eukaryotic cells for trans-kingdom RNAi.
  • the use of regulatory RNA has received great attention as a as a novel treatment of many diseases failing conventional small molecule therapy.
  • the use of therapeutic ribozymes, apatamers, and small interfering RNA (siRNA) in post-transcriptional gene silencing (PTGS) has demonstrated the broad potential and utility of RNA-based nucleic acid therapeutics in recent clinical trials.
  • RNA-based nucleic acid therapies to the microbiome for regulation of bacterial gene expression and the delivery of shRNA to mammalian cells.
  • small RNAs sRNA are known be present in and play a regulatory role in signal transduction and metabolism in bacteria. Interactions between prokaryotic sRNA and its target mRNA is sequence specific, mediated by bacterial chaperones, and usually results in the suppression of targeted gene translation.
  • RNA-based nucleic acid therapies can be tuned to effectively deliver RNA-based nucleic acid therapies to the microbiome for regulation of bacterial gene expression.
  • the therapeutic phage particles 4 can also be used for the delivery of nucleic acids to Porphyromonas gingivalis in the oral cavity.
  • Periodontitis is chronic inflammatory disease with high morbidity in the adult population. It typically leads to the destruction of the tooth-supporting structures such as the gingiva and the underlying alveolar bone, and it has been linked to adverse systemic health, such as atherosclerosis, diabetes, rheumatoid arthritis, and adverse pregnancy outcomes.
  • adverse systemic health such as atherosclerosis, diabetes, rheumatoid arthritis, and adverse pregnancy outcomes.
  • One of the hallmarks of periodontitis is the massive accumulation of neutrophils, thus linking the disease to an imbalance of the immune system.
  • Porphyromonas gingivalis a component of the oral microbiome, has long been associated with human periodontitis and recent studies suggest that P. gingivalis is a keystone organism leading to microbial dysbiosis and a proinflammatory response. Overall, P. gingivalis can impair host defenses in ways that alter the growth and development of the entire microbial community, thereby triggering a destructive change in the normally homeostatic relation with the host. Crosstalk between P. gingivalis with cells of the immune system, such as dendritic cells, can lead to the recruitment of pro-inflammatory T cells. Moreover, P. gingivalis inhibits production of Th1 -recruiting chemokines as well as cell production of interferon IFNy.
  • step 1 phage display using E. coli modified to express P. gingivalis FimA is used to select and amplify g3p sequences for absorption of bacteriophage particles to P. gingivalis.
  • Type I FimA are amplified by PCR from P. gingivalis ATCC 33277 and subdoned into an E.
  • E. coli pET expression vector encoding the transmembrane signal from Pseudomonas aeruginosa EstA (NCBI Accession number AF005091 ) as an anchoring motif for display of recombinant proteins on the surface of E. coli.
  • E. coli BL21 (DE3) cells F-, ompT
  • Phage display vector fADL-1 are modified to encode a Pill protein with a random mutagenized DM in the g3p N2 domain (fADL-1 -mN2).
  • Gene blocks containing mutated DII-N2 sequences are synthesized and placed into the fADL-1 vector.
  • the fADL-1 -mN2 plasmids and a pool of phage expressing the mutated Pill are propagated in electrocompetent F1 - E. coli TG1 DUOs.
  • the phage particles are used to infect the Type I FimA expressing cells, and phage are amplified in and purified from the infected cells.
  • ssDNA is isolated from purified phage and the sequence of g3p N2 determined.
  • step 2 the identified g3p N2 sequences conferring absorption to P. gingivalis Type I FimA are subdoned into a phagemid shuttle vector capable of replication in both E. coli (for amplification of plasmid DNA and production of BNPs) and P. gingivalis.
  • the shuttle phagemid in addition to sequences for propagation on E. coli, contain the minimum origin of replication for P. gingivalis and the erythromycin resistance cassette from plasmid pTO-1 , a luciferase reporter gene under the transcriptional control of a P.
  • E. coli TG1 DUO cells are co-transformed with the shuttle phagemid and M13 packaging vector (containing a chloramphenicol [cat] resistance cassette) and transformed TG1 DUOs selected and propagated in 2XTY plus kanamycin and chloramphenicol. Phage particles are isolated and concentrated from the culture medium by PEG precipitation. Infected P.
  • gingivalis are plated onto TSA blood agar plus containing erythromycin and plasmids purified from single colony isolates will sequenced for determination of g3p sequences conferring infection of P. gingivalis ATCC 33277.
  • the identified g3p sequences are subcloned into the shuttle phagemid encoding a codon optimized nanoluciferase (nLuc) reporter gene [REF] and BNPs programmed with a luciferase reporter gene are constructed in E. coli and characterized for delivery of nucleic acids encoding peptide therapeutics to P. gingivalis in vitro and in vivo.
  • nLuc nanoluciferase
  • the BNPs generally are specific for Type I FimA P. gingivalis strains due to the antigenic differences of the FimA proteins.
  • the range of the BNPs can be expanded, or tuned to specific FimA proteins, using the E. coli FimA expression plasmid to encode alternate FimA proteins.
  • nucleic acid therapies to P. gingivalis can be delivered for the effective local expression of immunoregulatory proteins and a reduction in the inflammatory responses associated with periodontal disease.
  • the BNPs can be for delivery of IL-10 to P. gingivalis.
  • the therapeutic phage particles 4 encode a codon optimized IL-10 containing signal sequences for POR secretion system of P. gingivalis.
  • IL-10 is an immunoregulatory cytokine that limits and terminates inflammatory responses, including the expression of I L-1 ⁇ and TNFa, and regulates the differentiation and proliferation of several immune cells to mediate immunostimulatory properties that help to eliminate infectious and noninfectious particles.
  • gingivalis uses a channel complex to secrete substances containing C-terminal peptide signals in from the cytoplasm across the inner and outer membranes to the outer bacterial surface and into the extracellular space.
  • Phagemids are modified to express codon optimized rlL-10 or with a C-terminal POR secretion signal (CTD).
  • CTD C-terminal POR secretion signal
  • Phagemids expression nLuc with the CTD POR secretion signal are engineered for use as a control.
  • nLuc is an ATP- independent bioluminescent enzyme.
  • the FimA expression plasmid can encode sequences for FimA Types ll-V and used for selection of BNP particles to transduce additional P. gingivalis strains.
  • the expression plasmid can express a FimA consensus sequence and peptides encoding homologous regions between the FimA proteins to generate a ubiquitous BNPs for transduction of P. gingivalis.
  • nucleic acid sequences encoding sRNA can be expressed.
  • P. gingivalis harbors an arsenal of virulence factors, which along with its many interactions with the host immune system strongly support its potency as a pathogen.
  • P. gingivalis also expresses a wide variety of sRNA in response to different environmental stimuli.
  • the BNPs for the expression of sRNA can be used to regulate virulence factors.
  • references to singular components or items are intended, unless otherwise specified, to encompass two or more such components or items. Also, to the extent that the terms “including”, “includes”, “having”, “has”, “with”, or variants thereof are used in the detailed description and/or in the claims, such terms are intended to be inclusive in a manner similar to the term “comprising”.

Abstract

L'invention concerne des compositions pour une particule de phage. La particule de phage est non réplicante et comprend au moins une séquence d'acides nucléiques hétérologue qui peut être exprimée dans une bactérie cible. La séquence d'acides nucléiques hétérologue exprimée est non létale pour les bactéries cibles.
PCT/US2015/012894 2014-01-29 2015-01-26 Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques WO2015116531A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2016548341A JP6755801B2 (ja) 2014-01-29 2015-01-26 治療的な使用のための、核酸送達のための治療用ファージおよび方法
US15/114,634 US10351452B2 (en) 2014-01-29 2015-01-26 Compositions for in vivo expression of therapeutic sequences in the microbiome
EP15742945.7A EP3099173A4 (fr) 2014-01-29 2015-01-26 Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques
US18/198,519 US20230285508A1 (en) 2014-12-05 2023-05-17 Treatment delivery system and method

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201461933032P 2014-01-29 2014-01-29
US61/933,032 2014-01-29
US201462063031P 2014-10-13 2014-10-13
US62/063,031 2014-10-13
US201462088073P 2014-12-05 2014-12-05
US62/088,073 2014-12-05
US14/605,320 US9676641B2 (en) 2014-01-29 2015-01-26 Therapeutic phages and methods for delivery of nucleic acids for therapeutic uses
US14/605,320 2015-01-26

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/114,634 A-371-Of-International US10351452B2 (en) 2014-01-29 2015-01-26 Compositions for in vivo expression of therapeutic sequences in the microbiome
US16/429,474 Division US20190315642A1 (en) 2014-12-05 2019-06-03 Compositions for in vivo Expression of Therapeutic Sequences in the Microbiome

Publications (1)

Publication Number Publication Date
WO2015116531A1 true WO2015116531A1 (fr) 2015-08-06

Family

ID=53757657

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/012894 WO2015116531A1 (fr) 2014-01-29 2015-01-26 Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques

Country Status (1)

Country Link
WO (1) WO2015116531A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019500049A (ja) * 2016-01-03 2019-01-10 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 免疫原性組成物
JP2019512227A (ja) * 2016-03-07 2019-05-16 中国科学院広州生物医薬与健康研究院Guangzhou Institutes Of Biomedicine And Health,Chinese Academy Of Sciences 自己発光部材を送達可能なマイコバクテリウムファージ及びその使用

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030148501A1 (en) * 1999-03-31 2003-08-07 Picardal Flynn W. Compositions and methods useful in bioremediation of polychlorinated biphenyls
US20050107326A1 (en) * 1999-04-14 2005-05-19 The Penn State Research Foundation, a Commonwealth of Pennsylvania Tissue-specific and pathogen-specific toxic agents and ribozymes
US20090148408A1 (en) * 2002-03-08 2009-06-11 Osel, Inc. Lactobacilli expressing biologically active polypeptides and uses thereof
US20100047885A1 (en) * 2006-09-22 2010-02-25 Rice University Pantothenate Kinase Overexpression and Pantothenic Acid Supplementation in Actinomycetes
US20100226890A1 (en) * 2005-07-12 2010-09-09 The University Of Leeds Bacteriophage and their uses
US20100239546A1 (en) * 2007-06-15 2010-09-23 Beth Israel Deaconess Medical Center Bacterial mediated tnf alpha gene silencing
US20100240025A1 (en) * 2007-06-15 2010-09-23 Timothy Read Bacteriophage with Enhanced Lytic Activity
US20120070414A1 (en) * 2000-10-04 2012-03-22 Paul Stamets Controlling disease vectors from insects and arthropods using preconidial mycelium and extracts of preconidial mycelium from entomopathogenic fungi

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030148501A1 (en) * 1999-03-31 2003-08-07 Picardal Flynn W. Compositions and methods useful in bioremediation of polychlorinated biphenyls
US20050107326A1 (en) * 1999-04-14 2005-05-19 The Penn State Research Foundation, a Commonwealth of Pennsylvania Tissue-specific and pathogen-specific toxic agents and ribozymes
US20120070414A1 (en) * 2000-10-04 2012-03-22 Paul Stamets Controlling disease vectors from insects and arthropods using preconidial mycelium and extracts of preconidial mycelium from entomopathogenic fungi
US20090148408A1 (en) * 2002-03-08 2009-06-11 Osel, Inc. Lactobacilli expressing biologically active polypeptides and uses thereof
US20100226890A1 (en) * 2005-07-12 2010-09-09 The University Of Leeds Bacteriophage and their uses
US20100047885A1 (en) * 2006-09-22 2010-02-25 Rice University Pantothenate Kinase Overexpression and Pantothenic Acid Supplementation in Actinomycetes
US20100239546A1 (en) * 2007-06-15 2010-09-23 Beth Israel Deaconess Medical Center Bacterial mediated tnf alpha gene silencing
US20100240025A1 (en) * 2007-06-15 2010-09-23 Timothy Read Bacteriophage with Enhanced Lytic Activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3099173A4 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2019500049A (ja) * 2016-01-03 2019-01-10 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 免疫原性組成物
JP2021112202A (ja) * 2016-01-03 2021-08-05 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 免疫原性組成物
JP2019512227A (ja) * 2016-03-07 2019-05-16 中国科学院広州生物医薬与健康研究院Guangzhou Institutes Of Biomedicine And Health,Chinese Academy Of Sciences 自己発光部材を送達可能なマイコバクテリウムファージ及びその使用

Similar Documents

Publication Publication Date Title
US20190315642A1 (en) Compositions for in vivo Expression of Therapeutic Sequences in the Microbiome
US10351452B2 (en) Compositions for in vivo expression of therapeutic sequences in the microbiome
Furuyama et al. Outer membrane vesicles (OMVs) produced by gram-negative bacteria: structure, functions, biogenesis, and vaccine application
CN109152848B (zh) 抗-crispr化合物以及使用方法
Secor et al. Pf bacteriophage and their impact on Pseudomonas virulence, mammalian immunity, and chronic infections
JP7275043B2 (ja) 増大したhATファミリートランスポゾン媒介遺伝子導入ならびに関連する組成物、システムおよび方法
US11827890B1 (en) Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
CN109996880A (zh) 基于模块化aav递送系统的crispr-cas基因组工程
CN102131927A (zh) 可调型基因自杀机制组合物和方法
Brüggemann et al. Bacteriophages infecting Propionibacterium acnes
TW200408410A (en) Clinical grade vectors based on natural microflora for use in delivering therapeutic compositions
CN108472391A (zh) 免疫原性组合物
JP2019010125A (ja) 細菌中の接合性プラスミドの低減方法
WO2015116531A1 (fr) Phages thérapeutiques et méthodes d'administration d'acides nucléiques pour applications thérapeutiques
ES2240758T3 (es) Metodo de estibilizacion de plasmidos mediante delecion in vivo de la resistencia a antibioticos y seleccion con un gen esencial.
US20230285508A1 (en) Treatment delivery system and method
ES2686341T3 (es) Nuevo método de preparación de antibióticos y sistema de plataforma basado en el mismo
KR20140002463A (ko) 디포신 및 그의 사용 방법
ES2561667T3 (es) Genes sintéticos que codifican fragmentos de péptido de proteínas naturales de mielina para la inducción de la tolerancia oral, fragmento de ADN que comprende estos genes, medios para obtener estos péptidos en un sistema microbiano (bacteriano) y su aplicación médica
CN116669557A (zh) 微生物群落中微生物的控制
CN116685204A (zh) 调节抗微生物肽半衰期
JP2004065237A (ja) 遺伝子導入用担体、前記担体を用いた遺伝子導入剤、前記導入剤を用いた遺伝子導入方法、およびそれによって遺伝子を導入した細胞
Fermin et al. Viruses as Tools of Biotechnology: Therapeutic Agents, Carriers of Therapeutic Agents and Genes, Nanomaterials, and More
KR102555172B1 (ko) Gm-csf의 분비를 위한 유전자 구조물 및 이에 의해 형질전환된 항암 재조합 균주
RU2745626C1 (ru) Способ создания живой вакцины против коронавирусной инфекции COVID-19 на основе пробиотического штамма Enterococcus faecium L3 и живая вакцина Enterococcus faecium L3-pentF-covid-19

Legal Events

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

Ref document number: 15742945

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2016548341

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15114634

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

REEP Request for entry into the european phase

Ref document number: 2015742945

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015742945

Country of ref document: EP