NZ764519B2 - Coated adenoviruses for immunotherapy - Google Patents

Coated adenoviruses for immunotherapy Download PDF

Info

Publication number
NZ764519B2
NZ764519B2 NZ764519A NZ76451915A NZ764519B2 NZ 764519 B2 NZ764519 B2 NZ 764519B2 NZ 764519 A NZ764519 A NZ 764519A NZ 76451915 A NZ76451915 A NZ 76451915A NZ 764519 B2 NZ764519 B2 NZ 764519B2
Authority
NZ
New Zealand
Prior art keywords
polypeptides
tumor
adenoviral vector
specific
cells
Prior art date
Application number
NZ764519A
Other versions
NZ764519A (en
Inventor
Cristian Capasso
Vincenzo Cerullo
Mari Hirvinen
Koskela Markus Vaha
Original Assignee
Valo Therapeutics Oy
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 Valo Therapeutics Oy filed Critical Valo Therapeutics Oy
Priority claimed from NZ726112A external-priority patent/NZ726112A/en
Publication of NZ764519A publication Critical patent/NZ764519A/en
Publication of NZ764519B2 publication Critical patent/NZ764519B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/522Bacterial cells; Fungal cells; Protozoal cells avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6075Viral proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/235Adenoviridae
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/46Ingredients of undetermined constitution or reaction products thereof, e.g. skin, bone, milk, cotton fibre, eggshell, oxgall or plant extracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10042Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Abstract

The present invention relates to adenoviral vectors, wherein the viral capsid has been coated with polypeptides, which are capable of stimulating a peptide-specific immune response in a subject and uses thereof. Furthermore, the present invention relates to methods of treating diseases, e.g. viral infections, by adenoviral vectors which have been coated by polypeptides causing peptide-specific immune response. Also the present invention relates to a method of coating adenoviral vectors by specific peptides as well as to a method of identifying those peptides suitable for coating the capsid of an adenoviral vector. nfections, by adenoviral vectors which have been coated by polypeptides causing peptide-specific immune response. Also the present invention relates to a method of coating adenoviral vectors by specific peptides as well as to a method of identifying those peptides suitable for coating the capsid of an adenoviral vector.

Description

(12) Granted patent specificaon (19) NZ (11) 764519 (13) B2 (47) aon date: 2021.12.24 (54) COATED ADENOVIRUSES FOR IMMUNOTHERAPY (51) Internaonal Patent Classificaon(s): A61K 39/00 (22) Filing date: (73) Owner(s): 2015.05.18 Valo Therapeutics Oy (23) Complete specificaon filing date: (74) Contact: 2015.05.18 Adams Pluck (62) Divided out of 726112 (72) Inventor(s): CERULLO, Vincenzo (30) Internaonal Priority Data: VÄHÄ-KOSKELA, Markus FI 20145449 2014.05.19 HIRVINEN, Mari CAPASSO, Cristian (57) Abstract: The present invenon relates to adenoviral vectors, wherein the viral capsid has been coated with polypepdes, which are capable of smulang a pepde-specific immune response in a subject and uses thereof. Furthermore, the present invenon relates to methods of g diseases, e.g. viral infecons, by adenoviral s which have been coated by pdes causing specific immune response. Also the present invenon relates to a method of coang adenoviral vectors by specific pepdes as well as to a method of idenfying those pepdes suitable for coang the capsid of an iral vector.
NZ 764519 B2 COMPLETE ICATION APPLICANT VALO THERAPEUTICS OY TITLE COATED ADENOVIRUSES FOR IMMUNOTHERAPY COATED ADENOVIRUSES FOR IMMUNOTHERAPY FIELD OF THE INVENTION The present ion relates to adenoviral vectors, wherein the viral capsid has been coated with ptides, which are capable of stimulating a peptide-specific immune response in a subject and uses thereof. Furthermore, the t disclosure relates to methods of treating diseases, e.g. cancer, by adenoviral s which have been coated by polypeptides causing especific immune response. Also the present disclosure relates to a method of coating adenoviral vectors by specific peptides as well as to a method of identifying those peptides suitable for coating the capsid of an adenoviral vector.
BACKGROUND OF THE INVENTION Cancer is a lethal disease in need of more effective treatments. tic viruses are of significant interest since they have the potential to be safer and more effective than any other standard therapy. However, in cancer patients the overall therapeutic effect has been modest. There are many s on modifying the iral vectors in order to find optimal tools for therapies. One aspect of regulating the function of adenoviruses is to modify the surface of the virus. Both genetic as well as non-genetic modifications of surfaces of adenoviruses are well known.
For example Stevenson M et al. (Cancer Gene Therapy (2007) 14, 335– 345) concentrate on enhancing the delivery of adenoviral vectors to target sites.
Stevenson et al. describe a study wherein adenoviral vectors are ed to infect cells via integrins that are selectively expressed on metastatic tumor cells. For this purpose a n-derived peptide (-SIKVAV-) was incorporated onto the surface of the polymer-coated viruses.
WO2013/116778 describes an immunologically enhanced adenovirus for . An adenovirus was modified by inserting a tumor antigen transgene into its genome in a way that the tumor antigen is expressed during the virus’s replication cycle and presented directly to MHC-I. This method is very slow and too laborious and expensive for personalized therapies, because the generation of a new virus is needed for every different tumor n (e.g. one must clone a new virus for every peptide that is wanted to be expressed).
Followed by page 2 Indeed, a need exists for simple and improved adenoviral tools and methods for therapeutics, especially for personalized ies. The present disclosure provides adenoviral applications for ing the immune response in a subject while using the virus as delivery system of peptides but not involving genetic manipulation of the virus.
The present disclosure further relates to the use of oncolytic adenoviruses as rm to r patient- and disease-specific peptides and consequently convert the anti-capsid immunity into a peptide specific immune response (e.g. anti-tumor ty).
BRIEF DESCRIPTION OF THE ION The present sure provides a new and potent customizable immunovirotherapy (e.g. cancer immunovirotherapy) platform. It is an advantage that in one or more forms there may be provided an adenoviral vector with a modified viral surface, uses thereof and a method for treating a disease by stimulating a peptide-specific (i.e. anti-peptide) immune response to ameliorate problems of e.g. inefficient, slow, expensive and laborious adenoviral therapies as well as the ability of the adenoviral therapies for personalized medicine, or to at least provide a useful alternative. The t matter of the invention is characterized by what is stated in the independent claims. The preferred embodiments of the invention are sed in the dependent claims.
By the present disclosure, one or more problems of prior art e.g. lack of specificity and the immune nce of oncolytic adenoviruses may be ameliorated.
Immune responses generated by adenovirus infection target mainly the virus and not the tumor. Furthermore, the majority of the viral immunity is directed against the proteins of the capsid. T he present invention in one or more forms is based on the idea that coating the viral capsid with peptides derived from tumor proteins diverts viral immunity to the tumor (Figure 3). In particular, the major histocompatibility x I (MHC-I) restricted peptides mounted onto the oncolytic adenovirus capsid may in one or more forms of the present disclosure, divert the capsid immunity into anti-tumor immunity.
Simply, when peptide(s) and virus(es) are administered as a single physically linked entity, both danger signal (virus) and tumor-antigen de) will enter the same antigen presenting cell for maximal anti-tumor effect. Clinical experience has already indicated that e vaccination alone only leads to a transient and imal immune response incapable of controlling tumor 1.
Correspondingly, while oncolytic viruses show promise as monotherapy, the immune se they elicit is mainly ed against the virus, not the tumor.
Even if peptide and virus are injected in the same anatomical location, since they are not joint in a single therapeutic entity, they inefficiently enter the same cell— aspects which are critical for ing proper and maximal immune activation2.
The physical conjunction of peptide and iral virus in a single therapeutic entity is a significant improvement over existing virus and peptide cancer vaccine technologies. In contrast to recombinant viruses of the prior art engineered to s one tumor-associated antigen or peptide, the present invention makes it possible to achieve personalized medicines in a much quicker and more costeffective way than before. Indeed, according to the t invention peptides attached onto a viral capsid are not encoded by the adenoviral vector.
One form of the present disclosure is the logy allowing constant and rapid monitoring of tumor antigen presentation as small peptides (MHC-I restricted). The present disclosure takes advantage of disease - (e.g. tumor-) and patient-specific peptides, which are presented simultaneously on tumor cells both before and after adenoviral therapy (i.e. which are not masked or edited away after therapy) and on dendritic cells (DCs) following adenoviral therapy. After identification of these specific peptides they can be synthetized and mounted onto the tic adenovirus capsid to achieve high anti-tumor immunity. This way it is possible to ensure that the tumor is effectively targeted by cytotoxic T-cells (CTLs) also after erapy so that immunological escape becomes impossible as the immune system targets the virus. Conversely, by comparing peptides appearing on DCs after virus therapy in the presence or absence of tumor, it is le to eliminate “virus-only” peptidesand find those deriving from the tumor cells that induce CTL response.
A personalized coated adenovirus can be obtained in as little as two weeks from biopsy; this is made possible because isolation and sequencing of peptides from MHC’s as well as automated synthesis are rapid processes, and the virus (e.g. the same backbone virus for all peptides) can be stockpiled in large quantities to await coating. g itself is med in one hour, after which the coated adenovirus is ready for injection. This is very unique feature of the present disclosure as it bypasses any genetic manipulation of the virus that slows down the s making the “personalized-vaccine approach” ible.
The present disclosure in one or more forms also provides for the possibility of discovery of novel immunogenic tumor-specific peptides.
In addition to cancer therapy, the coated adenovirus of the t invention may be used for treating any other es in a situation where higher and peptide-specific immune response is needed.
In particular, in one aspect of the invention there is provided an adenoviral vector comprising polypeptides that stimulate a peptide-specific antivirus immune response in a subject, wherein the ptides are attached onto the viral capsid and have not been genetically encoded by said adenoviral vector and further wherein the polypeptides attached onto the viral capsid are selected from the group ting of Major Histocompatibility Complex of class I (MHC-I)- specific polypeptides and Major Histocompatibility x of class II (MHC-II)- specific polypeptides, and are disease specific polypeptides.
In another aspect of the invention there is provided the use of an adenoviral vector embodied by the invention in the manufacture of a medicament to treat an infection.
In a related embodiment there is provided an adenoviral vector comprising polypeptides attached onto the viral capsid for use in stimulating a peptide-specific immune response in a subject. In another related embodiment there is provided an adenoviral vector comprising polypeptides attached onto the viral capsid for use in stimulating a peptide-specific immune se in a subject, wherein the polypeptides have not been genetically encoded by said adenoviral vector.
In a related embodiment there is provided a method of ating a peptide-specific immune response in a t in need thereof, wherein the method comprises administration of adenoviral vectors comprising polypeptides ed onto the viral capsid to the subject. In r related embodiment there is provided a method of stimulating a peptide-specific immune response in a subject in need thereof, wherein the method comprises administration of adenoviral vectors comprising polypeptides attached onto the viral capsid to the subject, wherein the polypeptides have not been genetically encoded by said adenoviral vector.
In another related embodiment there is provided a method of treating cancer in a subject in need thereof, wherein the method comprises administration of adenoviral vectors comprising polypeptides, which are capable of stimulating a peptide-specific immune response in the subject and which have been attached onto the viral capsid, to the subject. In another related embodiment there is provided a method of treating cancer in a subject in need thereof, wherein the method comprises administration of iral vectors comprising ptides, which are capable of stimulating a peptide-specific immune response in the subject and have been attached onto the viral capsid, to the t, wherein the ptides have not been genetically encoded by said adenoviral vector.
Also, in r d embodiment the present sure s to an adenoviral vector comprising polypeptides, which are capable of stimulating a peptide-specific immune response in a subject and which have been attached onto the viral capsid, for use in treating cancer in a t. The t invention also relates to an adenoviral vector comprising polypeptides, which are capable of stimulating a peptide-specific immune response in a subject and which have been attached onto the viral capsid, for use in ng cancer in a subject, wherein the polypeptides have not been genetically encoded by said adenoviral vector.
Furthermore, the t invention relates to an adenoviral vector, wherein the viral capsid has been attached with polypeptides and wherein the adenoviral vector attached with polypeptides is capable of stimulating a peptide- specific immune response in a subject.
Furthermore, the present invention relates to a method of coating the capsid of an adenovirus, wherein said method comprises linking polypeptides, which are capable of stimulating a peptide-specific immune response in a subject, to the iral capsid covalently or non-covalently. The present invention also relates to a method of modifying the capsid of an adenovirus, wherein said method comprises linking of polylysine-modified polypeptides to the adenoviral capsid covalently or non-covalently, wherein the modified adenoviral vector is capable of stimulating a peptide-specific immune response in a subject.
Still, the present invention relates to use of ptides (e.g. polylysine-modified polypeptides), which are capable of stimulating a peptidespecific immune response in a subject, for coating the capsid of an adenovirus by covalently or valently attaching or linking the polypeptides to the capsid.
The adenoviral vector and methods as described herein may be used for converting antiviral immunity into anti-peptide immunity wherein the modified viral vector of the invention causes anti-peptide response in a t.
Still further, the present invention relates to a pharmaceutical ition comprising an adenoviral vector embodied by the invention.
And still, in another related ment there is provided a method for identifying tumor-specific and MHC-I-specific polypeptides from a subject, said method comprising i) infecting tumor cells of the subject with adenoviral vectors; ii) infecting dendritic cells of the subject with iral vectors; iii) isolating MHC-I molecules from tumor cells of step i) and from dendritic cells of step ii) and identifying the MHC-I-associated polypeptides from both groups; iv) isolating MHC-I molecules from uninfected tumor cells and identifying the associated polypeptides; v) identifying those polypeptides which have been presented by the infected and uninfected tumors of steps iii) and iv) and dendritic cells of step iii).
Throughout this specification, the word “comprise”, or variations thereof such as ises” or “comprising”, will be understood to imply the inclusion of a stated element, integer or step, or group of elements integers or steps, but not the exclusion of any other element, integer or step, or group of elements, rs or steps.
Any discussion of nts, acts, materials, devices, articles or the like which has been included in this specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the t ion as it existed in New Zealand or elsewhere before the priority date of this application.
BRIEF DESCRIPTION OF THE DRAWING In the following the invention will be described in r detail by means of preferred embodiments with reference to the attached drawings, in which Figure 1 shows a tic of the present invention, wherein the modified adenovirus is capable of replicating and killing cancer cells while diverting the anti-viral immune response against the tumor.
Figure 2 shows dominance of anti-adenovirus response (left bar) vs tumor se (right bar). Mice) C57BL/6 mice bearing B16-OVA tumor were treated with PBS (Mock), Ad5D24 (unmodified oncolytic virus) and (Ad5D24-CpG, a more immunogenic tic virus). T cells from the tumor were harvested and IFNgamma ELISPOT was performed to assess anti-tumor response and anti-adenovirus response. Cancer Patients) IFNgamma ELISPOT was performed on PBMCs from patients treated with an GMCSF-armed oncolytic adenovirus (Ad5D24-GMCSF)15. Ad5 -derived peptides (anti-viral) and survivin- derived peptides (anti-tumor) were used to stimulate PBMCs before the ELISPOT.
Figure 3 reveals that the coated adenoviruses of the present invention represent an advantage vs existing technology. A) Oncolytic adenovirus has the ability to trigger APCs to present not only viral antigens (that leads to ral se) (another antigen presented on the cell of the Figure A) but also, as side effect, tumor antigens er n presented on the cell of the Figure A) that leads to mor immunity. Anti-tumor T cells are marked as the two lowest cells of the T cell group. B) The coated adenoviruses of the present invention will favor tumor antigen tation (marked as both antigens presented on the cell of the Figure B) because its capsid is covered by MHC-I ready-to-use tumor-specific antigens (peptides). In this way the anti-capsid immunity can be reverted into anti-tumor immunity. Anti-tumor T cells are marked as the four lowest cells of the T cell group. As Followed by page 7 used herein APC refers to antigen presenting cells, TAA refers to tumor associated antigen and “PRR tion" refers to pattern recognition receptor activation. PRRs are proteins expressed by cells of the innate immune system to identify pathogen~associated molecular patterns, which are associated for example with microbial pathogens.
Figure 4 shows the top upregulated Bio-Function networks of dendritic cells exposed to oncolytic irus. Human primary dendritic cells were harvested and cultured for two weeks with IL4 and GMCSF. The cells were pulsed with an oncolytic adenovirus (Ad5D24) at 10VP/cell. 72h later total RNA was collected, and analyzed on Agilent SurePrint G3 human 8x60k (mRNA). Data were then analyzed with Ingenuity Pathway software.
Figure 5 shows a tic representing the discovery of novel immunogenic tumor-associated MHCI restricted es. ent conditions allow us to match the peptides, which the tumor is expressing, with the e of the same tumor that tic cells are presenting. This is a key feature in the system to facilitate the identification of immunogenic peptides. A) Dendritic cells were pulsed with tumor sate to allow tumor ns presentation. 8) Unpulsed dendritic cells were matured and analyzed. This serves as a control to subsequently eliminate the self—peptides presented by the DCs. C) Infected tumor cell line (the same as ion A) were infected with tic adenovirus and analyzed before complete lysis (less than 48h). This condition helps us to discriminate if the adenovirus has a significant impact on the quality of the tumor antigens presented. D) This is uninfected tumor which presents tumOr antigens and self-peptides (Oic course these two can be the same) on MHCI.
Figure 6 shows a schematic of OVA-specific coated viruses. A) in this case, as we know all the processed peptide of the chicken ovalbumin (OVA) we coated the virus with OVA Specific immunogenic peptide (SllNFEKL) (SEQ ID NO: 1). Then we generated other coated viruses to be used as controls such as SllNFDL (SEQ lD NO: 2) (antagonist) and FILKSINE (SEQ lD NO: 3) (scramble) as well as uncoated viruses. B) Once the proof-of-concept had been proved we started with the study of II generation adenoviruses that are coated with different peptides. (PeptiCRAd refers to an oncolytic adenovirus coated with peptides.) Figure 7 shows a schematic representing three ent strategies to te the peptide—coated oncolytic adenovirus.
Figures 8 show complex formation between oncolytic adenoviruses and tumor specific peptides and interaction between modified epitopes and oncolytic adenoviruses. Figure 8A shows a complex formation between Ad5D24 tic adenovirus and tumor-specific peptides. “Z—potential” line) 1X101O viral particles were conjugated with different concentration of positively charged specific peptide. After the reaction Z-potential of the single particles was ed. “Size” line) 1X1010 virai particles were conjugated with different concentration of positively charged tumor—specific peptide. After, the size of the single les was measured and ed in on of the peptide concentration. When the Z—potential is n -20mV and +20mv there is a drastic change in size of the complex showing high degree of poly- dispersity (likely virus ation), but this state returned to normality at higher concentration of peptides suggesting that the complex (PeptiCRAd) is completed coated with no possibility to form dipole that promotes the formation of the aggregates (high polydispersity). Figure 8B reveals the interaction between the modified MHC-l epitope SIINFEKL and oncolytic adenoviruses.
The virus/peptide interaction was measured by SPR. An APTES silica Si02 sensor was coated with Ad5D24, and increasing concentrations (0.15, 0.3, 0.6, 1.2, 2.4 and 7.2 uM) of either SIINFEKL (dashed line) or ponK-SllNFEKL (solid line) were injected into the flowing system. The SPR signal se is shown in relation to the duration of the experiment.
Figure 9 shows that the coated irus Ad5D24 of the present invention (PeptiCRAd) displays an enhanced cell killing activity compared to uncoated oncolytic virus. Representative cell viability assay (MTS assay) performed on lung cancer arcinoma cell line (A549). Cells were seeded on day 0, infected at indicated multiplicity of infection on day 1 and the test was stopped and analyzed on day 3.
Figures 10 show that OVA—specific adenovirus enhances the OVA- specific ty. Mice bearing subcutaneous BtB-OVA tumors were intratumorally injected with: PBS, Oncolytic virus (Ad5D24), Oncolytic virus + SIINFEKL peptides (Not xed), Oncolytic virus + SIINFEKL (Complexed as single entity, PeptiCRAd). A) Tumor growth was measured and ed at shown time points. B) SllNFEKL specific immunity was assessed by flow try (pentamer analysis).
Figure 11 shows the consistency of the peptide coating technique.
The figure shows the net charge of two different oncolytic adenoviruses coated with ed peptide (BK-SIINFEKL). The two viruses used in this example are Ad5D24—CpG (oncolytic adenovurs genetically modified to have its genome rich in CpG islands) and Ad5D24-RFP which is an oncolytic adenovirus encoding for the Red fluorescent protein for tating the ing in vitro and in vivo; (RFP refers to Red Fluorescent Protein).
Figure 12 shows the correlation between net charge of PeptiCRAd and its size. in this example we started with a naked virus (net charge about - —30 mV) and then adding increasing concentration of peptides to form the complex we call RAd. it shows that the more peptides we added the more the net charge of the virus changed from negative to positive values, at the end, when the x PeptiCRAd was formed the net charge of the virus coated with the e was about +30—35mv.
Figures 13 show cross-presentation of modified SIINFEKL analogs on MHC-l ed or not adsorbed onto the viral capsid. Spleens were collected from C57BL/6 mice (H-ZKb), and a single~cell suspension was prepared in RPMI-1640 growth media with 10% FBS. (A) A total of 2x106 splenocytes were ted with 200 pl of media containing unmodified SIINFEKL ive control), the amino caproic acid-containing SllNFEKL-AHX- polyK ive control), the C-terminus-extended SIINFEKL-polyK or the N- terminus-extended polyK-SllNFEKL (0.19 ug/ul). After 2 h of incubation at 37°C, the cells were washed and stained with APC anti-H-2Kb bound to SIINFEKL or isotype control. (B) Similar to (A), fresh murine splenocytes were infected with of OVA-PeptiCRAd (100 l + 37.5 ug of peptide) and 37.5 ug of SIINFEKL (positive control) or polyK—SIINFEKL. After 2 h of incubation, the samples were washed and analyzed by flow cytometry. The data are shown as the mean i SEM (n22). Significance was assessed using one-way ANOVA with Bonferroni‘s multiple comparison test; * P<0.05, ** P<0.01, *** 1.
Figures 14 show that PeptiCRAd retains intact oncolytic activity and diSplays increased infectivity in cell lines with low CAR expression. (A) Cells were seeded at a density of 1X104 cells per well and infected with OVA- PeptiCRAd or naked Ad5D24 using different volcell ratios (0.1, 1, 10 and 100).
The peptide polyK-SIINFEKL (dashed line, circles) was included as a control.
The cell viability was then determined by MTS assay. The data are shown as the mean i SEM (n=3). (3) Study of viral infectivity by ICC. A total of 2><1O5 cells per well were seeded in a 24-well plate and infected with 100 pl of viral dilution (10 vp/cell) containing either OVA-PeptiCRAd or Ad5D24 (control) on the following day. After two days of incubation, anti-hexon ICC was performed, and five non-overlapping images were acquired using a digital microscope.
The average number of spots per visuai field is presented. The data from a representative experiment are shown as the mean i SEM (n=2-3).
Significance was assessed using the unpaired t—test with s correction; * P<0.05, ** P<0.01, *** P<0.001.
Figures 15 show anti~tumor efficacy of PeptiCRAd and immunological is of antigen-specific CD8+ T cells and 00s. (A) C57BL/6 mice (n=6) received 3><105 BiG-OVA cells in both flanks. ent was ted 9 days later and included saline solution (mock), peptide alone (SllNFEKL), virus alone (Ad5D24-CpG), a mixture of virus and peptide (Ad5D24-CpG+SlINFEKL) and virus-peptide complex (OVA-PeptiCRAd). The mice were d three times (on days 0, 2 and 7). Tumor size was then measured and is presented as the mean :1: SEM as a function of time.
Statistical analysis was performed using two-way ANOVA with Bonferroni’s multiple comparison test. * P<0.05, ** P<0.01, *** P<0.00l. Tumors, spleens and inguinal lymph nodes were collected from mice (n=3-4) at two time : the 7th day ) (B) and the 16th day (late) (C). The proportion of SIINFEKL— specific CD8+ T cells was then determined by gating out (3019‘ cells. The percentage of CDS+OVA+ T cells is presented as the mean 1: SEM. (D) The average tumor size at the end of the experiment r y axis) was plotted against the average percentage of double-positive CD8*OVA+ T cells (logm x axis). The Pearson’s rand rzvalues were also calculated and graphed for each set of samples. (E) The fold change in DCs showing a mature profile and cross-presenting SliNFEKL on their MHC-l molecules was determined. Mature DCs were defined as CDlQ‘CD3‘CD11 atopsshlgl‘ cells. APC anti-mouse H—2kb bound to KL was used to track the cross-presentation of SllNFEKL on MHC-I in the selected pool of DCs.
Figures 16 show that targeting two tumor antigens with PeptiCRAd reduces the growth of both d and distant, untreated tumors. One y tumor was engratted in CS7BL/6 mice on the right flank using 1x105 BiG—Flo melanoma cells. Treatment started at day 10. At day 16, the mice received 3><105 Bl6—F10 cells on their left flank. (A) The growth of the primary (right) tumor is reported, and the data are presented as the mean t SEM (n=5).
Significance was determined using two-way ANOVA with roni’s multiple comparison test; * P<0.05, ** P<0.01, *** P<0.001. (B) The size of the secondary (left) tumors at the end of the experiment is reported on a log2 scale.
Significance was assessed using the Mann-Whitney U-test; * P<0.05, ** P<0.01, *** P<0.001. (C) Spleens and inguinal lymph nodes were harvested, and the level of TRP and hgp100-specific CD8+ T cells was determined in each organ by MHC-I pentamer staining. The percentage of e-specific CD8+ T cells found in each organ was normalized against mock and is presented as the cumulative relative response for each mental group.
Figures 17 show efficacy of PeptiCRAd in humanized mice bearing human melanomas. Triple-knockout NGS mice received 2×106 human melanoma cells L-2) on each flank. When the tumors reached an e er of 4-5 mm, a group of mice (n=3) received human PBMCs from an HLA-A-matched healthy donor, whereas another group of mice (n=2) did not receive PBMCs. The mice were then treated (at days 0, 2 and 4) with one of the following: i) saline solution (mock), ii) -GM-CSF, and iii) MAGE-A1 PeptiCRAd. The tumor volume of the humanized mice (A) is presented as the mean ± SEM. icance was assessed using two- P<0.05, ** P<0.01, *** P<0.001, **** P<0.0001. (B) For each group of humanized mice, the area under the curve (AUC) relative to the size of the tumor is presented.
(C) The tumor volume of non-humanized mice is reported as the mean ± SEM (**** P<0.0001).
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS OF THE INVENTION Tumor immunology and the immunopeptidome Dendritic cells (DC) are bone marrow derived professional antigen presenting cells. DCs are optimal antigen presenting cells for presenting tumor antigen epitopes to CD8+ and CD4+ T cells3. Exogenous antigens can be loaded - T cell4. Cross-presentation is a phenomenon whose outcome is ined by the tion status of the DCs5. In cancer cells, the extent of DC maturation that leads to tumor-antigen crosspresentation is usually very low due to the hostile tumor microenvironment and tumor-derived immunosuppression also at local lymph nodes. These obstacles can be overcome by oncolytic virotherapy, as tumor-destroying viruses both provide activation and ere with tumor immunosuppression to expose hidden immunogenic antigens“.
Oncolytic adenoviruses, also known as Conditionally Replicating adenoviruses (CRAds), are cally modified to ate and kill only cancer cellsg'm' it is known that virus-induced tumor apoptosis and/or necrosis leads to release of large amounts of tumor-associated proteins not normally accessible by antlgen~presenting cells, which drives ent cross‘ presentation by tumor~associated DCs in the tumor draining lymph nodes" Virus therapy of cancer has generally been found well ted, however, the overall treatment efficacy has ed modest; upon scrutiny of the immunological effects of virotherapy a clear dominance of virus over tumor has been observed in both mice and human (Figure 2). g the adenovirus’s capsid with synthetic MHOl-restricted tumor-specific peptides will “trick” antigen presenting cells (APCs) to present these tumor antigens as part of the virus. in other words, the present invention utilizing adenovirus capsid as a scaffold to deliver MHC-l restricted peptides would shift the immune response away from the virus and instead toward the tumor.
As used herein “Major Histocompatibility Complex of class I” les refer to one of two primary classes of major histocompatibility complex (MHC) molecules (the other being MHC class II) and are found on nearly every nucleated cell of the body. Their function is to y fragments of proteins from within the cell to T cells; healthy cells will be ignored, while cells containing foreign proteins will be attacked by the immune system. Class I MHC molecules bind es generated mainly from degradation of cytosolic proteins by the some. The MHC lzpeptide complex is then inserted into the plasma membrane of the cell. The peptide is bound to the extracellular part of the class I MHC molecule. Thus, the function of the class i MHC is to display intracellular proteins to cytotoxic T cells (CTLs). However, class I MHC can also present peptides generated from exogenous proteins, in a process known as cross-presentation. As used herein “MHC-l-specific polypeptides" refer to those peptides, which are bound to MHC-l, is. the extracellular part of the class I MHC molecule, and displayed to CTLs.
All the MHC-l peptides (MlPs) are collectively called the immunOpeptidomeM' Only recently, with the use of advanced l09ies there has been the possibility to start looking into the MHC-l immunopeptidome. The crucial difference in the present invention, compared to other strategies attempting to broadly screen the whole immunopeptidome, is that the present invention focuses on specific peptides that are present simultaneously on tumor cells both before and after therapy (Le. which will not be masked or edited away after y) and on 005 following therapy e A significant difference between the present invention and the traditional peptide—based therapy is that the present invention takes full age of the fact that viruses, and in particular adenoviruses, have a privileged means to interact with DCs (hence there is no obligatory need to target DC). Adenoviruses stimulate several n Recognition Receptors (PRRs), Toll~like Receptors‘s'w’ NOD-like receptor family” and inflammasome‘g' predisposing 008 for robust antigen presentation and CTL tion”. To this purpose we show that human primary DCs pulsed with oncolytic adenovirus activate pathways involved in cellular adhesion, cell-cell interaction and signaling, maturation and antigen presentation suggesting that the adenovirus is capable ofpromoting maturation and ion of immature primary dendritic cells (Figure 4).
As used herein “stimulating a peptide-specific immune response" refers to causing an immune response wherein cells representing the specific peptides will be attacked and destroyed. “Immune response” refers to a system involving lymphocytes (Le. white blood cells), either Tor B lymphocytes or the both. T lymphocytes attack ns directly and help in controlling the immune response. They also release chemicals, known as nes, which control the entire immune response. B lymphocytes become cells that produce antibodies. dies attach to a specific antigen and make it easier for the immune cells to destroy the antigen. in one embodiment of the invention one or more polypeptides attached onto a viral capsid are ed from the group consisting of fragments of tyrosinase-related n 2 (TRP-Z), fragments of human melanoma antigen gpiOO (hgpiOO), fragments of melanoma-associated antigen A1 (MAGE-Ai), SllNFEKL, polyK-SIINFEKL, SIlNFEKL-polyK, SLFRAVITK (SEQ ID NO: 4), polyK-SLFRAVITK, SLFRAVITK-polyK, SVYDFFVWL (SEQ lD NO: 5), polyK-SWDFFVWL, SVYDFFVWL-polyK, KVPRNQDWL (SEQ lD NO: 8), ponK-KVPRNQDWL and DWL- ponK. in one embodiment of the invention one type or more polypeptides ed onto a viral capsid comprise SllNFEKL, SLFRAVITK, SVYDFFVWL or KVPRNQDWL. in a r embodiment polypeptide fragments of TRP-2 and hgpiOO (e.g. SVYDFFVWL or KVPRNQDWL) are attached onto the adenoviral capsid. In one embodiment of the invention the polypeptides used in the present invention are sine (polyK) modified. As used herein, ponK may be selected from the group consisting of 3K—15K, 3K—10K, 3K-8K, 5K-8K, 5K-7K and 6K. As used herein “polylysine-modified polypeptide" refers to a polypeptide, wherein a poiylysine sequence has been inserted. Addition of a polylysine sequence to a ptide causes change in the charge of the peptide and the consequent absorption on the surface of the virus.
Adenoviral vector Adenoviruses coated with peptides may be of any type and species of adenoviridae (e.g. not limited to human irus). in one ment of the invention, the adenoviruses are capable of replicating and killing cancer cells while diverting the iral immune reSponse against the tumor (Figure 1). The cancer destroying virus of the present invention coated with patient derived tumor-specific immune-activating peptides enhance and divert the anti- viral ty into anti-tumor immunity.
The adenoviral vectors used in the present invention can be any adenoviral vectors suitable for treating a human or animal. atively, various types of iral vectors can be used ing to the present invention. Also, the vectors may be modified in any way known in the art, eg. by deleting, inserting, mutating or modifying any viral areas. The vectors can be made tumor specific with regard to replication. For example, the adenoviral vector may comprise modifications in E1, E3 and/or E4 such as insertion of tumor specific promoters, deletions of areas and insertion of transgenes. in one embodiment of the invention, the iral vector is an oncolytic iral vector. As used herein “an oncolytic adenoviral vector” refers to an adenoviral vector e of infecting and killing cancer cells by selective replication in tumor versus normal cells. in one embodiment of the invention the vectors are replication competent only in cells, which have defects in the Rb—pathway, specifically Rb-pi6 pathway. These defective cells include all tumor cells in animals and humans. As used herein “defects in the Rb-pathway" refers to mutations and/or epigenetic s in any genes or proteins of the pathway. A tumor specific oncolytic adenovirus may be engineered for example by deleting 24 base pairs (D24) of the constant region 2 (CR2) of E1. As used herein “D24” or “24 bp deletion“ refers to a on of nucleotides corresponding to amino acids 122-129 of the vector according to Heise C. et al. (2000, Nature Med 6, 1134-1139). in one embodiment of the invention the adenoviral vector comprises the 24bp deletion (oncolytic virus) or E1 gene deletion (second generation virus) or the vector is a - dependent . E1 gene deletion may be partial or total on of the E1 region. As used herein “a Helper—dependent vector" refers to a vector, which does not e genes encoding the enzymes and/or structural proteins required for replication and therefore is dependent on the assistance of a helper virus in order to replicate.
The backbone of the adenoviral vector may be of any serotype. In one embodiment of the invention the serotype of the adenovirat vector backbone is selected from serotype 3 or 5. As used herein, “adenovirus serotype 5 (Ad5) nucleic acid backbone" refers to the genome of Ad5 and “adenovirus pe 3 (Ad3) nucleic acid backbone” refers to the genome of Ad3.
Further, the vectors may be chimeric vectors, e.g. Ad5/3, Ad3/5 or Ad5/35 vectors. As an example, “Ad5/3 vector” refers to a chimeric vector having parts of both Ad5 and Ad3 vectors.
In one embodiment of the invention the iral vector comprises a capsid modification (Le. a modification in nucleotide sequences encoding proteins forming the capsid of the . ”Capsid” of the adenovirus refers to the protein shell of a virus. The capsid ts of l oligomeric structural subunits made of proteins called protomers.
Furthermore, fiber knob areas of the vector can be modified. in one embodiment of the invention the adenoviral vector is Ad5/3 or Ad5/35 comprising an Ad5 nucleic acid backbone and a fiber knob selected from the group consisting of Ads fiber knob, Ad35 fiber knob, Ad5/3 chimeric fiber knob and Ad5/35 chimeric fiber knob. in a specific embodiment of the invention the oncolytic adenoviral vector is based on an adenovirus serotype 5 (Ad5) nucleic acid backbone and comprises the D24 on, optionally a transgene and optionally a CpG site. in another embodiment, the oncolytic adenoviral vector is based on an adenovirus serotype 5 (Ad5) nucleic acid backbone and comprises modification of the capsid (e.g. Ad3 fiber knob), optionally the D24 deletion and optionally a transgene.
Insertion of ous elements may enhance effects of vectors in target cells. The use of exogenous tissue or tumor-specific promoters is common in recombinant vectors and they can also be utilized in the present invention. Suitable promoters are well known to a person skilled in the art and they include, but are not limited to, hTERT, CMV, E2F.
The adenoviral vector may also cause expression of any transgene(s) (e.g. granulocyte macrophage colony stimulating factor (GM— CSF)). in one embodiment of the invention, the iral vector comprises one or more transgenes. One example of suitable transgenes is cytokines, which late increased trafficking of immune cells at the site affected by the e, eg. tumor site. Cytokines used in the present invention can be selected from any known cytokines in the art. in one embodiment of the invention the transgene is selected from the group consisting of chemokines and cytokines and signal peptides for the recruitment or manipulation of the logical stroma at the tumor site expecially for what concerns T cells, dendritic cells, macrophages, natural killer cells. The viral vectors of the invention may code for either one or several enes, e.g. nes (e.g. two, three, four, five or more). The adenoviral vector may for example express monoclonal antibodies to specifically block immunological checkpoints (e.g.
CTLA4, PD‘l, PDLi).
A transgene(s) may be placed to different ons of the aden0viral vector. The transgene may be placed for example into a partly or totally deleted E3 , either under the E3 promoter or an exogenous promoter, or into a partly or totally deleted E’l region, either under the E1 promoter or an ous promoter.
In one ment of the invention the adenoviral vector for coating is Ad5D24, Ad5D24CpG or Ad5D24-GMCSF. In Ad5D24-GMCSF GM—CSF transgene is in the place of deleted E3 region (i.e. deleted 6. 7K/gp19K) under the control of E3 promoter (Cerullo V et al. 2010, Cancer Research 70: 4297- 4309). As used herein, CpG refers to CpG moieties added into the adenovirus genome to make the virus more stimulatory. The insertion of CpG-rich regions in the adenovirus backbone increase the capability of adenovirus to stimulate TLRQ in antigen presenting cells hence increasing T cell stimulation and maturation as well as NK activation (Nayak S, Herzosi RW. Gene Ther. 2010 Mar;17(3):295-304.).
The viral vectors utilized in the present inventions may also se other modifications than described above. Any onal components or modifications may optionally be used but are not obligatory for the present invention.
Coating the adenoviral vector According to the present invention the capsid of an adenovirus is coated with synthetic polypeptides or peptides, which are capable of stimulating a e-specific immune response in a subject. The ptides used for coating the adenoviral vectors have not been genetically d by said adenoviral vectors. Herein, the terms eptide“ and “peptide“ are used interchangeably to refer to potymers of amino acids of any length.
The polypeptides can be attached to the capsid by any known suitable chemical or biochemical method. in one embodiment of the invention the peptides have been attached covalently or non—covalently onto the viral capsid. in another embodiment of the invention the polypeptides have been attached to the capsid by electrostatic, disulfide or amide bond linkage or co- delivered and attached to the capsid in a single nanoparticle. The nanoparticleis) may also be attached covalently or non-covalently, e.g. by electrostatic, disulfide or amide bond linkage, to the capsid. As used herein, “nanoparticles” refer to any particles, which are between 1 and 100 nanometers in size. The ostatic linkage strategy takes advantage of the fact that the adenovirus capsid has a negative net total charge, it implies a synthesis of positively charged peptides consisting of poly—lysine attached to a small linker that is attached to the peptide of st. The first gy has two potential ages: 1) it is rapid (for e about 15-30 minutes at room temperature or about 20 min at room temperature), which can be a key feature in personalized drugs and 2) transduction of adenovirus complexed with cation 2629' polymers is significantly increased The polypeptides attached onto the viral capsid may be all the same peptides or different peptides ed from two or more types of different tumor antiges. In one embodiment of the invention the adenoviruses are coated with more than one type of peptides. The peptides can be for example different MHC-I specific polypeptides of the same antigen, MHC-l polypeptides from different ns or a combination of MHC—l and MHC-Il restricted peptides. in one embodiment of the invention the ptides attached onto the viral capsid are selected from the group consisting of Major Histocompatibility Complex of class I (MHC-I)-specific polypeptides (polypeptides binding , Major Histocompatibility Complex of class II (MHC-II)-specific polypeptides (polypeptides binding MHC-II), e specific polypeptides (polypeptides associated with a disease), tumor specific polypeptides (polypeptides associated with tumors or a specific tumor) and DC specific polypeptides (polypeptides binding DC). In a specific embodiment of the ion the ptides attached onto the viral capsid are tumor-specific MHC-I restricted es. These es may be isolated directly from the tumor of patients with a process depicted in Figure 5. By utilizing the method of Figure 5 the polypeptides to be attached onto the viral capsid may be simultaneously presented on the MHC-I of the tumor and from the DCs that have been fed with tumor oncolysate.
DCs. presented by cells having a disease ype or infected by the disease.
The polypeptides to be attached to the capsid of an adenoviral vector include any polypeptides which are at the same time presented by disease or tumor cells and dendritic cells of one patient (e.g. tumor antigens or peptides derived from them). Examples of suitable peptides include, but are not limited to gp100.
The concentration of polypeptides on the capsid may vary and in one embodiment of the invention, the polypeptides are at a concentration of at least 500 nM.
According to the present disclosure, in the production of the patienttailored polypeptide coated adenoviruses disease cell-derived or tumor-derived MHC-I-loaded peptides can be isolated and identified, sized and admixed on to the capsid of a DC-stimulating oncolytic adenovirus. However, the method comprises at least two steps. First, the most immunogenic ptides loaded on MHC-I are identified, and secondly, these polypeptides are loaded on the tic adenovirus capsid.
Pharmaceutical itions The present disclosure provides not only therapeutic methods and uses for treating disorders but also pharmaceutical compositions for use in said methods and therapeutic uses. Such pharmaceutical compositions comprise coated adenoviruses, either alone or in combination with other agents such as a therapeutically effective agent or agents and/or a ceutically acceptable vehicle or vehicles.
A pharmaceutically acceptable vehicle may for example be selected from the group consisting of a ceuticaliy acceptable solvent, diluent, adjuvant, excipient, , carrier, antiseptic, filling, stabilising agent and ning agent. Optionally, any other components normally found in corresponding products may be included. in one embodiment of the invention the pharmaceutical composition comprises polypeptide coated adenoviruses and a pharmaceutically acceptable vehicle.
The pharmaceutical composition may be in any form, such as solid, semisolid or liquid form, suitable for stration. A formulation can be selected from the group consisting of, but not d to, for example solutions, emulsions or suspensions. Means and methods for formulating the present pharmaceutical preparations are known to persons skilled in the art, and may be manufactured in a manner which is in itself known.
Therapies Any disease or disorder, which can be d, which progress can be slowed down or wherein the symptoms can be ameliorated by stimulating the peptide-specific immune response against the abnormal cells caused by the disease, is included within the scope of the present invention. in one embodiment of the invention peptide-Specific immune reSponse is ed from the group consisting of anti-tumor (against primary and/or ary tumors), ancer (against primary and/or secondary malignant neoplasia), anti-infection and anti-virus immune response. in these cases the immune response is ed against a tumor (including both malignant and benign tumors as well as primary and secondary tumors), cancer (i.e. either primary or secondary malignant neoplasia), infectious disease (eg. malaria), viruses (in case of viral infection e.g. influenza, SARS—Cov or HIV) etc. pondingly.
For e any cancer can be a target of the coated adenovirus of the present invention. In one embodiment of the invention, the cancer is selected from the group consisting of nasopharyngeal cancer, synovial cancer, hepatocellular cancer, renal cancer, cancer of connective tissues, melanoma, lung cancer, bowel cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, throat cancer, oral cancer, liver cancer, bone cancer, pancreatic cancer, choriocarcinoma, gastrinoma, pheochromocytoma, prolactinoma, T- cell leukemia/lymphoma, neuroma, von Hippel-Lindau disease, Zollinger- Ellison syndrome, adrenal cancer, anal cancer, bile duct cancer, bladder cancer, ureter , brain cancer, oligodendroglioma, neuroblastoma, meningioma, spinal cord tumor, bone cancer, osteochondroma, chondrosarcoma, Ewing's sarcoma, cancer of unknown y site, carcinoid, oid of gastrointestinal tract, fibrosarcoma, breast cancer, Paget's disease, al cancer, colorectal cancer, rectal , gus cancer, gall bladder cancer, head cancer, eye cancer, neck cancer, kidney cancer, Wilms' tumor, liver cancer, Kaposi's sarcoma, prostate cancer, lung cancer, testicular cancer, Hodgkin's disease, dgkin's lymphoma, oral , skin cancer, mesothelioma, multipie myeloma, ovarian cancer, endocrine pancreatic cancer, glucagonoma, pancreatic cancer, yroid cancer, penis cancer, pituitary cancer, soft tissue sarcoma, retinoblastoma, small ine cancer, stomach , thymus cancer, thyroid cancer, trophoblastic cancer, diform mole, uterine cancer, endometrial cancer, vagina cancer, vulva cancer, acoustic neuroma, mycosis des, insulinoma, carcinoid syndrome, somatostatinoma, gum cancer, heart , lip cancer, meninges cancer, mouth cancer, nerve cancer, palate cancer, parotid gland cancer, peritoneum cancer, pharynx cancer, pleural cancer, salivary gland cancer, tongue cancer and tonsil cancer.
As used herein, the term “treatment“ or “treating“ refers to administration of at least coated adenoviral vectors or a pharmaceutical composition comprising coated adenovlral vectors to a subject. The term “treating“, as well as words stemming therefrom, as used , do not necessarily imply 100% or complete treatment or increase. Rather, there are varying degrees of which one of ordinary skill in the art recognizes as having a ial benefit or therapeutic effect. in this respect, the present inventive methods and uses can provide any degree of treatment or prevention of a disease. Therefore, “treating“ includes not only complete cure but also for example prophylaxis, ration, or alleviation of disorders or symptoms related to a disease in question, such as cancer, tumor, infectious e or viral infection. Therapeutic effect may be assessed by any method known to a person skilled in the art, for example by monitoring the symptoms of a patient or disease markers in blood.
As used herein, the term “subject" refers to a subject, which is selected from the group consisting of an animal, a mammal or a human. In one embodiment of the invention, the subject is a human or an animal.
The adenovirus coated with polypeptides is administered to a subject in a therapeutically effective amount, which causes the peptide-specific immune response. As used herein, the term "therapeutically effective amount" refers to an amount of coated adenovirus with which the harmful effects of a disease or disorder (e.g. ) are, at a minimum, ameliorated. The harmful effects include any able or noticeable effects of a subject such as pain, dizziness or swelling.
Only one administration of coated adenoviral vectors or pharmaceutical composition of the invention may have therapeutic effects. On the other hand the treatment may contain several administrations. Adenoviral vectors or pharmaceutical composition may be stered for example from i to 10 times during 2, 3, 4, or 8 weeks, or during the ent period. The length of the treatment period may vary, and for example may last from two to 12 months or more. in some cases it is also possible to use l treatment periods for one patient.
The effective dose of vectors depends on at least the subject in need of the treatment, type of the disease and stage of the e. The dose may vary for example from about ‘le08 viral particles (VP) to about ‘le014 VP, specifically from about 1x109 VP to about 1x1013 VP and more ically from about 5x109 VP to about 1x1012 VP.
Administration of the coated adenovirus can be conducted through any suitable method known to a person skilled in the art. in one embodiment of the invention, the administration of the adenoviral vectors is conducted through an intratumoral, infra-arterial, intravenous, intrapleural, esicular, intracavitary or peritoneal injection, or an oral administration. it is also possible to combine ent routes of administration.
The coated adenoviruses may also be used together (simultaneously or sequentially) with other therapeutic agents or therapeutic s or a combination of treatments. For example the method or use of the ion may further comprise radiotherapy, herapy, administration of other drugs or any clinical operatibns.
Before classifying a human or animal t as suitable for the therapy of the present invention, the clinician may examine a patient. Based on the results deviating from the normal and revealing a disease, such as cancer, the clinician may t methods or treatment as described herein for a patient.
Identification of specific peptides for coating The present sure further relates to a method for identifying at least tumor-specific and MHC-I-specific polypeptides from a subject. The method utilizes qualitative and quantitative study on MHC-I immunopeptidome of tumors and DCs d to tumor lysate, ically in vitro. The methodology in short, summarized in Figure 5, involves isolation of MHC I molecules from both tumor cells and DCs pulsed with oncolysate in vitro (virus infected tumor cells) and sequencing of the MHC-associated polypeptides by mass-spectrometry based technology. logically relevant peptides will be presented by both, tumors and dendritic cells pulsed with tumor . For example, the use of the OVA- expressing mouse model may facilitate the validation of the system, in fact well known immunogenic OVA derived peptides (e.g. SIINFEKL) result from the mouse experiments and may serve as positive control.
Tumor cells of a subject before and after in vitro adenoviral infection are used in the method in order to block those polypeptides which are displayed by the cell due to the viral infection. DCs pulsed with tumor oncolysate in vitro are also used in the method in order to allow presentation of tumor antigen. The advantage of using not only tumor but also DC pulsed with tumor oncolysate for the isolation of tumor specific es is to better identification of the immunological active es (only if a peptide is presented on both tumor and DC there will be an ent immune response). Isolation of MHC-I molecules from the tumor cells and dendritic cells may be conducted by any suitable isolation method of the art.
Thereafter, cing of the polypeptides can be carried out by any suitable mass-spectrometry based logy (e.g. LC MS/MS) for identifying the MHC- associated peptides. The polypeptides ted both by tumors and dendritic cells can be identified by comparing the polypeptides presented by these cells.
Common polypeptides in two groups i.e. polypeptides presented by DCs pulsed with lysate minus DCs not pulsed (to eliminate DC-self peptides) and polypeptides presented by virus-infected tumors and non infected tumors (to eliminate virusspecific peptides) are suitable for coating the adenoviruses. Comparison of polypeptides can be carried out ly or by any bioinformatics method known to a person skilled in the art. Optionally, in vitro, ex vivo and/or in vivo validation can be performed for any specific polypeptide or a combination thereof. in one embodiment of the invention, in addition to ing MHC-l molecules from infected and uninfected tumor cells as well as infected dendritic cells, the method further ses isolating MHC-l molecules from uninfected dendritic cells and identifying the MHC-Lassociated ptides; and identifying those polypeptides which have been presented by the hfected and uninfected tumors of steps iii) and iv) and by the infected dendritic cells of step iii) but not by the uninfected dendritic cells. In a specific ment of the invention infection of tumor cells and DCs with adenoviral vectors takes place in vitro. Adenoviral vectors used for the method of the present invention can be any iral vector, for example any one of these vectors described in the earlier chapters. in one embodiment of the invention the method for identifying tumor-specific and specific polypeptides from a subject is used for selecting one or more tumor-specific and MHC-i-specific polypeptides for coating the adenoviral capsid. Any of these tumor-specific and MHC-l-specific polypeptides or a combination thereof can be used for coating.
It will be obvious to a person skilled in the art that, as the logy advances, the ive concept can be implemented in various ways. The invention and its embodiments are not d to the examples described above but may vary within the scope of the claims.
EXAMPLES The following es demonstrate at least analysis of the tumor MHC-l immunopeptidome for isolating and ing tumor—specific polypeptides, generation and physical characterization of tumor-specific polypeptide—coated oncolytic adenoviruses, and characterization of the coated adenoviruses in animal models (eg. i) therapeutic efficacy, ii) capacity to divert the anti-virus immunity into anti-tumor immunity and iii) capacity to recruit cells of the immune system and to promote T cell responses).
Oncolytic adenovirus preparation All oncolytic adenoviruses (OAd) were generated and ated using standard ols, as previously described (8). Briefly, viruses were amplified by infecting 10 T175 flasks with 70—80% confluent A549 cells at a multiplicity of infection (MOl) of 30. Three days post-infection, the cells were collected and lysed through four freeze (~80°C) and thaw (37°C) cycles.
Adenoviral particles were then separated from the cell debris and impurities by two ultra-centrifugations (22,000 and 27,000 rpm) on 030! gradients. The recovered bands were purified by overnight dialysis at 4°C against A195 buffer with continuous stirring. Specifically, dialysis cassettes with a molecular weight ’iO cutoff of 10,000 kDa (Pierce, Life Technologies) were used. The purified s were recovered from the cassettes, aliquoted and stored at -80°C.
The integrity of the iral genome was assessed by PCR using primers specific for the E3 gene and the D24 deletion in the E‘lA gene.
The viral particle titer was determined using the spectrophotometric method, whereas the infectious titer was ined by immunocytochemical staining, as described ere in this section. The protein concentration of the viral preparation was determined by the Bradford assay using Bio-Rad n Assay Dye Reagent Concentrate (Bio-Rad Laboratories, Hercules, CA, USA). All spectrophotometric readings were performed with a SPECTROstar Nano spectrophotometer (BMG Labtech, Ortenber, Germany).
All viruses used in this study have been previously reported: Ad5D24 is an adenovirus that es a 24-base-pair deletion (D24) in the EA gene (9), Ad5D24-CpG is an OAd bearing a CpG-enriched genome in the E3 gene (30), and Ad5D24-GM-CSF is an OAd expressing GM-CSF under the l of the viral E3 er (8).
Analysis of the tumor MHC-l immunopeptidome to isolate and select tumor-specific peptides Method ta: Mouse CD11c+—sorted bone morrow dendritic cells were harvested from C57BL/6 mice and cultured for 1 weekza' Cells were then exposed to: A) PBS as control, B) Oncolysate from VA cells (the oncolysate comes from Bits-OVA cells infected with oncolytic adenovirus Ad5024 until their te Iysis), C) B‘lS—OVA cell lysate obtained by freezing and thawing of the cells.
At different time points MHC-l loaded with peptides were isolated from viable DCs using mild acid elution25' At the time of the analysis, peptides were dissolved in aqueous solution and analyzed by nano LC-MS/MS on a bitrap Elite mass spectrometer (Thermo Fisher Scientific). Database searches were performed against the ational protein Index mouse database version 3.23 containing 51536 sequences and 24497860 residues, httpzl/www.ebi.ac.uk/lPl/lPlhelp.html). nt peptides were in the group formed by the peptides that are commonly present in both the groups, DC- pulsed with Iysate minus DC-not pulsed (to eliminate DC-self peptides) and Bl6-OVA virus-infected minus B‘lB-OVA-non infected (to eliminate virus— specific es).
Method 1b: We first reduced the complexity of the immunopeptidome of Method 1 a in silico. Prediction of MHC-l class peptides /Iwww.syfpeithi.de/home.htm), Functional tion of the proteins (http://davld.abcc.ncifcrf.gov) and (http://www.ingenuity.com) were used.
Oncomine analysis (https://www.oncomine.org) was used to suggest the level of expression of a given protein in different human cancers and cell lines. Most importantly, we validated our peptides using an e tool predictor . mentally, to select the most immunogenic peptides we used a mouse lFN-gamma T (Mabtech AB, Sweden) on splenocytes, tumors and lymph nodes harvested from 057BL/6 mice and pulsed with all the different peptides isolated from method We.
Briefly, CSYBL/B mice bearing BiG—OVA tumors were treated with oncolytic adenovirus (Ad5024). One to two weeks after treatment, mice were euthanized and organs and tumors were harvested and reduced to a single cell su3pension for the lFN-gamma ELISPOT analysis (Mabtech, Palo Alto CA). uently, once we had identified a pool of a few of the most immunogenic peptides we generated custom er or Pentamer (Proimmune, UK) for flow cytometer-based detection of specific CD8 T cetls recognizing these peptides on MHC-l molecules.
Generation and physical characterization of tumor-specific peptide- coated oncolytic adenoviruses Because OVA-derived es are very well known, as proof-of- concept we first generated an OVA-specific coated virus (Figure 6). More specifically, we generated a SllNFEKL-coated adenovirus (SllNFEKL (SEQ lD NO: 1) is the most genic OVA derived peptide); a SllNFEDL—coated virus (SIINFEDL (SEQ ID NO: 7) is an antagonist of SllNFEKL peptide); a NE-coated virus (FlLKSlNE (SEQ ID NO: 3) is a scramble peptide of SllNFEKL).
Method Za: in order to generate a peptide-coated oncolytic adenovirus different strategies were taken into account (Figure 7).
One will use electrostatic binding between the virus and the peptides and two others will involve covalent bonds between virus and peptides. l. Electrostatic interaction. Positively d peptides complexed with negative virus capsid ll. nt bond. Disulphide bond with the cysteine of the protein of the capsid 27’28‘ ill. Covalent bond. Amidic bond. Succinimidyl ester reaction with amine groups of Lysine of capsid The methods of linking are described in the corresponding reference documents.
In one embodiment of the invention peptide—coated oncolytic adenoviruses were prepared as follows: PeptiCRAd complex formation All PeptiCRAd complexes described in this work were prepared by mixing oncolytic viruses (as described under the title ”Oncolytic adenovirus preparation") and polyK-epitopes at a 1:500 ratio (see s 8A and 12) according to the following protocol: i) for each microliter of viral preparation used, the ponding number of micrograms of n present was calculated; ii) then, for each microgram of viral protein, 500 pg of peptide was added; iii) after vortexing, the e was incubated at room temperature (RT) for 15 min; and iv) the solution was vortexed and used for assays or animal injections. New PeptiCRAds were prepared before each experiment using fresh reagents. All dilutions of virus and peptides ed before tion were med in sterile Milli-Q water adjusted to pH 7.4. The PeptiCRAds were then diluted with the buffer required by the assay.
Method 2b: lnfectivity of this peptide coated virus from Method 2a was assessed in vitro by rase assay and by qPCR in different cell lines (human and murine)“. To assess infectivity, a panel of different tumor cell lines with different levels of expression of CAR were infected with different concentrations of coated virus expressing luciferase (Ad5D24~Luc) (1, 10, 100, 1000 VP/cell); uncoated virus was always used as control. At different time points luciferase expression was quantified. Simultaneously, total DNA was harvested and viral DNA replication was quantified by qPCR. tic activity in vitro was tested by TCID50 and MTS “ in one embodiment of the invention, the infectivity was studied by ICC as s: Infectivity assay by ICC Tumor cells were seeded at 2.O><105 cells per well on 24-well plates in 3 or 5 replicates. The following day, the cells were infected with 100 pl of viral dilutions. The plates were then centrifuged for 90 min at 1,000 rcf at 37°C, followed by incubation for 48 h. After the incubation period, the culture media were removed, and the cells were fixed by incubation with 250 pl of ice~cold methanol for 15 min. Once the methanol was removed, the cells were washed 3 times with 300 pl of PBS supplemented with 1% bovine serum albumin (BSA). The cells were then stained with 250 pl of mouse monoclonal anti- hexon antibody (Novus Biologicals, Littleton, CO, USA), diluted 122,000, for 1h at RT in the dark. The cells were then washed and stained with 250 pl of -streptavidin-conjugated goat ouse antibody, diluted 1:500 with PBS/1% BSA, for 1h at RT in the dark. The cells were subsequently incubated with 250 pl of extravidin-peroxidase (Sigma-Aldrich, St. Louis, MO, USA), diluted 1:200, for 30 min at RT. The cells were washed extensively, and DAB staining solution (Sigma-Aldrich, St. Louis, MO, USA) was prepared according to the manufacturer's instructions. A total of 250 ul of DAB staining solution was then applied to each well, and the cells were monitored under a microscope for the appearance of dark spots. When the l signal-to-noise ratio was reached, the reaction was quenched by the addition of PBS/1% BSA (500 pl per well). For each replicate (is, well), 5 images of non-overlapping fields were acquired using an AMG EVOS XL microscope (AMG group, Life Technologies). The following formula was used to ine the infectious titer: wall area 1 1 ml 1nfec wasc‘ t'tL er , = x * ——-—-—-—~a< —————-——-—-—-— ”—* field area dilution factor Volume of dilution d For the ivity comparisons, the data are presented as the average number of spots in each field.
In support of Methods 2: The negatively charged adenovirus capsid was coated electrostatically with tumor specific peptide. This complex had a variation in Z- potential that is proportional to the amount of peptides. This change of Z- potential showed that positively d peptides were binding the viral capsid determining the inversion of charge (Figure 8A line with dots). Once all the negative s of the capsid had been saturated, the Z-potential seemed to rich a plateau (Figure 12 line with circle). Uniform monodispersed complex can be formed with concentration of polypeptides more than 500nM for proceeding to in vitro and in vivo efficacy.
To further terize the peptide coated adenovirus complex we med several viability assays (MTS assay) comparing the efficacy of cell killing of PeptiCRAd with uncoated tic virus (Figure 9). The results indicate that the coating of the virus constantly result in unaltered or better cell killing ty compared with uncoated oncolytic viruses.
In one ment of the invention the viability assay was d out as follows: Viability assay Tumor cells were seeded at 1.0x104 cells per well on 96-well plates in growth media with 5% PBS. The next day, the media were removed, and 50 pl of virus, diluted in gowth media with 2% F88, was used to infect the cells for 2 h at 37°C. Afterwards, 100 pl of growth media with 5% PBS was added, and the cells were again incubated at 37°C. The gowth media were changed every other day. When the most infective conditions (100 vp/cell) showed an extensive cytopathic effect (>90%), cell viability was determined by MTS assay according to the manufacturer’s protocol (CellTiter 96 AQueous One Solution Cell Proliferation Assay; Promega, Nacka, Sweden). Spectrophotometric data were acquired with Varioskan Flash Multimode Reader (Thermo Scientific, Carlsbad, CA, USA).
Study design The sample size was ined using the following equation: n=1+2C(—)23 where C is a constant based on o and (3 values, 3 is the estimated ility and d is the effect to be observed (34). For all of the animal ’10 experiments, a power (1-3) of at least 80% and a significance (or) of 0.05 were considered. The rules for stopping the data collection were i) death of more than 60% of the mice in one or more groups and ii) total clearance of the tumors. All of the mice that died before the end of the experiment were excluded from the growth curves to preserve the statistical integrity of the analysis.
The objective of the research was to use melanoma models to test whether OAds could represent a valid adjuvant for a peptide cancer-vaccine approach. Additionally, two specific questions were posed: i) Can PeptiCRAd limit the growth of distant, untreated tumors? ii) Can the efficacy of PeptiCRAd be enhanced by targeting multiple tumor antigens instead of a single one? To answer these questions, we utilized immunocompetent or humanized mice bearing melanoma tumors. The mice were randomly assigned to each experimental group, and no blinding was d.
Cell lines, ts and human samples The human lung oma cell line A549, the human colorectal adenocarcinoma cell line CACO-Z, the human malignant ma cell line SK-MEL-2, the human melanoma cell line H8294T and the mouse melanoma cell line Bio-F10 were purchased from the American Type Culture Collection (ATCC; as, VA, USA). The ceil tine Bis-OVA (35), a mouse melanoma cell line expressing n OVA, was kindly provided by Prof. Richard Vile (Mayo , Rochester, MN, USA).
The A549, CACO-2 and BtS-OVA cell lines were cultured in low- glucose DMEM (Lonza, Basel, Switzerland), the H8294T cell line was cultured in high-glucose DMEM (Gibco, Life Technologies, Carlsbad, CA, USA), the SK-MEL-Z cell line was cultured in EMEM (ATCC), and the B‘iB—F10 cell line was cultured in RPMI-1640 (Gibco, Life Technologies). All media were mented with 10% fetal bovine serum (FBS; Gibco, Life Technologies), 2 mM GiutaMAX (Gibco, Life logies), and 100 U/ml penicillin and 0.1 mg/mi streptomycin (Gibco, Life Technologies). The BlS-OVA cell line was also ed in the presence of 5 mg/mi Geneticin (Gibco, Life Technologies) to ensure the selection of OVA~expressing cells. During the culture period or when needed for assays, the cells were washed with 1X phosphate—buffered saline (PBS) and detached by incubation with 1X TrprE Express (Gibco, Life Technologies) for 3 min at 37°C.
SIINFEKL (OVA257-264). poiyK-SHNFEKL, SllNFEKL-polyK, poiyK- AHX-SIINFEKL, poiyK-SVYDFFVWL (TRP—Ziso-iaa), DOIyK-KVPRNQDWL (hgp10025-33) and polyK—SLFRAVITK (MAGE—A195.1g4) peptides were sed from Zhejiang Ontores Biotechnologies Co. (Zhejiang, China). The purity of all peptides was estimated to be >80%, and they were analyzed by mass spectral analysis.
In the examples chapter polyK refers to 6K.
The net charge of peptides was calculated by the e Property Calculator Ver. 3.1 online tool (h_ttp://www.biosyn.com/PeptidePropertyCalculator/PeptidePropefiyCalculator. aspa- The genotype of the SK-MEL-2 cell line was HLA-A*03 - *26; B*35 - *38; 004 ~ *12. Buffy coat from a healthy donor was also obtained from the h Red Cross service, and the genotype was ined as HLA—A*O3 — *03; B*O7 ~ *27; (3’01 — *0? terization of coated adenoviruses in animal models Method3a: We tested in vivo the efficacy, immunogenicity, toxicity, biodistribution of the coated-viruses vs ed regular oncolytic viruses.
Efficacy and immunogenicity were tested in C57BL/6 mice g BiB-OVA tumors. The SiINFEKL-coated virus presented a more robust anti-OVA response that translated into a mere preminent tumor control (efficacy), compared with other coated viruses (antagonist, le and uncoated).
Simultaneously, through adaptive transfer of radioiabeied cells (DOS and T cells) the trafficking of these cells to the tumor microenvironment was also ed. Finally, toxicity and biodistribution of the modified adenoviral vector was also d.
To study the efficacy of the coated viruses, different groups of 057BL/6 mice (N=15 per group) bearing syngeneic Bi6-OVA tumors (two tumors per mouse) were treated as follows: a) SliNFEKL-coated virus b) SilNFEDL-coated virus 0) FiLKSINE—coated virus and d) uncoated virus as control. At different time points starting from 3 days after the administration of the virus, two mice per group was euthanized and spleen, lymph nodes and tumor were harvested into a singie cell suspension for ELISPOT, coucuiture and flow cytometry analysis. aneously tumor growth was measured with standard caliper over time. Flow cytometry analysis revealed directly the quantity of SIiNFEKL-specifc T cells in the tumor, in the spleen and in the lymph nodes (tumor draining and not). For this analysis we used SllNFEKL— specific pentamers (eg. 31). Mouse lFN-gamma ELlSPOT also gave us quantitative indication of anti-OVA SIINFEKL) T cell activation. in the co- e experiment we tested in vitro the capability of T cells (harvested from experimental mice) to kill 816 and B16-OVA. Cells were tured at different ceii:target ratios and 816 and Bits-OVA viability was assessed by MTS or MTT assay. In all this experiment T ceii harvested from OT—I mice was used as control. CMT64—OVA model, which is a murine tumor expressing OVA where the human adenovirus is semi—permissive”, was also used.
Method 3b: The anti—tumor activity and immunogenicity of a virus coated with: i) OVA—peptide (SIINFEKL (SEQ iD NO: 1)), ii) 816 peptide TRP2 (SVYDFFVWL (SEQ ID NO: 5)), iii) hgpiOO peptide QDWL (SEQ iD NO: 6)) or iv) new peptides identified in method 1 were compared.
These viruses were tested for their efficacy and capacity to induce an umor immune response. iral response was compared with the anti-tumor se (ELISPOT and Pentamer analysis). The capacity to induce an immune response to a different epitope (e.g. OVA-virus trigger a TRP2 response, epitope ing) was also assessed. Methods used in this method have already been described in method 3a.
Studies based on methods 2 and 3: We ted an OVA-specific PeptiCRAd (SIINFEKL—coated oncolytic adenovirus) as described in Figure 7 gy I. Briefly, synthetic SIINFEKL peptides were sized and attached to a poly-lysine linker (polyK—SIINFEKL) to confer to the peptides a positive net charge and xed with naked virus that has a negative net charge, 30 minutes prior injection. The complex was then intratumorally administered to mice bearing subcutaneous 816~OVA tumors. Tumor growth was monitored and at the end of the experiment mice were euthanized, tumors were collected and OVA— specific T cells were quantified by flow cytometry (Figure 10). 1O This experiment demonstrates the superiority of the modified adenoviral vector of the present invention ed to virus alone and to virus and peptides administered separately. it also shows the importance of the correct formulation of the coated virus, as with higher concentrations of peptides it seems to induce less tumor specific T cells (data not .
Second generation coated adenovr'ruses Method 4: Second, Generation PeptiCRAd were generated by coating oncolytic viruses with more than a single peptide to elicit a more robust and polyvalent immune response. These new viruses were characterized as in Method 2 and the efficacy was assessed as described in method 3.
Subsequently, we coated a ne-armed oncolytic adenovirus with several types of polypeptides. The polypeptides can either be different MHC-l specific peptides of the same antigen, or MHC-i peptides from different antigens, or a combination of MHC-l and MHC~li restricted peptides.
Methods used for analyzing coated tic viruses Zeta potentiai and c light scattering (DLS) analysis Coated tic virus samples were prepared as described under the title “PeptiCRAdcomplex formation". Each sample was then vortexed and diluted to a final volume of 700 pl with sterile Milli—Q water ed to pH 7.4, after which the sample was transferred to a polystyrene diSposable e to determine the size of the complexes. The sample was then recovered from the cuvette and transferred to a DTS’iO‘IO disposable capillary cell (Malvern, Worcestershire, UK) for zeta potential measurements. All measurements were performed at 25°C with a Zetasizer Nano ZS (Malvern).
The interaction of polyK-SllNFEKL or SllNFEKL with OAds was evaluated using SPR. Measurements were performed using a mum-parametric SPR NaviTM 220A instrument (Bionavis Ltd, Tampere, d). This instrument comprises a temperature-controlled dual flow channel with an integrated fiuidic system and an auto-sampler for buffer and sample handling.
Q water with its pH adjusted to 7.4 was used as a running .
Additionally, a constant flow rate of 30 ul/min was used throughout the experiments, and temperature was set to +20°C. Laser light with a wavelength of 670 nm was used for surface plasmon excitation.
Prior to the SPR experiment, a sensor slide with a silicon dioxide surface was activated by 3 min of plasma treatment ed by coating with APTES ((3-aminOpropyl)triethoxysilane) by incubating the sensor in 50 mM APTES in toluene solution for 1 h. The sensor was then placed into the SPR device, and the OAds were immobilized in situ on the sensor surface of the test channel by injecting 50 ug/ml OAds in Milli-Q water (pH 7.4) for approximately 12 min, followed by a 3 min wash with 20 mM CHAPS (Ci—[(3— cholamidopropyl)dimethylammonio]propanesulfonate). The second flow channel was used as a reference and was injected with Milli-Q water (pH 7.4), followed by washing with CHAPS. The baseline was observed for at least 10 min before sample injections. PoiyK-SilNFEKL or SllNFEKL was then injected into both fiow channels of the flow cell in parallel, with increasing concentrations.
Cross-presentation experiment Fresh spleens were collected from naive CSYBL/G mice and forced h a “IO-um cell strainer (Fisher ific, Waltham, MA, USA). Red blood cells were lysed by incubating the samples with 5 ml of ACK lysis buffer (Life logies) for 5 min at RT. Afterwards, splenocytes were washed and prepared for the assay 6 cells in 800 pl of 10% RPMl-164O e media for each condition tested). A total of 200 pl of SIlNFEKL, polyK-SlINFEKL, SllNFEKL-polyK or SllNFEKL-AHX-polyK peptide dilution (0.19 ) was added to the splenocytes. To test OVA~PeptiCRAd, an infectious condition of 100 vp/cell was used (a total of 7.9x10g vp mixed with 37.5 pg of ponK- SllNFEKL in 200 pl of 10% RPMl-1640). The PeptiCRAd complex was prepared as described under Method 2. The splenocytes were then ted for 2 h at 37°C. Afterwards, the cells were ively washed and stained with either APC anti-mouse H—2Kb bound to SllNFEKL or APC Mouse lgG1, K lsotype Ctrl (BioLegend, San Diego, CA, USA). After a 30-min incubation on ice, the samples were washed and analyzed by flow cytometry.
Flow cytometry analysis The tumors, spleens and lymph nodes of treated mice were collected, forced through a 70-pm cell er and ed overnight in 10% RPMl—164O media. When ary, the samples were frozen in RPMl-164O (with 10% FBS and 10% DMSO) and stored at -80°C. Singlecell sions were stained with fluorochrome-conjugated monoclonal antibodies and analyzed using 3 BD LSR ll (BD Biosciences) or a Gallios (Beckman Coulter)‘ flow cytometer and FIowJo software (Tree Star, Ashland, OR, USA). Sterile PBS was used as the staining buffer. Epitope-specific T cells were studied using MHC Class l Pentamers (Prolmmune, Oxford, UK). Other antibodies used included the following: murine and human Fc block CDtB/32 (BD Pharmingen); FITC anti-mouse CD8 and FITC anti-human CD8 (Prolmmune); PE/Cy7 anti—mouse CD38, PE/Cy?’ anti-mouse CD19, FlTC anti-mouse CD11 0, PeGC/Cy5.5 anthmouse CD86, APC anthouse H-2Kb bound to SllNFEKL and APO Mouse IgGi, K lsotype Ctrl (BioLegend). All staining ures were performed according to the manufacturer’s endations.
Statistical analyses Statistical icance was determined using GraphPad Prism 6 (GraphPad Software, lnc., La Jolla, CA, USA). A ed description of the statistical methods used to analyze the data from each experiment can be found in each Brief Description of the Drawing.
Animal experiments and ethical Issues Animal experiments were done under the Finnish and European law and ation. The animal permit (ESAVl/5924/04.10.03/2012) has been revised and accepted by the Finnish authorities (the Experimental Animal Committee of the University of Helsinki and the Provincial Government of Southern d). Fully immunocompetent 6 mice were obtained from Scanbur (Karlslunde, Denmark), and immunodeficient triple-knockout NOD.Cg- PrkchC’d-ILQrg’mW’JSZJ mice were obtained from Jackson Laboratories (Bar Harbor, ME, USA). All mice were purchased at 4~6 weeks of age and were quarantined for 2 weeks before the study. The mice were kept in cages with isolated and controlled airflow, and they had unlimited access to food during the entire study period. The health status of the mice was frequently monitored, and the animals were sacrificed at the first signs of pain or distress.
All procedures were performed in a biosafety level 2 cabinet under sterile conditions.
For the efficacy experiments, tumor cells were ted at 60-70% confluence (logarithmic phase of growth) and were injected subcutaneously (s.c.) into the flanks of mice. The number of tumor cells injected into each flank varied according to the cell line type: 3X105 BiB-OVA, 1X105 BfG-FtO, and 2><iO6 SK-MEL-Z. In all experiments, three treatment injections were given.
The tumor growth was then followed, and the tumor volume was determined using the formula.
According to our license, the humane endpoints were as follows: i) weight loss of 25%, ii) a tumor diameter >15 mm, and iii) evident signs of pain (reduced ty or ulceration of the tumor). Euthanasia was performed by carbon dioxide inhalation followed by cervical dislocation.
Results The negative charge of the adenovirus capsid can be used to complex positive!y d immunogenic peptides, forming PeptiCRAd.
Adenovirus capsids bear a highly negative net charge (36); hence, we hypothesized that a positively charged MHC-i-restricted peptide would bind to the capsid by electrostatic interaction, covering the virus with immunologically relevant peptides (i.e., tumor-specific restricted peptides). To test our esis, we used the BiS-OVA tumor model (37).
This cell line expresses chicken ovalbumin (OVA) and presents the OVA- d peptide SllNFEKL, which we used as a model e, on MHC-l.
To allow for electrostatic interactiOn between the neutral, hobic SllNFEKL e and the negative viral surface, we added a poly—lysine ) chain to the peptide sequence. This chemical modification increased the net charge of the e from O to +6 mV under physiological conditions. Next, we investigated the interaction between the viral capsid and modified es by e plasmon resonance (SPR). In particular, we coated an APTES silica SiOg sensor with OAds and injected increasing concentrations of SllNFEKL or polyK—SliN into the flowing system (Figure SB).
No increase in the signal was observed with the unmodified peptide (Figure 88, dashed line), whereas a concentration-dependent increase in the signal was observed with the modified peptide (Figure 8B, solid line), demonstrating that the cation of the peptide significantly sed the ction with the adenovirus capsid.
Next, we investigated the optimal concentration of peptide required to efficiently cover the viral surface. To this end, we evaluated the net charge and hydrodynamic diameter of the virus-peptide complexes resulting from different OAdzpeptide ratios (1 :5, 1:50, 1:100 and 1:500). We observed a clear relationship between the amount of positive peptide in the reaction and the net charge of the complexes (Figure 8A). The lowest ratio (1:5) was able to increase the charge of the viral particles from -29.7:0.5 to +6.310.06 mV, although under these conditions, heavy aggregation was ed, as indicated by an increase in the size of the complexes (8002135 nm). Above 1:5, the net charge increased, reaching plateau-like kinetics; in fact, we measured zeta potentials of +17.5:i:0.2, +18.4;i:0.1 and 8 mV for the 1:50, 1:100 and 1:500 ratios, respectiveiy. However, only at a ratio of 1:500 did the diameter of the x decrease below 120 nm, which represents the normal diameter of adenoviral particles. (Figure 8A) The same experiment has also been repeated with concentration of the es and not ratio to facilitate repeatability (Figure 12). ed MHC—l es adsorbed onto PeptiCRAd are efficiently cross- presented.
To induce an effective cytotoxic T-lymphocyte-mediated immune response, peptides must be cross-presented to naive CD8+ T lymphocytes via MHC-l on APCs. Therefore, we investigated whether the presence and the on of the polyK chain could affect the efficienCy of cross-presentation. For this purpose, we pulsed ex viva-cultured splenocytes (from CSYBL/S mice) with either natural SllNFEKL or two different lysine-extended versions: polyK- SIINFEKL minus extended) and SIINFEKL-polyK (C-terminus extended).
As a negative control, we included ed KL containing an amino caproic (AHX) residue, which is a well-known analog of lysine that can inhibit the proteolytic activity of the proteasome. We then assessed the cross- presentation of SIINFEKL with the use of an antibody that specifically izes MHC-l loaded with SIINFEKL (38).
As expected, 98.5% of the SllNFEKL-pulsed splenocytes were positive for the presence of SllNFEKL on the MHC-l le on splenocyte membranes (Figure 13A). interestingly, the on of the polyK chain in the sequence of the e significantly changed the proportion of cells stained.
In fact, 94.5% of the splenocytes pulsed with the N-terminus-extended peptide cross-presented SIINFEKL. in contrast, when the cytes were pulsed with the C-terminus-extended SllNFEKL-polyK, the stained population decreased to 27.1%. When pulsed with the negative control SIlNFEKL-AHX- polyK, only 1.36% of the cytes cross-presented the SIINFEKL peptide.
Based on these findings, we chose the lnus-extended version (polyK— SIINFEKL) for r studies.
Next, we investigated whether the adsorption of the modified SIINFEKL onto the viral capsid could affect its cross-presentation. As in the previous experiment, we incubated mouse Splenocytes with the peptide KL or polyK—SIINFEKL or with OVA-PeptiCRAd. We found that the N- terminus-extended polyK-SllNFEKL complexed with an OAd, forming PepthRAd, allowed for efficient MHC‘Lrestricted presentation of the SllNFEKL peptide (Figure 138).
PeptiCRAd shows unaltered infectivity and intact oncolytic activity compared with unmodified viruses.
OAds can selectively infect tumor cells and lyse them via the OAd replication cycle. Thus, we investigated whether coating the viruses with modified peptides would affect their biological properties. We chose to study a human colorectal adenocarcinoma cell line (CACO-2) expressing low levels of coxsackie and adenovirus receptor (CAR) and two human melanoma cell lines (SK-MEL-2 and A2058) expressing higher levels of CAR. An in vitro viability assay comparing OVA-PeptiCRAd with the unmodified virus Ad5D24 was first performed (Figure 14A), and the results showed no significant differences with regard to oncolytic activity. As expected, the most infectious condition (100 vp/cell) correlated with the lowest viability in all cell lines. In addition, we showed that the peptide SllNFEKL had no toxic effect on cells.
Next, we evaluated the ivity of PeptiCRAd by immunocytochemistry (lCC) assays using the same cell lines in vitro (Figure 148). Whereas we did not observe any difference in the SK-MEL-Z cell line, in the CACO—Z and A2058 cell lines, PeptiCRAd showed a significant increase 1) in infectivity compared with the naked adenovirus. This increase was likely due to the different charges of RAd and the naked adenovirus (36).
Studies of the anti-tumor efficacy and immunology of a PeptiCRAd cancer vaccine in a murine model of melanoma.
To thoroughly study the anti-tumor efficacy of PeptiCRAd and the anti-tumor immunity that it promotes, we first used a murine model of melanoma over-expressing chicken OVA (Bits-OVA) (35). Specifically, 816- OVA was implanted in the flanks of mice, after which the established tumors were d. The experiment was performed using an OAd bearing the D24 deletion in 5D24) (37) and then repeated with a CpG-rich irus (Ad5D24~CpG) (39) to further boost immunity (Figure 15). The study groups included mice treated with OVA-PeptiCRAd, with non-complexed Ad5024-CpG and SIINFEKL (Ad5D24-CpG+SllNFEKL), with OAd (Ad5D24—CpG) or peptide (SllNFEKL) alone or with saline solution (mock).
PeptiCRAd ent significantly reduced tumor growth compared with mock treatment or the mixture of OAd and SlINFEKL (P<0.01). At the end of the experiment, the average volume of the tumors in the OVA-PeptiCRAd- treated mice was lower than in all other groups :31.6 mm3 vs. 697.7:350 mm3 in mock, 255i61.5 mm3 in SllNFEKL, and 713.7i292.6 mm3 in Ad5D24-CpG, 48971732 mm3 in AdSDZ4-CpG+SIINFEKL; Figure 15A).
At two different time points (days 7 and 16 for the early and late time points, respectively), the mice were iced, and spleens, tumors and draining lymph nodes were collected for immunological analysis. This analysis revealed the presence of a large population of SllNFEKL-Specific CD8+ T cells (CD8*OVA+ T cells) in the al draining lymph nodes in the group of mice treated with PeptiCRAd (7.4% at day 7 and 3.2% at day 16). The same analysis showed no drastic difference in tumors at the early time point, whereas a substantial increase was observed at the late time point (0.23% in OVA-PeptiCRAd vs. 0.02% in mock, 0.03% in KL, 0.01% in Ad5D24— CpG and 0.02% in Ad5D24—CpG+SllNFEKL at day 16; Figure 158 and C).
We then studied the correlation between the sizes of the tumors and the population of OVA—specific T cells (CD8*OVA+ T cells) in the spleen, lymph nodes and tumors. We calculated the Pearson’s r value to estimate the nature of the ation (negative value, negative correlation; positive value, positive correlation) and observed a ve correlation between the tumor volume and the extent of the anti-OVA response (Figure 150), indicating that the groups of animals with smaller tumors corresponded to the groups of s with a more robust population of CD8“OVA+ T cells. ards, the r2 value was calculated for each set of samples to evaluate the strength of this correlation n, r2=0.5719; lymph nodes, r2=0.6385; tumors, r2=0.7445). stingly, in the correlation analyses, the PeptiCRAd group (red dots in Figure 15D) consistently showed the st tumor volume and the greatest immunological response.
Finally. to deepen our understanding of the mechanisms of PeptiCRAd, we ted the proportion of mature DCs (CD19'CD3' coiictcoashtg“ cells) presenting the SllNFEKL peptide on MHC-l in the spleens of the mice. At the late time point, the proportion of mature SllNFEKL- presenting DCs was significantly higher (P<0.05) in the mice treated with OVA- PeptiCRAd than in the mice treated with the non-complexed Ad5D24- CpG+SllNFEKL. When both time points are considered, PeptiCRAd was the only treatment that induced an increase in mature KL-presenting DCs, as shown by the 9.67-fold increase in the coachighovrt+ DC population (Figure 15E).
These results suggest that expansion of the mature and epitope- Specific DC pool could be the basis for the higher anti-tumor efficacy of PeptiCRAd.
Multiva/ent PeptiCRAd shows enhanced anti-tumor activity toward distant, untreated melanomas.
One of the main advantages of using tic vaccines is that the immune response elicited tates targeting not only the primary tumors but also disseminated metastasis. For this reason, we investigated the anti-tumor efficacy of PeptiCRAd toward ted lateral tumors in a murine model of melanoma. in the same set of experiments, we also studied whether targeting two tumor antigens (via multivalent PeptiCRAds), rather than a single one, would increase the l efficacy. Therefore, we chose two tumor- specific MHC-l-restricted es to coat the oncolytic virus Ad5D24-CpG: SVYDFFVWL (TRPuzigmgg; restricted to the murine MHC—l molecule H-ZKb) and KVPRNQDWL (human gp10025.33, or hgplOO; cted to the murine MHC-l molecule H—2Db (40)). For these experiments, we employed the highly aggressive melanoma 0, which expresses both tumor ns (41).
The peptides were modified with a poiyK chain at their N-terminus to favor their adsorption onto the viral capsid, as before for SllNFEKL.
We first implanted ‘lxiO5 Bi6~FlO cells into the right flank of C57BL/6 mice. After 10 days, treatments were initiated as follows: i) saline solution (mock), ii) naked oncolytic virus (AdSDZ4-CpG), and iii) double-coated TRP-Z-hgplOO-PeptiCRAd. The treatments were administered intratumorally every second day, as shown in the schematic in Figure 6A. Two days after the last round of injections, 3><105 lO cells were injected into the left flank of the mice, and the growth of melanomas followed. The mice treated with the double-coated PeptiCRAd showed significantly reduced tumor growth (P<0.001) compared with the control (at day ll; Figure 16A). Analysis of secondary and untreated tumors revealed an age of the double-coated PeptiCRAd over all other groups. in particular, at the end of the experiment, the secondary tumors in this group were significantly smaller compared with those in the controls receiving saline solution or only Ad5024-CpG (P<0.0i; Figure 163).
To better clarify the mechanisms underpinning these results, we performed a flow cytometry analysis to study the specific T-cell responses to both epitopes. In mice treated with TRP—Z—hgplOO PeptiCRAd, we observed a larger cumulative population of epitope-speclfic CD8+ T cells (Figure 16C) than in all other groups.
Taken together, these results demonstrate that the RAd ch is effective against a less immunogenic and more aggressive melanoma model. in addition, targeting le antigens results in a strong effect on both treated and untreated tumors. Hence, it is possible to te multivalent PeptiCRAds, and they can give us the possibility to target different tumor antigens hence ming the some immunological escape of the tumor.
RAd displays enhanced efficacy and anti-tumor immunity in humanized mice bearing human tumors. y, we wanted to assess the efficacy of PeptiCRAd in a model that could provide information on the feasibility of its translation to the clinical g. Therefore, we chose a more ticated humanized mouse model.
To this end, triple-knockout mice (NOD.Cg-Prkdcsc’d-IL2rg‘mW""JSzJ, or NSG) were first engrafted with the human melanoma cell line SK-MEL-Z. When the tumor reached a palpable size, partially matched human peripheral blood mononuclear cells (PBMCs) from a healthy donor were engrafted into the same mice. One day later, the mice were d with PeptiCRAd, uncoated virus or saline solution. For this experiment, we chose a peptide derived from melanoma-associated antigen A1 A196-1o4; ITK) and modified it to allow for interaction with the viral capsid -SLFRAVITK). In this experiment, as we were studying a completely human immune system, we selected an OAd expressing human GM-CSF, which we have previously shown to have enhanced activity in an immunocompetent system, including in cancer patients (8).
We found that MAGE-A1 PeptiCRAd showed increased efficacy compared with the control treatments, as shown by the rapid reduction in the tumor volume (Figure 17A and B). Finally, we investigated whether a stronger immunological response could explain the increased anti-tumor efficacy of PeptiCRAd in this model. To this end, we studied the presence of MAGE-A195- wit-specific CUES+ T cells by pentamer ng (Figure 17C), and we found the largest population of human MAGE-speclfic T cells (CD8*MAGE-A1*) in the spleens of mice treated with PeptiCRAd.
These data confirm our previous findings that PeptiCRAd stimulates the tumor~specific immune response by taking advantage of the natural immunogenicity of oncolytic viruses, hence improving the cy of cancer immunovirotherapy.
Analysis of MHC-i specific polypeptides on any disease and coating of the iral capsid and uses thereof Any MHC-l specific polypeptide(s) is(are) identified by comparing MHC-I-resticted polypeptides represented by DOS and infected disease cells of a subject. One or more polypeptides presented by both cell groups are selected for g an adenovirai vector.
Any adenovirai vector is selected and coated according to any method described in Method 2.
The coated s are used for treating the disease of a patient.
References 1. in, S (2012). Peptides in melanoma therapy. Curr Pharm Des 18: 820—831. 2. Lipscomb, MF and Masten, BJ. Dendritic Cells: Immune Regulators in Health and Disease. vphysiologyorg. 3. Degli-ESposti, MA and Smyth, MJ (2005). Close encounters of different kinds: dendritic cells and NK cells take centre stage. Nat Rev Immunol 5: 112-424. 4. Trinchieri, G, Aden, DP and Knowles, BB (1976). Cell-mediated cytotoxicity to SV40-specific tumour—associated antigens. Nature 261: 312— 314.
. Steinman, RM, Hawiger, D and Nussenzweig, MC (2003).
Tolerogenic dendritic cells. Annu. Rev. l. 21: 1. 6. Wongthida, P, Diaz, RM, Galivo, F, Kottke, T, Thompson, J, Melcher, A, et al. (2011). VSV oncolytic virotherapy in the 816 model depends upon intact MyD88 signaling. Mol Ther 19: 8. 7. Prestwich, RJ, Errington, F, Diaz, RM, Pandha, HS, Harrington, KJ, Melcher, AA, et al. (2009). The case of oncolytic viruses versus the immune system: waiting on the judgment of Solomon. Hum Gene Ther 20: 1119—1132. 8. Cerullo, V, n, S, Diaconu, I, Escutenaire, S, Arstila, PT, Ugolini, M, et al. . tic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res 70: 4297—4309. 9. Heise, C, Hermiston, T, Johnson, L, Brooks, G, Sampson— Johannes, A, Williams, A, et al. . An adenovirus E1A mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nat Med 6: 1134—1139.
. Heise, C, Sampson-Johannes, A, Williams, A, McCormick, F, Hoff, von, DD and Kim, DH (1997). ONYX-015, an E1B gene-attenuated adenovirus, causes tumor-specific cytolysis and antitumoral efficacy that can be augmented by standard chemotherapeutic agents. Nat Med 3: 639—645. 11. Nowak, AK, Lake, RA, Marzo, AL, Scott, B, Heath, WR, Collins, EJ, et al. (2003). Induction of tumor cell sis in vivo increases tumor antigen presentation, cross—priming rather than cross-tolerizmg host tumor-specn‘ic CD8 T cells. J lmmunol 170: 4905-4913. 12. Zitvogel, L, Apetoh, L, Ghiringhelli, F and Kroemer, G (2008). immunological aspects of cancer chemotherapy. Nat Rev lmmunol 8: 59-73. 13. van der Most, RG, Currie, AJ, Robinson, BWS and Lake, RA (2008). Decoding dangerous death: how cytotoxic chemotherapy invokes inflammation, immunity or nothing at all. Cell Death Differ 15: 13—20. 14. lstrail, S, Florea, L, Halldérsson, BV, Kohlbacher, O, Schwartz, RS, Yap, VB, et al. (2004). Comparative immunopeptidomics of humans and their pathogens. Proc Natl Acad Sci USA 101: 13268—13272.
. Cerullo, V, Pesonen, S, Diaconu, l, Escutenaire, S, Arstila, PT, Ugolini, M, et al. (2010). Oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral immunity in cancer patients. Cancer Res 70: 4297—4309. 16. Cerullo, V, , MP, Mane, V, ti-Pierri, N, Clarke, C, , TK, et al. (2007). Toll-like receptor 9 triggers an innate immune response to helper-dependent adenoviral vectors. Moi Ther 15: 378—385. 17. Suzuki, M, Cerullo, Bertin, T, Cela, Clarke, Guenther, M, et al. (2009). MyD88-dependent silencing of transgene expression during the innate and ve immune response to Helper-dependent adenovirus. Hum Gene Therdoi:10.1089/hum.2009.155. 18. Suzuki, M, Ceia, R, Bertin, TK, Sule, G, Cerullo, V, s, JR, et al. (2011). NODZ signaling contributes to the innate immune se against helper-dependent adenovirus vectors independently of MyDS8 in vivo. .
Hum Gene Ther 22: 10 1082. 19. Muruve, DA, li, V, Zaiss, AK, White, LR, Clark, SA, Ross, PJ, et al. . The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune se. Nature 452: 103—107.
. Tewalt, EF, Grant, JM, Granger, EL, Palmer, DC, Heuss, ND, Gregerson, DS, et al. (2009). Viral Sequestration of Antigen Subverts Cross Presentation to CD8+ T Cells. PLoS Pathog 5: e1000457. 21. Cerullo, V, Seiler, MP, Mane, VP, Brunetti-Pierri, N, , C, Bertin, TK, et al. (2007). Toll-like receptor 9 triggers an innate immune response to helper-dependent adenoviral vectors. Mol Ther 15: 378—385. 22. Suzuki, M, Cerullo, V, , TK, Cela, R, Clarke, C, Guenther, M, et al. . MyD88—dependent Silencing of transgene expressron during the innate and ve immune response to helper-dependent adenovirus.
Hum Gene Ther 21: 325—336. 23. , MP, Gottschaik, S, Cerulio, V, ake, M, Mane, VP, , C, et ai. (2007). Dendritic cell on after gene transfer with irus—caicium phosphate co-precipitates. Moi Ther 15: 386—392. 24. Suzuki, M, Cela, R, Bertin, TK, Suie, G, Ceruiio, V, Rodgers, JR, et al. (2011). NODZ Signaling Contributes to the Innate immune Response Against Helper—Dependent Adenovirus Vectors independently of MyD88 in Vivo. Hum Gene i:10.1089lhum.2011.002. 25. Fortier, M-H, Caron, E, Hardy, M-P, Voisin, G, Lemieux, S, Perreault, C, et ai. (2008). The MHC class l peptide repertoire is molded by the transcriptome. Journal of Experimental Medicine 205: 595—610. 26. Fasbender, A, Zabner, J, Chiiion, M, Moninger, TO, Puga, AP, Davidson, BL, et al. (1997). Complexes of adenovirus with poiycationic polymers and cationic lipids increase the efficiency of gene transfer in vitro and in vivo. J Biol Chem 272: 6479-6489. 27. Croyle, MA, Yu, QC and Wilson, JM (2000). Development of a rapid method for the PEGylation of adenoviruses with enhanced transduction and improved stability under harsh storage conditions. Hum Gene Ther 11: 1713—1722. 28. Wonganan, P and Croyle, MA . PEGylated Adenoviruses: From Mice to Monkeys. Viruses 2: 468—502. 29. , K, Ooboshi, H, Chu, Y, Fasbender, Al, Davidson, BL, Welsh, MJ, et al. Cationic Polymer and Lipids Enhance Adenovirus-Mediated Gene Transfer to Rabbit Carotid Artery. strokeahajournaisorg.
. Ceruiio, V, Diaconu, l, Romano, V, Hirvinen, M, Ugolini, M, Escutenaire, S, et al. (2012). An Oncoiytic irus Enhanced for Toll-like Receptor 9 Stimulation increases Antitumor immune Responses and Tumor Clearance. Moi Ther 20, 2076. 31. Ceruiio, V, Diaconu, i, , V, Hirvinen, M, Ugolini, M, Escutenaire, S, et ai. (2012). An Oncoiytic Adenovirus Enhanced for Toii-iike Receptor 9 Stimulation increases Antitumor immune Responses and Tumor Clearance. Moi Ther 20, 2076. 32. Dias, JD, Lilkanen, l, Guse, K, FoIOppe, J, cka, M, Diaconu, i, et al. (2010). ed chemotherapy for head and neck cancer with a chimeric oncoiytic adenovirus coding for bifunctional suicide n FCU1. Clin Cancer Res 16: 2540—2549. 33. Edukulia, R, Woller, N, Mundt, B, Knocke, S, Gurlevik, E, Saborowski, M, et al. (2009). Antitumoral immune response by tment and expansion of tic cells in tumors infected with telomerase-dependent oncolytic viruses. Cancer Res 69: 14484458. 34. R. B. Dell, 8. Holleran, R. Ramakrishnan, Sample size determination. ILAR journal / National Research Council, Institute of Laboratory Animal Resources 43, 207 (2002). 35. M. W. Moore, F. R. Carbone, M. J. Bevan, Introduction of e protein into the class i pathway of antigen processing and presentation. Cell 54, 777 (Sep 9, 1988). 36. A. Fasbender et al., Complexes of adenovirus with tionic polymers and cationic lipids increase the efficiency of gene er in vitro and in vivo. The Journal of biological chemistry 272, 6479 (Mar 7, 1997). 37. C. Heise et al,, An adenovirus E1A mutant that demonstrates potent and selective systemic anti-tumoral efficacy. Nature medicine 6. 1134 (Oct, 2000). 38. Y. Deng et al., Assembly of MHC class i molecules with biosynthesized endoplasmic reticulum—targeted peptides is cient in insect cells and can be enhanced by protease inhibitors. J l 161, 1677 (Aug , 1998). 39. V. Cerullo at a/., An oncolytic adenovirus enhanced for toll-iike or 9 stimulation increases antitumor immune responses and tumor clearance. Molecular therapy : the journal of the American Society of Gene Therapy 20, 2076 (Nov, 2012). 40. W.W. Overwin et al., gp100/pmel 17 is a murine tumor rejection antigen: induction of "self-reactive, tumoricidal T cells using ffinity, altered peptide ligand. The Journal of experimental ne 188, 277 (Jul 20, 1998) 41. W.W. Overwijk, N. P. Restifo, 816 as a mouse model for human melanoma. Current protocols in immunology / edited by John E. Coligan [at al.] Chapter 20, Unit 20 1 (May, 2001).

Claims (15)

1. An adenoviral vector comprising polypeptides that stimulate a peptide-specific anti-virus immune response in a subject, wherein the 5 polypeptides are attached onto the viral capsid and have not been genetically encoded by said adenoviral vector and further n the polypeptides attached onto the viral capsid are selected from the group ting of Major Histocompatibility Complex of class I (MHC-I)-specific polypeptides and Major Histocompatibility Complex of class II (MHC-II)-specific ptides, and are 10 disease specific polypeptides.
2. The adenoviral vector of claim 1, wherein the subject is a human or an animal.
3. The adenoviral vector of claim 1 or 2, wherein the vector is formulated for via intratumoral, intra-arterial, intravenous, intrapleural, 15 esicular, intracavitary or peritoneal injection, or oral administration.
4. The adenoviral vector of any one of claims 1-3, wherein the polypeptides have been attached ntly or non-covalently onto the viral
5. The adenoviral vector of any one of claims 1-4, wherein the 20 polypeptides are attached to the viral capsid by linking sine-modified polypeptides to the adenoviral capsid covalently or non-covalently.
6. The adenoviral vector of any one of claims 1-5, wherein the polypeptides have been attached to the capsid by electrostatic, ide or amide bond e. 25
7. The adenoviral vector of any one of claims 1-6, wherein the polypeptides have been attached to the capsid and are provided with the vector in a single nanoparticle.
8. The adenoviral vector of any one of claims 1-7, wherein the polypeptides attached onto the viral capsid are all the same polypeptides or 30 different polypeptides ed from two or more types of different polypeptides.
9. The adenoviral vector of any one of claims 1-8, wherein the polypeptides attached onto the viral capsid are at the same time both MHC-I- specific and disease specific polypeptides, or, at the same time MHC-I-specific, DC specific and disease specific polypeptides. 35
10. The adenoviral vector of any one of claims 1-9, wherein the serotype of the adenoviral vector backbone is selected from serotype 3 or 5.
11. The adenoviral vector of any one of claims 1-10, wherein the adenoviral vector is an tic adenoviral vector.
12. The adenoviral vector of any one of claims 1-11, wherein the adenoviral vector comprises the 24bp deletion or E1 gene deletion or the vector 5 is a Helper-dependent vector.
13. The iral vector of any one of claims 1-12, wherein the adenoviral vector comprises one or more transgenes.
14. The adenoviral vector of any one of claims 1-13, wherein the adenoviral vector comprises a capsid modification. 10
15. The iral vector of any one of claims 1-14, n the adenoviral vector is Ad
NZ764519A 2014-05-19 2015-05-18 Coated adenoviruses for immunotherapy NZ764519B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FI20145449 2014-05-19
FI20145449 2014-05-19
NZ726112A NZ726112A (en) 2014-05-19 2015-05-18 Coated adenoviruses for immunotherapy

Publications (2)

Publication Number Publication Date
NZ764519A NZ764519A (en) 2021-08-27
NZ764519B2 true NZ764519B2 (en) 2021-11-30

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020203816B2 (en) Coated Oncolytic Adenoviruses for Cancer Vaccines
ES2938749T3 (en) cancer therapy
CN112912507A (en) Alphavirus novel antigen vector and interferon inhibitor
US20220202932A1 (en) Compositions and methods for organ-protective expression and modulation of coding ribonucleic acids
TW202000906A (en) Immune checkpoint inhibitor co-expression vectors
Zhao et al. Photo‐enhanced CRISPR/Cas9 system enables robust pd‐l1 gene disruption in cancer cells and cancer stem‐like cells for efficient cancer immunotherapy
Lauterbach et al. Development and characterization of a novel non-lytic cancer immunotherapy using a recombinant arenavirus vector platform
US20230062122A1 (en) Modified adenoviruses for infectious disease vaccine development
Ye et al. Folate‐conjugated herpes simplex virus for retargeting to tumor cells
NZ764519B2 (en) Coated adenoviruses for immunotherapy
WO2021036245A1 (en) Chimeric antigen receptor t cell carrying safety switch and targeting egfrvⅲ, and preparation method therefor and use thereof
WO2021036247A1 (en) Chimeric antigen receptor t cell targeting her2 and interfering with il-6 expression, preparation method therefor and use thereof
BR112016026745B1 (en) USE OF AN ONCOLYTIC ADENOVIRAL VECTOR, ONCOLYTIC ADENOVIRAL VECTOR, METHOD FOR MODIFICATION OF THE CAPSID OF AN ADENOVIRUS, USE OF POLYPEPTIDES MODIFIED BY POLYLYSINE, AND PHARMACEUTICAL COMPOSITION
WO2021036244A1 (en) Chimeric antigen receptor t cell carrying safety switch and targeting her2, preparation method therefor, and application thereof
US11235072B2 (en) Adenovirus complex for gene delivery and gene iherapy
Gonzales Development of therapeutic vaccine strategies and pre-clinical animal tumor models for head and neck cancers
Rojas Expósito Blood barriers for oncolytic adenovirus efficacy: study of binding to erythrocytes via CAR and albumin‐mediated evasion of neutralizing antibodies
Recagni SELF-AMPLIFYING RNA VECTORS ENCODING FOR SURVIVIN AS ANTI-TUMORAL VACCINE CANDIDATES
Toyoda et al. Oncolytic Poliovirus Therapy in a Mouse Model of Neuroblastoma: Preclinical Data Analysis and Future Studies