WO2024059629A1 - Combinaison de thérapie génique utilisant l'il-12 et d'inhibiteurs de point de contrôle immunitaire pour le traitement du cancer - Google Patents

Combinaison de thérapie génique utilisant l'il-12 et d'inhibiteurs de point de contrôle immunitaire pour le traitement du cancer Download PDF

Info

Publication number
WO2024059629A1
WO2024059629A1 PCT/US2023/074062 US2023074062W WO2024059629A1 WO 2024059629 A1 WO2024059629 A1 WO 2024059629A1 US 2023074062 W US2023074062 W US 2023074062W WO 2024059629 A1 WO2024059629 A1 WO 2024059629A1
Authority
WO
WIPO (PCT)
Prior art keywords
aspects
administered
dose
immune checkpoint
combination therapy
Prior art date
Application number
PCT/US2023/074062
Other languages
English (en)
Inventor
Khursheed Anwer
Nicholas Borys
Original Assignee
Imunon, Inc.
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 Imunon, Inc. filed Critical Imunon, Inc.
Publication of WO2024059629A1 publication Critical patent/WO2024059629A1/fr

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0041Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • 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
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/50Vector systems having a special element relevant for transcription regulating RNA stability, not being an intron, e.g. poly A signal

Definitions

  • the present disclosure relates to the fields of cancer therapy, gene therapy, and immunology.
  • IL-12 is one of the most active cytokines for stimulating an immune response against cancer.
  • the GEN-1 is an IL-12 DNA plasmid vector formulated using a lipopolymeric delivery system. GEN-1 can be delivered locally (e.g., intraperitoneally), offering the potential for cytokines to be expressed specifically in the tumor micro-environment with the goal of achieving increased efficacy while minimizing potential systemic toxicity.
  • immunocytokine therapies are designed to elicit tumor killing by enhancing the immune system against cancer cells.
  • GEN-1 reduces the toxicity issues associated with IL-12. Its nanoparticle profile allows for cell transfection followed by persistent, local secretion of IL-12 at therapeutic levels, while avoiding the toxicities associated with recombinant IL-12.
  • a nucleic acid vector e.g., a plasmid
  • a nucleic acid vector comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and
  • an immune checkpoint inhibitor e.g., an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and (ii) an immune checkpoint inhibitor.
  • IL- 12 interleukin- 12
  • Certain aspects of the disclosure are related to a method of treating a subject suffering from cancer comprising administering to the subject a combination therapy comprising: (i) a nucleic acid vector (e.g., a plasmid) comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and
  • a nucleic acid vector e.g., a plasmid
  • IL- 12 interleukin- 12
  • a lipopolymer e.g., a nanoparticle
  • the polynucleotide encodes human IL-12.
  • nucleic acid vector (e.g., a plasmid) comprises a promoter operably linked to a nucleic acid encoding a p35 subunit of IL- 12 and a promoter operably linked to a nucleic acid encoding a p40 subunit of IL12.
  • the promoter is a CMV promoter.
  • nucleic acid vector e.g., a plasmid
  • the lipopolymer comprises a polyethyleneimine (PEI) covalently linked independently to cholesterol and polyethylene glycol (PEG) groups (e.g., the lipopolymer of FIG. 2).
  • PEI polyethyleneimine
  • PEG polyethylene glycol
  • the combination further comprises an anticancer agent.
  • the anticancer agent is a chemotherapeutic agent.
  • the chemotherapeutic agent is selected from a group consisting of topoisomerase inhibitors (e.g., irinotecan, topotecan, doxorubicin, epirubicin, idarubicin), anti -microtubule agents (e.g., paclitaxel, docetaxel), alkylating agents (e.g., cyclophosphamide, dacarbizine), platinum-based drugs (cisplatin, carboplatin, oxaliplatin), anti-metabolites (e.g., gemcitabine, methotrexate, 5 -fluorouracil), or combinations thereof.
  • topoisomerase inhibitors e.g., irinotecan, topotecan, doxorubicin, epirubicin, idarubicin
  • anti -microtubule agents e.g., paclitaxel, docetaxel
  • alkylating agents e.g., cyclophos
  • the chemotherapeutic agent is selected from the group consisting of doxorubicin, paclitaxel, carboplatin, docetaxel, nab-paclitaxel, olaparib, and any combination thereof.
  • the anticancer agent is doxorubicin.
  • the anticancer agent is paclitaxel.
  • the anticancer agent is carboplatin.
  • the anticancer agent is docetaxel.
  • the anticancer agent is nab-paclitaxel.
  • the anticancer agent is olaparib.
  • the immune checkpoint inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 (and its ligands PD- L1 and PD-L2), and/or LAG-3.
  • the immune checkpoint inhibitor is an antibody.
  • the immune checkpoint inhibitor is a small molecule inhibitor.
  • the immune checkpoint inhibitor is a PD-1 antagonist selected from the group consisting of nivolumab, pembrolizumab, dostarlimab, and cemiplimab.
  • the immune checkpoint inhibitor is a PD-1 antagonist, wherein the PD-1 antagonist is nivolumab.
  • the immune checkpoint inhibitor is a PD-L1 antagonist selected from the group consisting of atezolizumab, durvalumab, and avelumab.
  • the immune checkpoint inhibitor is a CTLA-4 antagonist is ipilimumab.
  • the immune checkpoint inhibitor is a LAG-3 antagonist, wherein the LAG-3 antagonist is relatlimab.
  • the combination therapy further comprises (c) a second immune checkpoint inhibitor.
  • the second immune checkpoint inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 (and its ligands PD-L1 and PD-L2), and/or LAG-3.
  • second the immune checkpoint inhibitor is an antibody.
  • the second immune checkpoint inhibitor is a small molecule inhibitor.
  • the second immune checkpoint inhibitor is a PD-1 antagonist selected from the group consisting of nivolumab, pembrolizumab, dostarlimab, and cemiplimab.
  • the second immune checkpoint inhibitor is nivolumab.
  • the second immune checkpoint inhibitor is a PD-L1 antagonist selected from the group consisting of atezolizumab, durvalumab, and avelumab.
  • the second immune checkpoint inhibitor is a CTLA-4 antagonist is ipilimumab.
  • the second immune checkpoint inhibitor is a LAG-3 antagonist, wherein the LAG-3 antagonist is relatlimab.
  • the method further comprises a surgery to remove all or part of a tissue or tumor (e.g., interval cytoreductive surgery) in the subject.
  • a surgery to remove all or part of a tissue or tumor e.g., interval cytoreductive surgery
  • the nucleic acid vector formulated with the lipopolymer is administered intratum orally or intraperitoneally.
  • the nucleic acid vector formulated with the lipopolymer is administered intravenously.
  • the immune checkpoint inhibitor is administered intratumorally, intraperitoneally, intravenously, intravesicularly, or any combination thereof.
  • the immune checkpoint inhibitor is administered intratumorally or intraperitoneally.
  • the immune checkpoint inhibitor is administered intravenously.
  • the immune checkpoint inhibitor is administered intravesicularly.
  • nucleic acid vector formulated with the lipopolymer is administered prior to, concurrently with, or after the immune checkpoint inhibitor
  • the nucleic acid vector formulated with the lipopolymer is administered prior to, concurrently with, or after the anticancer agent.
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), and followed by administration of the immune checkpoint inhibitor (e.g., third).
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), followed by the immune checkpoint inhibitor (e.g. third), and followed by a surgery to remove all or part of a tissue or tumor (e.g., interval cytoreductive surgery) (e.g., fourth).
  • a surgery to remove all or part of a tissue or tumor is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer is administered (e.g., second), and followed by administration of an immune checkpoint inhibitor (e.g., third, fourth, etc. depending on how many immune checkpoint inhibitors are administered).
  • an immune checkpoint inhibitor e.g., third, fourth, etc. depending on how many immune checkpoint inhibitors are administered.
  • an anticancer agent is administered, followed by the administration of a nucleic acid vector comprising a polynucleotide encoding IL-12 (e.g., a DNA plasmid), followed by the immune checkpoint inhibitor, followed by an interval cytoreductive surgery.
  • a nucleic acid vector comprising a polynucleotide encoding IL-12 (e.g., a DNA plasmid)
  • the immune checkpoint inhibitor followed by an interval cytoreductive surgery.
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of 25 - 350 mg/m 2 , optionally followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the interleukin- 12 (IL-12) formulated with a lipopolymer is administered at a dose of about 35 mg/m 2 to about 80 mg/m 2 .
  • the cancer is selected from a group consisting of ovarian cancer, fallopian tube cancer, primary peritoneal cancer, cervical cancer, breast cancer, prostate cancer, colorectal cancer, bladder cancer, brain cancer (e.g., glioblastoma), lung cancer, and any combination thereof, and metastasis of any of the cancers.
  • the cancer is selected from a group consisting of ovarian cancer, fallopian tube cancer, primary peritoneal cancer, and any combination thereof.
  • the subject is a human.
  • the lipopolymer e.g., nanoparticle
  • the lipopolymer is administered at a dose of about 60 mg/m 2 .
  • the immune checkpoint inhibitor is nivolumab, and wherein nivolumab is administered at about 240 mg.
  • nivolumab and the lipopolymer are administered every 1-4 weeks (e.g., every 2 weeks) for the duration of treatment.
  • the second inhibitor is ipilimumab, and wherein ipilimumab is administered at about 1 mg/kg.
  • ipilimumab is administered every 2-8 weeks (e.g., every 6 weeks) for the duration of treatment.
  • the nucleic acid vector is a plasmid.
  • the lipopolymer is a nanoparticle.
  • FIG. 1 shows an exemplary human IL- 12 (hIL-12) expression plasmid.
  • FIG. 2 shows the PEG-PEI-Cholesterol structure.
  • an effective amount or “pharmaceutically effective amount” or “therapeutically effective amount” as used herein refers to the amount or quantity of a drug or pharmaceutically active substance which is sufficient to elicit the required or desired therapeutic response, or in other words, the amount which is sufficient to elicit an appreciable biological response when administered to a patient.
  • Transfecting shall mean transport of nucleic acids from the environment external to a cell to the internal cellular environment, with particular reference to the cytoplasm and/or cell nucleus.
  • nucleic acids may be delivered to cells either after being encapsulated within or adhering to one or more cationic polymer/nucleic acid complexes or being entrained therewith.
  • Particular transfecting instances deliver a nucleic acid to a cell nucleus.
  • Nucleic acids include DNA and RNA as well as synthetic congeners thereof.
  • nucleic acids include missense, antisense, nonsense, as well as protein producing nucleotides, on and off and rate regulatory nucleotides that control protein, peptide, and nucleic acid production.
  • they can be genomic DNA, cDNA, mRNA, tRNA, rRNA, hybrid sequences or synthetic or semi -synthetic sequences, and of natural or artificial origin.
  • the nucleic acid can be variable in size, ranging from oligonucleotides to chromosomes.
  • These nucleic acids may be of human, animal, vegetable, bacterial, viral, or synthetic origin. They may be obtained by any technique known to a person skilled in the art.
  • the term "pharmaceutical agent” or “drug” or any other similar term means any chemical or biological material or compound suitable for administration by the methods previously known in the art and/or by the methods taught in the present disclosure, which induce a desired biological or pharmacological effect, which may include but are not limited to (1) having a prophylactic effect on the organism and preventing an undesired biological effect such as preventing an infection, (2) alleviating a condition caused by a disease, for example, alleviating pain or inflammation caused as a result of disease, and/or (3) either alleviating, reducing, or completely eliminating a disease from the organism.
  • the effect may be local, such as providing for a local anesthetic effect, or it may be systemic.
  • biocompatible or “biodegradation” is defined as the conversion of materials into less complex intermediates or end products by solubilization hydrolysis, or by the action of biologically formed entities which can be enzymes and other products of the organism.
  • an effective amount means the amount of a nucleic acid or a bioactive agent that is sufficient to provide the desired local or systemic effect and performance at a reasonable risk/benefit ratio as would attend any medical treatment.
  • peptide means peptides of any length and includes proteins.
  • polypeptide and oligopeptide are used herein without any particular intended size limitation, unless a particular size is otherwise stated.
  • a "derivative" of a carbohydrate includes, for example, an acid form of a sugar, e.g. glucuronic acid; an amine of a sugar, e.g. galactosamine; a phosphate of a sugar, e.g. mannose-6-phosphate and the like.
  • administering means delivering the composition to the individual being treated such that the composition is capable of being circulated systemically where the composition binds to a target cell and is taken up by endocytosis.
  • the composition is preferably administered systemically to the individual, typically by subcutaneous, intramuscular, transdermal, intravenous, or intraperitoneal routes.
  • injectables for such use can be prepared in conventional forms, either as a liquid solution or suspension, or in a solid form that is suitable for preparation as a solution or suspension in a liquid prior to injection, or as an emulsion.
  • Suitable excipients that can be used for administration include, for example, water, saline, dextrose, glycerol, ethanol, and the like; and if desired, minor amounts of auxiliary substances such as wetting or emulsifying agents, buffers, and the like.
  • "efficacy” and similar terms means disappearance of tumor or shrinkage of tumor in size or reduction in tumor density or increase in lymphocyte count or increase in neutrophil count or improvement in survival, or all of the above.
  • toxicity is defined as any treatment related adverse effects on clinical observation including but not limited to abnormal hematology or serum chemistry results or organ toxicity.
  • promoter/regulatory sequence refers to a nucleic acid sequence required to express a gene product operably linked to a promoter/regulatory sequence.
  • constitutive refers to a nucleotide sequence that, when operably linked to a polynucleotide encoding or specifying a gene product, results in the production of a gene product in the cell under most or all physiological conditions of the cell.
  • inducible promoter means that when operably linked to a polynucleotide encoding a specified gene product, it basically results in the production of a gene in the cell only when the inducer corresponding to the promoter is present in the cell the nucleotide sequence of the product.
  • the term "expression” refers to a process by which a gene produces a biochemical, for example, a polypeptide.
  • the process includes any manifestation of the functional presence of the gene within the cell including, without limitation, gene knockdown as well as both transient expression and stable expression. It includes without limitation transcription of the gene into messenger RNA (mRNA), and the translation of such mRNA into polypeptide(s). Expression of a gene produces a "gene product.”
  • mRNA messenger RNA
  • expression of a gene produces a "gene product.”
  • a gene product can be either a nucleic acid, e.g., a messenger RNA produced by transcription of a gene, or a polypeptide which is translated from a transcript.
  • Gene products described herein further include nucleic acids with post transcriptional modifications, e.g., polyadenylation, or polypeptides with post translational modifications, e.g., methylation, glycosylation, the addition of lipids, association with other protein subunits, proteolytic cleavage, and the like.
  • expression vector refers to a vector comprising a recombinant polynucleotide comprising an expression control sequence operably linked to the nucleotide sequence to be expressed.
  • the expression vector contains sufficient cis-acting elements for expression; other elements for expression can be provided by the host cell or in an in vitro expression system.
  • Expression vectors include expression vectors known in the art, including cosmids, plasmids (for example, naked or contained in liposomes), and viruses incorporating recombinant polynucleotides (for example, lentivirus, retrovirus, adenovirus, and adeno-associated virus).
  • operably linked refers to a functional linkage between a regulatory sequence and a heterologous nucleic acid sequence, which results in the expression of the latter.
  • first nucleic acid sequence and the second nucleic acid sequence are arranged in a functional relationship, the first nucleic acid sequence and the second nucleic acid sequence are operably linked.
  • the promoter affects the transcription or expression of a coding sequence, the promoter is operably linked to the coding sequence.
  • the operably linked DNA sequences may be adjacent to each other, and for example, in the case where two protein coding regions need to be linked, the DNA sequences are in the same reading frame.
  • transfer vector refers to a composition containing an isolated nucleic acid and a substance that can be used to deliver the isolated nucleic acid to the inside of a cell.
  • Many vectors are known in the art, including but not limited to linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses.
  • transfer vector should also be interpreted to further include nonplasmid and non-viral compounds that facilitate the transfer of nucleic acids into cells, such as polylysine compounds, liposomes, and the like.
  • the term "host cell” can be any type of cell, e.g., a primary cell, a cell in culture, or a cell from a cell line.
  • the term “host cell” refers to a cell transfected with a nucleic acid molecule and the progeny or potential progeny of such a cell. Progeny of such a cell may not be identical to the parent cell transfected with the nucleic acid molecule, e.g., due to mutations or environmental influences that may occur in succeeding generations or integration of the nucleic acid molecule into the host cell genome.
  • Percent (%) amino acid sequence identity with respect to a polypeptide sequence as set forth herein is defined as the percentage of amino acid residues in a candidate sequence of interest to be compared that are identical with the amino acid residues in a particular polypeptide sequence as set forth herein (e.g. a particular polypeptide sequence characterized by a sequence identifier in the sequence listings), after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity.
  • a sequence alignment performed for determining percent amino acid sequence identity can be carried out according to procedures known in the art, as described for example in EP 1 241 179 Bl, which is incorporated herewith by reference, including in particular page 9, line 35 to page 10, line 40 with the definitions used therein and Table 1 regarding possible conservative substitutions.
  • a skilled person can use publicly available computer software.
  • Computer program methods for determining sequence identity include, but are not limited to BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software.
  • the software alignment program used can be BLAST.
  • a skilled person can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences subjected to comparison.
  • the % identity values can be generated using the WU-BL AST-2 computer program (Altschul et al., 1996, Methods in Enzymology 266:460-480, which is incorporated herewith by reference).
  • the following parameters are used, when carrying out the WU-BLAST-2 computer program: Most of the WU-BLAST-2 search parameters are set to the default values.
  • the HSP S and HSP S2 parameters which are dynamic values used by BLAST-2, are established by the program itself depending upon the composition of the sequence of interest and composition of the database against which the sequence is being searched.
  • a % sequence identity value can be determined by dividing (a) the number of matching identical amino acid residues between a particular amino acid sequence as set forth herein which is subjected to comparison (e.g. a particular polypeptide sequence characterized by a sequence identifier in the sequence listings) and the candidate amino acid sequence of interest to be compared, for example the number of matching identical amino acid residues as determined by WU-BLAST-2, by (b) the total number of amino acid residues of the polypeptide sequence as set forth herein which is subjected to comparison (e.g. a particular polypeptide sequence characterized by a SEQ. ID. NO. in the sequence listings).
  • Percent (%) nucleic acid sequence identity with respect to a nucleic acid sequence as set forth herein is defined as the percentage of nucleotides in a candidate sequence of interest to be compared that are identical with the nucleotides in a particular nucleic acid sequence as set forth herein (e.g. a particular polypeptide sequence characterized by a sequence identifier in the sequence listings), after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity.
  • the term “homology” or “identity” refers to the identity of subunit sequence between two polymer molecules, for example, between two nucleic acid molecules, such as two DNA molecules or two RNA molecules, or between two polypeptide molecules.
  • subunit positions in two molecules are occupied by the same monomer subunit; for example, if the position of each of two DNA molecules is occupied by adenine, they are homologous or identical at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions; for example, if half of the positions in the two sequences (for example, 5 positions in a polymer of 10 subunits in length) are homologous, the two sequences are 50% homologous; if 90% of the positions (for example, 9 out of 10) are matched or homologous, then the two sequences are 90% homologous.
  • identity percent refers to two or more sequences that are the same.
  • identity percent refers to two or more sequences that are the same.
  • sequence comparison algorithms e.g., 60% identity, optionally 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity over a specified region, or if not specified, over the entire sequence), then the two sequences are "substantially the same".
  • the identity exists over a region of at least about 50 nucleotides (or 10 amino acids) in length, or more preferably over a region of 100 to 500 or 1000 or more nucleotides in length (Or 20, 50, 200 or more amino acids).
  • a sequence serves as a reference sequence against which the test sequence is compared.
  • a sequence comparison algorithm is used, a test sequence and a reference sequence are input into a computer, and the sub-sequence coordinates and the sequence algorithm program parameters are specified, if necessary. Default program parameters can be used, or alternative parameters can be specified.
  • the sequence comparison algorithm calculates the percent sequence identity of the test sequence relative to the reference sequence based on the program parameters. Methods of sequence alignment for comparison are well known in the art as disclosed above.
  • Coding sequence or a sequence "encoding" a particular molecule (e.g., a therapeutic molecule) is a nucleic acid that is transcribed (in the case of DNA) or translated (in the case of mRNA) into polypeptide, in vitro or in vivo, when operably linked to an appropriate regulatory sequence, such as a promoter.
  • the boundaries of the coding sequence are determined by a start codon at the 5' (amino) terminus and a translation stop codon at the 3' (carboxy) terminus.
  • a "stop codon” (TAG, TGA, or TAA) is not translated into an amino acid, it can be considered to be part of a coding region, but any flanking sequences, for example promoters, ribosome binding sites, transcriptional terminators, introns, and the like, are not part of a coding region.
  • a coding sequence can include, but is not limited to, cDNA from prokaryotic or eukaryotic mRNA, genomic DNA sequences from prokaryotic or eukaryotic DNA, and synthetic DNA sequences.
  • a transcription termination sequence will usually be located 3' to the coding sequence.
  • nucleic acid sequences e.g., coding sequences, regulatory elements (e.g., promoters, enhancers, silencers, termination sequences), linkers (e.g., spacers, internal ribosome entry sites, cleavage sites) derived from a variety of sources, inserting nucleic acid sequences from a variety of sources in appropriate vectors (e.g., delivery vectors, expression vectors, integrating vectors), modifying or altering nucleotide sequences (e.g., by mutagenesis, insertion of modified nucleotides, 5'-capping, polyadenylation), synthesizing artificial nucleotide sequence.
  • nucleic acid sequences e.g., coding sequences, regulatory elements (e.g., promoters, enhancers, silencers, termination sequences), linkers (e.g., spacers, internal ribosome entry sites, cleavage sites) derived from a variety of sources, inserting nucleic
  • the term “recombinant”, refers to any nucleic acid (e.g., DNA, or RNA), peptide (e.g., oligopeptide, polypeptide, or protein), cell, or organism, which is made by combining genetic material from two or more different sources.
  • the recombinant nucleic acid, peptide, cell or organism comprises a portion of the genetic material from at least one source.
  • "recombinant DNA” molecules can include DNA molecules derived from one organism and inserted in a host organism to produce new genetic combinations.
  • recombinant RNA molecule
  • recombinant mRNA molecules can include RNA molecules derived from one organism and inserted in a host organism to produce the expression of a desired genetic product in the host organism.
  • recombinant peptide molecules can include amino acid molecules derived from an organism or cell, which are expressed from recombinant nucleic acid molecules.
  • isolated means changed or removed from the natural state.
  • a nucleic acid or peptide naturally present in a living animal is not “isolated”, but the same nucleic acid or peptide that is partially or completely separated from a substance co-existing in its natural state is “isolated.”
  • the isolated nucleic acid or protein may exist in a substantially purified form or may exist in a non-natural environment such as a host cell.
  • tumor refers to any mass of tissue that results from excessive cell growth or proliferation, either benign (non-cancerous) or malignant (cancerous), including pre-cancerous lesions.
  • primary tumor refers to the original, or first, tumor formed in the subject's body.
  • metalastasis refers to cancer (e.g., a tumor) formed by cancer cells derived from a primary cancer (e.g., tumor) that spread to further locations or areas of the body.
  • a primary cancer e.g., tumor
  • the term “specifically binds” refers to an antigen binding molecule that recognizes and binds a protein of a binding partner (such as a tumor antigen) present in a sample, but the antigen binding molecule does not substantially recognize or bind to other molecules in the sample.
  • tumor heterogeneity means that, after multiple divisions and proliferation during the growth of a tumor, daughter cells of the tumor its show molecular biological or genetic changes, so that there are differences in the growth rate, invasion ability, and drug sensitivity, prognosis and other aspects of the tumor. It is one of the characteristics of malignant tumors.
  • the term "cancer” refers to a broad group of various diseases characterized by the uncontrolled growth of abnormal cells (e.g., malignant cells) in the body. Unregulated cell division and growth results in the formation of malignant tumors that invade neighboring tissues through local spread and can also metastasize to distant parts of the body through the lymphatic system or bloodstream.
  • the methods of the present disclosure can be used to reduce the size of a primary tumor or a metastatic tumor, or treat a primary tumor or a metastatic tumor.
  • the conditions that can be treated or prevented by the method of the present disclosure include, for example, various neoplasms, including benign or malignant tumors, various hyperplasias, and the like.
  • the method of the present disclosure can achieve the inhibition and/or reversal of the undesirable hyperproliferative cell growth involved in such conditions.
  • the cancer can be ovarian cancer.
  • ovarian cancer refers to cancers arising in, or involving, the ovaries, e.g. in the epithelium of the ovaries.
  • cancer or “tumor” refers to an uncontrolled growth of cells which interferes with the normal functioning of the bodily organs and systems.
  • a subj ect that has a cancer or a tumor is a subj ect having obj ectively measurable cancer cells present in the subject's body. Included in this definition are benign and malignant cancers, as well as dormant tumors or micrometastases.
  • cytoreductive surgery refers to surgical removal of at least part of the ovarian cancer tissue from a subject. Cytoreductive surgery can remove varying amounts of tumor tissue from a subject, depending upon the location and character of the tumor tissue, the health of the subject, and complicating factors which one of skill in the art can assess. In some embodiments, cytoreductive surgery can remove at least 10% of the tumor tissue, e.g. 10% or more, 20% or more, 30% or more, 40% or more, 50% or more, 60% or more, 70% or more, 80% or more, 90% or more, or 95% or more of the tumor tissue present in the subject.
  • transfected or “transformed” or “transduced” refers to the process by which exogenous nucleic acid is transferred or introduced into a host cell.
  • a “transfected” or “transformed” or “transduced” cell is a cell that has been transfected, transformed or transduced with exogenous nucleic acid.
  • the cells include primary cell of a subject and progenies thereof.
  • refractory refers to a disease, such as cancer, which does not respond to treatment.
  • a refractory cancer may be resistant to treatment before or at the beginning of the treatment.
  • a refractory cancer may become resistant during treatment.
  • Refractory cancers are also called resistant cancers.
  • refractory or recurrent malignant tumors can use the treatment methods disclosed herein.
  • relapsed refers to the return of the signs and symptoms of a disease (e.g. cancer) or the return of a disease such as cancer during a period of improvement, for example, after a therapy, such as a previous treatment of cancer therapy.
  • the term "combination therapy” means a therapy that includes more than one treatment (e.g., active agent or procedure).
  • the combination therapy herein comprises at least a gene therapy, an anticancer agent, and at least one immune checkpoint inhibitor, which can be administered together or separately.
  • compositions of the combination therapy are formulated together in a single composition or as separate compositions.
  • the terms “treat,” “treated,” and “treating” mean both therapeutic and prophylactic treatment or preventative measures wherein the object is to reverse, alleviate, ameliorate, lessen, inhibit, slow down progression, development, severity or recurrence of an undesired symptom, complication, condition, biochemical indicia of a disorder, or disease, or obtain beneficial or desired clinical results.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease.
  • treatment includes eliciting a clinically significant response without excessive levels of side effects.
  • treatment includes prolonging survival as compared to expected survival if not receiving treatment.
  • the term “amelioration” or “ameliorating” refers to a lessening of severity of at least one indicator of a condition or disease.
  • the term “preventing” or “prevention” refers to delaying or forestalling the onset, development or progression of a condition or disease for a period of time, including weeks, months, or years.
  • the term “prophylactic” e.g., “prophylactic agent”, “prophylactic treatment”, “prophylactically effective amount”
  • the terms “individual” and “subject” have the same meaning herein, and can be a human and animal from other species.
  • the terms “subject” and “patient” are used interchangeably.
  • the subject can be an animal.
  • the subject is a mammal such as a non-human animal (e.g., cow, pig, horse, cat, dog, rat, mouse, monkey or other primate, etc.).
  • the subject is a human.
  • the patient is a subject who has a disease, disorder, or condition, or is at risk of suffering from a disease, disorder, or condition, or is otherwise in need of the compositions and methods provided herein.
  • the terms “therapeutically effective amount”, “therapeutically effective”, “effective amount” or “in an effective amount” are used interchangeably herein and refer to the amount of a compound, preparation, substance or composition that is effective to achieve a specific biological result as described herein, such as but not limited to treating or reducing the growth of a cancer or tumor.
  • therapeutically effective amount refers to the amount of a compound, preparation, substance or composition that is effective to achieve a specific biological result as described herein, such as but not limited to treating or reducing the growth of a cancer or tumor.
  • anti-tumor effective amount anti-tumor effective amount
  • “tumor-suppressing effective amount” or “therapeutically effective amount” the precise number of immune effector cells and therapeutic agents of the present disclosure to be administered can be determined by a physician in consideration of the individual's age, weight, tumor size, degree of infection or metastasis, and the condition of a patient (subject).
  • An effective amount of immune effector cells refers to, but is not limited to, the number of immune effector cells which can increase, enhance or prolong the anti-tumor activity of immune effector cells; increase the number of anti-tumor immune effector cells or activated immune effector cells; promote tumor regression, tumor shrinkage and/or tumor necrosis.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, formulations, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • excipient refers to any substance, not itself a therapeutic agent, which can be used in a composition for delivery of an active therapeutic agent to a subject or combined with an active therapeutic agent (e.g., to create a pharmaceutical composition) to improve its handling or storage properties or to permit or facilitate formation of a dose unit of the composition.
  • Excipients include, but are not limited to, solvents, penetration enhancers, wetting agents, antioxidants, lubricants, emollients, substances added to improve appearance or texture of the composition and substances used to form hydrogels. Any such excipients can be used in any dosage forms according to the present disclosure.
  • excipients are not meant to be exhaustive but merely illustrative as a person of ordinary skill in the art would recognize that additional types and combinations of excipients could be used to achieve the desired goals for delivery of a drug.
  • the excipient can be an inert substance, an inactive substance, and/or a not medicinally active substance.
  • the excipient can serve various purposes.
  • a person skilled in the art can select one or more excipients with respect to the particular desired properties by routine experimentation and without any undue burden.
  • the amount of each excipient used can vary within ranges conventional in the art. Techniques and excipients which can be used to formulate dosage forms are described in Handbook of Pharmaceutical Excipients, 6th edition, Rowe et al., Eds., American Pharmaceuticals Association and the Pharmaceutical Press, publications department of the Royal Pharmaceutical Society of Great Britain (2009); and Remington: the Science and Practice of Pharmacy, 21st edition, Gennaro, Ed., Lippincott Williams & Wilkins (2005).
  • immune response refers to a biological response within an organism against a foreign agent or abnormal cell (e.g., a tumor cell), wherein the response protects the organism against such agents/cells and diseases caused by them.
  • An immune response is mediated by the action of a cell of the immune system (e.g., a T lymphocyte (T cell), B lymphocyte (B cell), natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil) and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results in selective targeting, binding to, damage to, destruction of, and/or elimination from the organism's body of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
  • T cell T lymphocyte
  • B cell B lymphocyte
  • NK natural killer
  • an immune reaction includes, e.g., activation or inhibition of a T cell, e.g., an effector T cell or a Th cell, such as a CD4+ or CD8+ T cell, or the inhibition of a regulatory T cell (Treg cell).
  • a T cell e.g., an effector T cell or a Th cell, such as a CD4+ or CD8+ T cell, or the inhibition of a regulatory T cell (Treg cell).
  • autologous refers to any material derived from an individual that will later be reintroduced into that same individual.
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • Papain digestion of intact antibodies produces two identical antigen-binding fragments, called “Fab” fragments containing each the heavy- and light-chain variable domains (VH and VL, respectively) and also the constant domain of the light chain (CL) and the first constant domain of the heavy chain (CHI).
  • Fab fragment thus refers to an antibody fragment comprising a light chain comprising a VL domain and a CL domain, and a heavy chain fragment comprising a VH domain and a CHI domain.
  • an "isolated" antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC) methods.
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs), including the complementarity determining regions (CDRs) (see, e.g., Kindt et al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007)).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • CDRs complementarity determining regions
  • a "paratope” or an “antigen binding site”, as used interchangeably herein, refers to a part of an antibody which recognizes and binds to an antigen.
  • An antigen binding site is formed by several individual amino acid residues from the antibody's heavy and light chain variable domains arranged that are arranged in spatial proximity in the tertiary structure of the Fv region.
  • the antigen binding site is defined as a set of the six CDRs comprised in a cognate VH/VL pair.
  • CDRs complementarity determining regions
  • antibodies comprise six CDRs: three in the VH domain (CDR-H1, CDR-H2, CDR-H3), and three in the VL domain (CDR-L1, CDR-L2, CDR- L3).
  • CDR residues and other residues in the variable domain are numbered herein according to the Kabat numbering system (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991).
  • An "acceptor human framework” for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • VL light chain variable domain
  • VH heavy chain variable domain
  • Framework refers to variable domain amino acid residues other than CDR residues.
  • the framework of a variable domain generally consists of four framework domains: FR1, FR2, FR3, and FR4. Accordingly, the CDR and FR amino acid sequences generally appear in the following sequence in the (a) VH domain: FR1-CDR-H1- FR2-CDR-H2-FR3-CDR-H3-FR4; and (b) in the VL domain: FR1-CDR-L1-FR2-CDR-L2- FR3-CDR-L3-FR4.
  • affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described herein.
  • epitope denotes the site on an antigen, either proteinaceous or non-proteinaceous, to which an antibody binds.
  • Epitopes can be formed both from contiguous amino acid stretches (linear epitope) or comprise non-contiguous amino acids (conformational epitope), e.g. coming in spatial proximity due to the folding of the antigen, i.e. by the tertiary folding of a proteinaceous antigen.
  • Linear epitopes are typically still bound by an antibody after exposure of the proteinaceous antigen to denaturing agents, whereas conformational epitopes are typically destroyed upon treatment with denaturing agents.
  • An epitope comprises at least 3, at least 4, at least 5, at least 6, at least 7, or 8-10 amino acids in a unique spatial conformation.
  • Screening for antibodies binding to a particular epitope can be done using methods routine in the art such as, e.g., without limitation, alanine scanning, peptide blots (see Meth. Mol. Biol. 248 (2004) 443-463), peptide cleavage analysis, epitope excision, epitope extraction, chemical modification of antigens (see Prot. Sci. 9 (2000) 487-496), and cross-blocking (see “Antibodies”, Harlow and Lane (Cold Spring Harbor Press, Cold Spring Harb., NY).
  • Antigen Structure-based Antibody Profiling also known as Modification- Assisted Profiling (MAP)
  • MAP Modification- Assisted Profiling
  • the antibodies in each bin bind to the same epitope which may be a unique epitope either distinctly different from or partially overlapping with epitope represented by another bin.
  • competitive binding can be used to easily determine whether an antibody binds to the same epitope, or competes for binding with, a reference antibody.
  • an "antibody that binds to the same epitope" as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • the reference antibody is allowed to bind to its antigen under saturating conditions. After removal of the excess of the reference antibody, the ability of an antibody in question to bind to its antigen is assessed.
  • the antibody in question binds to a different epitope than the reference antibody. But, if the antibody in question is not able to bind to its antigen after saturation binding of the reference antibody, then the antibody in question may bind to the same epitope as the epitope bound by the reference antibody. To confirm whether the antibody in question binds to the same epitope or is just hampered from binding by steric reasons routine experimentation can be used (e.g., peptide mutation and binding analyses using ELISA, RIA, surface plasmon resonance, flow cytometry or any other quantitative or qualitative antibody-binding assay available in the art).
  • This assay should be carried out in two set-ups, i.e. with both of the antibodies being the saturating antibody. If, in both set-ups, only the first (saturating) antibody is capable of binding to the antigen of interest, then it can be concluded that the antibody in question and the reference antibody compete for binding to the same antigen.
  • two antibodies are deemed to bind to the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate binding of one antibody also reduce or eliminate binding of the other.
  • Two antibodies are deemed to have "overlapping epitopes" if only a subset of the amino acid mutations that reduce or eliminate binding of one antibody reduce or eliminate binding of the other.
  • anti -turn or effect refers to a biological effect that can be manifested in various ways, including but not limited to, for example, reduction in tumor volume, reduction in the number of tumor cells, reduction in the number of metastases, increase in life expectancy, reduction in tumor cell proliferation, and reduction in tumor cell survival rate, or improvement in various physiological symptoms related to cancerous conditions.
  • the "anti-tumor effect” can also be expressed by the ability of the peptides, polynucleotides, cells and antibodies of the present disclosure to prevent or reduce the frequency of tumorigenesis.
  • chemotherapy refers to a wide variety of chemotherapeutic agents that may be used in accordance with the present embodiments.
  • chemotherapy refers to the use of drugs to treat cancer.
  • a “chemotherapeutic agent” is used to connote a compound or composition that is administered in the treatment of cancer.
  • composition refers to a preparation which is in such form as to permit the biological activity of the active ingredient to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the composition would be administered.
  • the composition can be sterile.
  • recombinant IL-12 has been demonstrated to induce profound T-cell mediated antitumor effects causing regression of established tumors, followed by systemic immune memory. See The Oncologist, 1996, vol. 1, 88. However, systemic administration of recombinant IL-12 has resulted in dose limiting toxicity in several experimental trials and in an initial human trial. See Lab Invest., 1994, vol. 71, 862; Science, 1995, vol. 270, 908; J. Interferon Cytokine Res., 1995, vol. 14, 335. Dose limiting toxicity was also observed with intraperitoneal administration of recombinant IL-12 in a recent human clinical trial. Clin. Cancer Res., 2002, vol. 8, 3686. A gene delivery approach that can provide therapeutic levels of IL-12 locally at the tumor site would have the advantage of generating an anticancer response without causing systemic toxicity.
  • IL-12 protein therapy When tumors were subjected to this gene therapy, they displayed all the characteristics of IL-12 protein therapy, e.g., an increased infiltration of NK cells, CD4 and CD8 T cells, coupled with an increased expression of major histocompatibility complex (MHC) class I molecules.
  • MHC major histocompatibility complex
  • IL-12 gene delivery was well tolerated and highly effective against both Renca and CT26 tumor bearing animals. Tumor rejecting mice were also protected from a subsequent rechallenge, suggesting the presence of a long lasting systemic immunity.
  • a functionalized and less toxic water soluble lipopolymer (WSLP) has been tested for delivery of the IL- 12 gene to CT26 colon carcinoma tumors. See Mahato et al, Mol. Ther., 2001, vol. 4, 130.
  • IL-12 plasmid (pIL-12) and WSLP (pIL- 12/WSLP) treatment gave higher levels of intratumoral gene expression than naked DNA.
  • Interleukin- 12 is a pro-inflammatory cytokine that plays an important role in innate and adaptive immunity. Gately, MK et al., Annu Rev Immunol. 16: 495-521 (1998). IL-12 functions primarily as a 70 kDa heterodimeric protein consisting of two disulfide- linked p35 and p40 subunits. IL-12 p40 homodimers do exist, but other than functioning as an antagonist that binds the IL- 12 receptor, they do not appear to mediate a biologic response. Id.
  • the precursor form of the IL-12 p40 subunit (e.g., NM_002187; NP_002178; also referred to as IL-12B, natural killer cell stimulatory factor 2, cytotoxic lymphocyte maturation factor 2) is 328 amino acids in length, while its mature form is 306 amino acids long.
  • the precursor form of the IL-12 p35 subunit (e.g., NM_000882; NP_000873; also referred to as IL- 12 A, natural killer cell stimulatory factor 1, cytotoxic lymphocyte maturation factor 1) is 219 amino acids in length and the mature form is 197 amino acids long. Id.
  • the genes for the IL- 12 p35 and p40 subunits reside on different chromosomes and are regulated independently of each other. Gately, MK et al., Annu Rev Immunol. 16:495- 521 (1998). Many different immune cells (e.g., dendritic cells, macrophages, monocytes, neutrophils, and B cells) produce IL-12 upon antigenic stimuli. The active IL-12 heterodimer is formed following protein synthesis. Id.
  • IL-12 protein Due to its ability to activate both NK cells and cytotoxic T cells, IL-12 protein has been studied as a promising anti-cancer therapeutic since 1994. See Nastala, C. L. et al., J Immunol 153: 1697-1706 (1994). But despite high expectations, early clinical studies did not yield satisfactory results. Lasek W. et al., Cancer Immunol Immunother 63:419-435, 424 (2014). Repeated administration of IL-12, in most patients, led to adaptive response and a progressive decline of IL- 12-induced interferon gamma (IFNy) levels in blood. Id.
  • IFNy interferon gamma
  • IL- 12-induced anti-cancer activity is largely mediated by the secondary secretion of IFNy
  • the concomitant induction of IFNy along with other cytokines (e.g., TNF-a) or chemokines (IP- 10 or MIG) by IL-12 caused severe toxicity. Id.
  • the marginal efficacy of the IL- 12 therapy in clinical settings may be caused by the strong immunosuppressive environment in humans.
  • IL-12 is mainly secreted from antigen presenting cells (phagocytes, dendritic cells) in response to pathogens, promoting CD4+ cell differentiation into Thl cells.
  • IL-12 IL-12 receptor
  • IL-12R The IL-12 receptor (IL-12R) consists of two chains, IL-12RP1 and IL-12RP213 and signals predominately through STAT4.
  • the IL-12R is expressed mainly by activated NK and T cells; it is barely detectable in resting T cells but is expressed at a low level in NK, probably explaining their rapid response to IL-12. Nevertheless, TCR activation and co-stimuli like B7, IFN-a, IFN-y and IL- 12 itself upregulate IL-12R expression (particularly IL-12RP2).
  • IL-12 Antitumor activity of systemic or local administration of IL-12 has been established in preclinical studies against various tumor cell lines. Dose and model dependent response as well as a memory antitumor effect were observed. Other studies suggest that IL- 12 is more efficacious against early stage or microscopic tumors than advanced disease. The T cell response is responsible for its antitumor activity and NK/NKT cell activation seems to prevent metastasis.
  • IL-12 inhibits tumor derived regulatory T cells (Treg) either through suppression of T cell IL-2 production, induction of apoptosis or by IFN-y mediated cell arrest, thus enhancing the Teff/Treg ratio both in vitro and in vivo.
  • IL-12 was also reported to mediate reprogramming of intratumoral myeloid derived suppressor cells (MDSC) to enhance CTL activity in a B16 melanoma model, reversing their suppressive role in vivo.
  • MDSC intratumoral myeloid derived suppressor cells
  • IL-12RP2 has also emerged as a potential tumor suppressor gene, particularly in hematologic malignancies where epigenetic IL-12RP2 gene silencing by promoter hypermethylation or gene downregulation were observed in multiple B-cell myeloma specimens and IL-12 significantly reduced tumor load upon receptor restoration. Consistently, IL-12RP2-/- mice develop spontaneous malignancies and lymphoproliferative diseases. IL-12 also has "direct" antitumor activity in IL-12RP1 expressing tumors through IFN-y mediated upregulation of MHC Class I molecules.
  • IL-12 is a key mediator of the Thl response
  • IL-12 has improved immune responses but this did not translate into clinically significant anti-tumor effects.
  • NHS-IL-12 is as immunokine consisting of two IL- 12 molecules fused to a tumor necrosis-targeting human IgGl with longer half-life. It has now entered phase II clinical trial testing.
  • Carson et al. conducted an NCI-sponsored phase I trial to determine the safety and optimal biologic dose of IL-12 when given in combination with trastuzumab.
  • Patients with metastatic HER2 -positive malignancies received trastuzumab on day 1 of each weekly cycle (4 mg/kg initially and 2 mg/kg thereafter). Beginning in week 3, patients also received intravenous (i.v.) injections of IL- 12 on days 2 and 5 of the weekly cycle.
  • the IL- 12 component was dose-escalated within cohorts of three patients (30, 100, 300, or 500 ng/kg). Fifteen patients were treated. The regimen was well-tolerated and no drug-related Grade 3 or 4 toxicities were noted.
  • the IL-12 component was dose-escalated in cohorts of three patients (100 ng/kg and then 300 ng/kg), but because of dose-limiting grade 3 fatigue at the 300 ng/kg dose level, the IL-12 component was reduced to 200 ng/kg subcutaneously. Overall there was a 52% rate of clinical benefit. There was significantly increased activation of ERK in peripheral blood mononuclear cells and increased plasma levels of IFN-y with clinical benefit (complete response, partial response, or stable disease), but not in patients with progressive disease. No patient with progressive disease had measurable levels of IFN-y in their circulation. Within any one cycle, IFN-y levels typically peaked following injections of IL-12 (range 124-1612 pg/ml) and then fell to baseline.
  • Ovarian cancer represents the fifth most common form of cancer affecting women and is the most lethal of gynecological malignancies, ranking fourth in cancer deaths among women.
  • First-line chemotherapy regimens for the treatment of ovarian cancer are typically platinum-based combination therapies that are administered intravenously (IV) for 4 to 6 treatments every 21 to 28 days. Although nearly 90% of women respond initially to platinumbased therapies, 55-75% of women will develop recurrent ovarian cancer within 2 years.
  • the second and third line therapies are generally ineffective and toxic including immune checkpoint inhibitors with a 11-15% response rate in platinum resistant, recurrent patients. There is an immediate need for safer and more effective therapies for the treatment of ovarian cancer.
  • Cancers can evade detection and destruction by the immune system despite the fact that many tumors elicit a strong immune response evident in lymphocyte infiltrates of the primary lesion.
  • Tumor immune evasion can be categorized into induction of immune tolerance and resistance to killing by activated immune effector cells.
  • the 'immunoediting' hypothesis suggests that tumors manipulate their microenvironment by creating complex local and regional immunosuppressive networks comprising various tumor-derived cytokines and other soluble factors. Therefore, by the time tumors have become clinically detectable, the tumor has already evolved mechanisms to evade the immune response mounted by the host against it. Such resistance mechanisms must be overcome to create effective and durable antitumor immunity.
  • TME tumor microenvironment
  • the GEN-1 nanoparticle comprises a DNA plasmid encoding IL-12 gene and a synthetic polymer facilitating plasmid delivery.
  • GEN-1 is designed to be delivered locally (e.g., intraperitoneally), offering the potential for cytokines to be expressed specifically in the tumor micro-environment with the goal of achieving increased efficacy while minimizing potential systemic toxicity.
  • GEN-1 has been studied in subjects with recurrent ovarian cancer as a single agent or combination agent with standard chemotherapy.
  • a phase I monotherapy trial evaluating 13 platinum-resistant EOC patients who were treated with IP GEN-1 at 0.6 mg/m 2 , 3 mg/m 2 , 12 mg/m 2 and 24 mg/m 2 weekly every 4 weeks evaluating for safety and tolerability.
  • the most frequent adverse events (AE) were fever and abdominal pain.
  • a GOG trial evaluated GEN-1 in 16 patients with persistent or recurrent platinum- resistant EOC.
  • higher doses of GEN-1 were assessed in combination with intravenous pegylated liposomal doxorubicin (PLD) 40 mg/m 2 (dose level 1 and 2) or 50 mg/m 2 (dose level 3) every 28 days and intraperitoneal GEN-1 at 24 mg/m 2 (dose level 1) or 36 mg/m 2 (dose level 2 and 3) on days 1, 8, 15, and 22 of a 28-day cycle. Cycles were repeated every 28 days until disease progression.
  • the GEN-1 DNA plasmid is delivered using a synthetic polymer facilitating plasmid delivery that is a lipopolymer.
  • the lipopolymer comprises a polyethyleneimine (PEI) covalently linked independently to cholesterol and polyethylene glycol (PEG) groups.
  • PEI polyethyleneimine
  • PEG polyethylene glycol
  • the present disclosure provides a polymeric system, PEG-PEI-Cholesterol (PPC), which differs from WSLP (PEI-Cholesterol) in that it contains PEG moi eties and yields significantly higher transfection efficiency in tumors.
  • PPC PEG-PEI-Cholesterol
  • the addition of PEG is designed to enhance the stability of the nucleic acid/polymer complexes in the biological milieu to circumvent for this deficiency in the prior art (WSLP).
  • WSLP prior art
  • the addition of PEG chains allows for the incorporation of ligands on to the PPC chain to improve the tissue selectivity of delivery.
  • the cholesterol moiety which is directly linked to the PEI back bone in the prior art (WSLP) may be extended farther from the PEI backbone to create a more flexible geometry for cell receptor interaction.
  • Controlling the number of PEG molecules per unit of the PEI backbone is important to achieve optimal enhancement in transfection activity.
  • a preferred range of composition was a PEG:PEI molar ratio of 2-4 at a fixed cholesterol content.
  • the optimal ratio between PEI and cholesterol was 1 :0.5 to 1 : 1.
  • Certain aspects of the disclosure are related to a combination therapy comprising: a (i) a nucleic acid vector (e.g., a plasmid) comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and (ii) an immune checkpoint inhibitor.
  • a nucleic acid vector e.g., a plasmid
  • IL- 12 interleukin- 12
  • lipopolymer e.g., a nanoparticle
  • Certain aspects of the disclosure are related to a method of treating a subject suffering from cancer comprising administering to the subject a combination therapy comprising: (i) a nucleic acid vector (e.g., a plasmid) comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and (ii) an immune checkpoint inhibitor.
  • a nucleic acid vector e.g., a plasmid
  • IL- 12 interleukin- 12
  • a lipopolymer e.g., a nanoparticle
  • the polynucleotide encodes human IL-12. In some aspects, the polynucleotide encodes a p35 subunit of IL-12 and a p40 subunit of IL-12.
  • nucleic acid vector (e.g., a plasmid) comprises a promoter operably linked to a nucleic acid encoding a p35 subunit of IL- 12 and a promoter operably linked to a nucleic acid encoding a p40 subunit of IL12.
  • the human IL-12 p35 comprises an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 85.
  • the human IL-12 p40 comprises an amino acid sequence having at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 86.
  • the human IL-12 p35 comprises the sequence: MGPARSLLLVATLVLLDHLSLARNLPVATPDPGMFPCLHHSQNLLRAVSNMLQK ARQTLEFYPCTSEEIDHEDITKDKTSTVEACLPLELTKNESCLNSRETSFITNGSCL ASRKTSFMMALCLSSIYEDLKMYQVEFKTMNAKLLMDPKRQIFLDQNMLAVIDE LMQALNFNSETVPQKSSLEEPDFYKTKIKLCILLHAFRIRAVTIDRVMSYLNAS (SEQ ID NO: 85).
  • the human IL-12 p40 comprises the sequence: MGHQQLVISWFSLVFLASPLVAIWELKKDVYVVELDWYPDAPGEMVVLTCDTP EEDGITWTLDQSSEVLGSGKTLTIQVKEFGDAGQYTCHKGGEVLSHSLLLLHKKE DGIWSTDILKDQKEPKNKTFLRCEAKNYSGRFTCWWLTTISTDLTFSVKSSRGSS DPQGVTCGAATLSAERVRGDNKEYEYSVECQEDSACPAAEESLPIEVMVDAVHK LKYENYTS SFFIRDIIKPDPPKNLQLKPLKNSRQVEVSWEYPDTWSTPHS YF SLTFC VQVQGKSKREKKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVPCS (SEQ ID NO: 86).
  • the polynucleotide encoding the human IL- 12 p35 has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 87. In some aspects, the polynucleotide encoding the human IL-12 p35 has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% identity to SEQ ID NO: 88.
  • the polynucleotide encoding the human IL-12 p35 comprises the sequence: atgggtccagcgcgcagcctcctccttgtggctaccctggtcctcctggaccacctcagtttggccagaaacctccccgtggcca ctccagacccaggaatgttcccatgccttcaccactcccaaaacctgctgagggccgtcagcaacatgctccagaaggccagac aaactctagaattttacccttgcacttctgaagagattgatcatgaagatatcacaaagataaaaccagcacagtggaggcctgt taccattggaattaaccaagaatgagagttgcctaaattccagagagacctctttcataactaatgggagttgcttcataactaatggg
  • the polynucleotide encoding the human IL-12 p35 comprises the sequence:
  • nucleic acid vector (e.g., a plasmid) comprises an intron, a 3' UTR (e.g., hGH 3' UTR), an antibiotic resistance gene, or any combination thereof (e.g., the elements of FIG. 1).
  • a 3' UTR e.g., hGH 3' UTR
  • an antibiotic resistance gene e.g., the elements of FIG. 1.
  • the lipopolymer comprises a polyethyleneimine (PEI) covalently linked independently to cholesterol and polyethylene glycol (PEG) groups (e.g., the lipopolymer of FIG. 2).
  • PEI polyethyleneimine
  • PEG polyethylene glycol
  • the nanoparticle disclosed herein comprises a DNA plasmid that encodes human IL- 12.
  • nanoparticle comprises a synthetic polymer facilitating plasmid delivery that is a lipopolymer.
  • the lipopolymer further comprises polyethyleneimine (PEI) covalently linked independently to cholesterol and polyethylene glycol (PEG) groups
  • the gene delivery polymer is a cationic polymer or a noncondensing polymer.
  • the cationic polymer is selected from the group comprising polylysine, polyethylenimine, functionalized derivatives of polyethylenimine (PEI), polypropylenimine, aminoglycoside-polyamine, dideoxy-diamino-b-cyclodextrin, spermine and spermidine.
  • a cationic gene delivery polymer suitable for the present disclosure is a PEI derivative comprising a PEI backbone, a lipid, and a hydrophilic polymer spacer wherein the lipid is directly bound to the polyethylenimine backbone or covalently bound to the polyethylene glycol spacer, which in turn is bound, via a bio compatible bond, to the PEI.
  • the cationic gene delivery polymer of the present disclosure may further comprise a targeting moiety including antibodies or antibody fragments, cell receptors, growth factor receptors, cytokine receptors, folate, transferrin, epidermal growth factor (EGF), insulin, asialoorosomucoid, mannose-6-phosphate (monocytes), mannose (macrophage, some B cells), Lewis x and sialyl Lewis x (endothelial cells), N-acetyllactosamine (T cells), galactose (colon carcinoma cells), and thrombomodulin (mouse lung endothelial cells), fusogenic agents such as polymixin B and hemaglutinin HA2, lysosomotrophic agents, nucleus localization signals (NLS) such as T-antigen, and the like.
  • a targeting moiety including antibodies or antibody fragments, cell receptors, growth factor receptors, cytokine receptors, folate, transferrin, epidermal growth factor (EGF
  • Another gene delivery polymer is a non-condensing polymer selected from the group comprising polyvinylpyrrolidone, polyvinylalcohol, poly(lactide-co-glycolide) (PLGA) and triblock copolymers of PLGA and PEG.
  • the gene delivery polymer may also be a non-condensing polymer.
  • non-condensing polymers examples include polyvinyl pyrollidone, polyvinyl alcohol, poloxamers, polyglutamate, gelatin, polyphosphoesters, silk-elastin-like hydrogels, agarose hydrogels, lipid microtubules, poly(lactide-co-glycolide) and polyethyleneglycol-linked poly(lactide-co- glycolide).
  • the gene delivery polymer is a cationic polymer or a non-condensing polymer.
  • the cationic polymer is selected from the group comprising polylysine, polyethylenimine, functionalized derivatives of polyethylenimine, polypropylenimine, aminoglycosidepolyamine, dideoxy-diamino-b-cyclodextrin, spermine and spermidine.
  • a cationic gene delivery polymer suitable for the present invention is a polyethylenimine derivative comprising a polyethylenimine (PEI) backbone, a lipid, and a polyethylene glycol spacer wherein the lipid is directly bound to the polyethylenimine backbone or covalently bound to the polyethylene glycol spacer, which in turn is bound, via a biocompatible bond, to the PEI.
  • PEI polyethylenimine
  • the gene delivery polymer comprises a lipopolyamine with the following formula: [0183] (Staramine).
  • the gene delivery polymer comprises a mixture of the lipopolyamine and an alkylated derivative of the lipopolyamine.
  • the alkylated derivative of the lipopolyamine is a polyoxyalkylene, polyvinylpyrrolidone, polyacrylamide, polydimethylacrylamide, polyvinyl alcohol, dextran, poly (L-glutamic acid), styrene maleic anhydride, poly-N-(2-hydroxypropyl) methacrylamide, or polydivinylether maleic anhydride.
  • the alkylated derivative of the lipopolyamine has the following formula:
  • n an integer from 10 to 100 repeating units containing 2-5 carbon atoms each.
  • the alkylated derivative of the lipopolyamine has the following formula:
  • n 11 (Staramine-mPEG515).
  • alkylated derivative of the lipopolyamine has the following formula:
  • the ratio of the lipopolyamine to the alkylated derivative of the lipopolyamine in the mixture is 1:1 to 10:1.
  • the lipopolyamine is present in an amount sufficient to produce a ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector from about 0.01:1 to about 50:1 (e.g., about 0.01:1 to about 40:1; about 0.01:1 to about 30:1; about 0.01:1 to about 20:1; about 0.01:1 to about 10:1, or about 0.01:1 to about 5:1).
  • the ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector is from about 0.1 : 1 to about 50: 1 (e.g., about 0.1:1 to about 40:1; about 0.1:1 to about 30:1; about 0.1:1 to about 20:1; about 0.1:1 to about 10:1, or about 0.1:1 to about 5:1). In some aspects, the ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector is from about 1 : 10 to about 10:1.
  • the gene delivery polymer comprises a lipopolyamine with the following formula:
  • the gene delivery polymer comprises a mixture of the lipopolyamine and an alkylated derivative of the lipopolyamine.
  • the alkylated derivative of the lipopolyamine is a polyoxyalkylene, polyvinylpyrrolidone, polyacrylamide, polydimethylacrylamide, polyvinyl alcohol, dextran, poly (L-glutamic acid), styrene maleic anhydride, poly-N-(2-hydroxypropyl) methacrylamide, or polydivinylether maleic anhydride.
  • the ratio of the lipopolyamine to the alkylated derivative of the lipopolyamine in the mixture is 1:1 to 10:1.
  • the lipopolyamine is present in an amount sufficient to produce a ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector from about 0.01:1 to about 50: 1 (e.g., about 0.01:1 to about 40:1; about 0.01:1 to about 30:1; about 0.01:1 to about 20:1; about 0.01:1 to about 10:1, or about 0.01 : 1 to about 5:1).
  • the ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector is from about 0.1 : 1 to about 50: 1 (e.g., about 0.1:1 to about 40: 1 ; about 0.1 : 1 to about 30:1; about 0.1:1 to about 20: 1 ; about 0.1 : 1 to about 10:1, or about 0.1 : 1 to about 5:1). In some aspects, the ratio of amine nitrogen in the lipopolyamine to phosphate in the nucleic acid vector is from about 1 : 10 to about 10:1.
  • the gene delivery polymer comprises a poloxamer back-bone having a metal chelator covalently coupled to at least one terminal end of the poloxamer backbone.
  • the metal chelator is coupled to at least two terminal ends of the poloxamer backbone.
  • the poloxamer backbone is a poloxamer backbone disclosed in U.S. Publ. No. 2010/0004313, which is herein incorporated by reference in its entirety.
  • the metal chelator is a metal chelator disclosed in U.S. Publ. No. 2010/0004313.
  • the gene delivery polymer comprises a polymer having the following formula:
  • A represents an integer from 2 to 141;
  • B represents an integer from 16 to 67;
  • C represents an integer from 2 to 141;
  • RA and RC are the same or different, and are R'-L- or H, wherein at least one of
  • RA and RC is R'-L-;
  • L is a bond, —CO—, — CH2— O— , or — O— CO— ;
  • R' is a metal chelator
  • the metal chelator is RNNH — , RN2N — , or (R" — (N(R") — CH2CH2)x)2 — N — CH2CO — , wherein each x is independently 0-2, and wherein R" is HO2C — CH2 — .
  • the metal chelator is a crown ether selected from the group consisting of 12-crown-4, 15-crown-5, 18-crown-6, 20-crown-6, 21-crown-7, and 24-crown- 8.
  • the crown ether is a substituted-crown ether, wherein the substituted crown ether has:
  • the metal chelator is a cryptand, wherein the cryptand is selected from the group consisting of (1,2,2) cryptand, (2,2,2) cryptand, (2,2,3) cryptand, and (2,3,3) cryptand.
  • the cryptand is a substituted-cryptand, wherein the substituted cryptand has:
  • the gene delivery polymer is Crown Poloxamer (aza-crown- linked poloxamer), wherein the Crown Poloxamer comprises a polymer having the following formula:
  • a represents an integer of about 10 units
  • b represents an integer of about 21 units
  • the gene delivery polymer is present in a solution with the nucleic acid vector from about 0.1% - about 5% or about 0.5% - about 5%.
  • the gene delivery polymer is a P-amino ester. In some aspects, the polymer is present in a solution with the nucleic acid vector from about 0.1% - about 5% or about 0.5% - about 5%.
  • the gene delivery polymer is a poly-inosinic-polycytidylic acid.
  • the poly-inosinic-polycytidylic acid is present in a solution with the nucleic acid vector from about 0.1% - about 5% or about 0.5% - about 5%.
  • the gene delivery polymer further comprises benzalkonium chloride.
  • the gene delivery polymer comprises BD15-12.
  • the ratio of nucleotide to BD15-12 polymer is 5: 1.
  • the gene delivery polymer comprises Omnifect.
  • the ratio of nucleotide to Omnifect polymer (N:P) is 10: 1.
  • the gene delivery polymer comprises Crown Poloxamer (azacrown-linked pol oxamer). In some aspects, the ratio of nucleotide to Crown Poloxamer (N:P) is 5: 1. In some aspects, the gene delivery polymer comprises Crown Poloxamer and a PEG- PEI-cholesterol (PPC) lipopolymer. In some aspects, the gene delivery polymer comprises Crown Poloxamer and benzalkonium chloride. In some aspects, the gene delivery polymer comprises Crown Poloxamer and Omnifect. In some aspects, the gene delivery polymer comprises Crown Poloxamer and a linear polyethyleneimine (LPEI). In some aspects, the gene delivery polymer comprises Crown Poloxamer and BD 15-12.
  • PPC PEG- PEI-cholesterol
  • the gene delivery polymer comprises Staramine and mPEG modified Staramine.
  • the mPEG modified Staramine is Staramine-mPEG515.
  • the mPEG modified Staramine is Staramine-mPEGl 1.
  • the ratio of Staramine to mPEG modified Staramine is 10: 1.
  • the nucleotide to polymer (N:P) ratio is 5: 1.
  • the gene delivery polymer comprises Staramine, mPEG modified Staramine, and Crown Poloxamer.
  • the gene delivery polymer comprises Staramine, Staramine-mPEG515, and Crown Poloxamer.
  • the gene delivery polymer comprises Staramine, Staramine-mPEGl 1, and Crown Poloxamer.
  • the gene delivery polymer comprises a poloxamer backbone disclosed in WO 2022/072910 Al, which is herein incorporated by reference in its entirety.
  • the nanoparticle that comprises a DNA plasmid that encodes interleukin- 12 (IL- 12) and a synthetic polymer facilitating plasmid delivery is delivered intraperitoneally.
  • the nanoparticle is administered at a dose of about 35 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 40 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 45 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 50 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 55 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 60 mg/m 2 to about 80 mg/m 2 .
  • the nanoparticle is administered at a dose of about 65 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 70 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 75 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 75 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 70 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 65 mg/m 2 .
  • the nanoparticle is administered at a dose of about 35 mg/m 2 to about 60 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 55 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 50 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 45 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 40 mg/m 2 .
  • the nanoparticle is administered at a dose of about 35 mg/m 2 , about 40 mg/m 2 , about 45 mg/m 2 , about 50 mg/m 2 , about 55 mg/m 2 , about 60 mg/m 2 , about 65 mg/m 2 , about 70 mg/m 2 , about 75 mg/m 2 , or about 80 mg/m 2 .
  • the nanoparticle is administered at a dose of about 60 mg/m 2 .
  • Immune checkpoint proteins interact with specific ligands which send a signal into T-cells that inhibits T-cell function. Cancer cells exploit this by driving high level expression of checkpoint proteins on their surface thereby suppressing the anti-cancer immune response.
  • An immune checkpoint inhibitor comprises any compound capable of inhibiting the function of an immune checkpoint protein. Inhibition includes reduction of function as well as full blockade.
  • the immune checkpoint protein is a human checkpoint protein.
  • the immune checkpoint inhibitor is an antagonist of an immune checkpoint protein. In some aspects, the immune checkpoint inhibitor is an agonist of an immune checkpoint protein.
  • the immune checkpoint protein is selected from the group consisting of CTLA-4, PD-1 (and its ligands PD-L1 and PD-L2), B7-H3, B7-H4, HVEM, TIM3, GAL9, LAG-3, VISTA, KIR, BTLA, TIGIT, IDO-1, CEA, PVRIG, GARP, STING, Siglec-15, CD20, CD27, CD38, CD39, CD47, CD66a (CEACAM1), CD73, CD80, CD86, CD93, CD96, and/or CD161.
  • the immune checkpoint inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 (and its ligands PD- L1 and PD-L2), and/or LAG-3.
  • the immune checkpoint inhibitor is a small molecule inhibitor.
  • the immune checkpoint inhibitor is an antibody or fragment thereof that specifically binds to PD-1. In some aspects, the immune checkpoint inhibitor is an antibody or fragment thereof that specifically binds to PD-L1. In some aspects, the immune checkpoint inhibitor is an antibody or fragment thereof that specifically binds to CTLA-4. In some aspects, the immune checkpoint inhibitor is an antibody or fragment thereof that specifically binds to LAG-3.
  • the immune checkpoint inhibitor is an antibody.
  • immune checkpoint inhibitors comprise an antibody or fragment thereof that specifically bind to an immune checkpoint protein.
  • the immune checkpoint inhibitor is a monoclonal antibody, a fully human antibody, a chimeric antibody, a humanized antibody or fragment thereof that capable of at least partly antagonizing an immune checkpoint protein.
  • the immune checkpoint inhibitor comprises a heavy chain variable region (VH) amino acid sequence and a light chain variable region (VL) amino acid sequence disclosed in Table 1.
  • the immune checkpoint inhibitor comprises a heavy chain (HC) amino acid sequence and a light chain (LC) amino acid sequence disclosed in Table 2.
  • the immune checkpoint inhibitor comprises a VH complementarity determining region (CDR) 1, a VH CDR2, a VH CDR3, a VL CDR1, a VL CDR2, and a VL CDR3 as disclosed in Table 3.
  • the immune checkpoint inhibitor is ipilimumab.
  • ipilimumab is administered at a dose of about 0.5 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 1 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 1.5 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 2 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 2.5 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 3 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 3.5 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 4 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 4 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 3 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 2.5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 2 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 1.5 mg/kg.
  • ipilimumab is administered at a dose of about 0.5 mg/kg, about 1 mg/kg, about 1.5 mg/kg, about 2 mg/kg, about 2.5 mg/kg, about 3 mg/kg, about 3.5 mg/kg, about 4 mg/kg, about 4.5 mg/kg, or about 5 mg/kg.
  • ipilimumab is administered at a dose of about 1 mg/kg.
  • ipilimumab is administered every 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.
  • ipilimumab is administered every 2-8 weeks (e.g., every 6 weeks) during treatment.
  • ipilimumab is administered intravenously.
  • the immune checkpoint inhibitor is nivolumab.
  • nivolumab is administered at a dose of about 120 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 140 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 160 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 180 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 200 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 220 mg to about 360 mg.
  • nivolumab is administered at a dose of about 240 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 260 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 280 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 300 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 320 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 340 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 340 mg.
  • nivolumab is administered at a dose of about 120 mg to about 320 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 300 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 280 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 260 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 220 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 200 mg.
  • nivolumab is administered at a dose of about 120 mg to about 180 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 160 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 140 mg. In some aspects, nivolumab is administered at a dose of about 240 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 260 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 280 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 300 mg to about 360 mg.
  • nivolumab is administered at a dose of about 320 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 340 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 140 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 160 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 180 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 200 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 220 mg to about 240 mg.
  • nivolumab is administered at a dose of about 120 mg, about 140 mg, about 160 mg, about 180 mg, about 200 mg, about 220 mg, about 240 mg, about 260 mg, about 280 mg, about 300 mg, about 320 mg, about 340 mg, or about 360 mg.
  • nivolumab is administered at a dose of about 240 mg.
  • nivolumab is administered intravenously.
  • nivolumab is administered every 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.
  • nivolumab is administered every 1-4 weeks (e.g., every 2 weeks) during treatment.
  • Human monoclonal antibodies that bind specifically to PD-1 with high affinity have been disclosed in U.S. Patent Nos. 8,008,449 and 8,779,105.
  • Other anti-PD-1 mAbs have been described in, for example, U.S. Patent Nos. 6,808,710, 7,488,802, 8,168,757 and 8,354,509, and PCT Publication No. WO 2012/145493.
  • the anti-PD-1 antibodies of the combination therapy disclosed herein include mAbs that bind specifically to human PD-1 with a KD of 1 x 10' 7 M or less, as determined by surface plasmon resonance using a Biacore biosensor system; (b) does not substantially bind to human CD28, CTLA-4 or ICOS; (c) increases T-cell proliferation in a Mixed Lymphocyte Reaction (MLR) assay; (d) increases interferon-y production in an MLR assay; (e) increases IL-2 secretion in an MLR assay; (f) binds to human PD-1 and cynomolgus monkey PD-1; (g) inhibits the binding of PD-L1 and/or PD-L2 to PD-1; (h) stimulates antigen-specific memory responses; (i) stimulates Ab responses; and (j) inhibits tumor cell growth in vivo.
  • the anti-PD-1 antibodies of the combination therapy disclosed herein include mAbs that bind specifically to human bind specifically
  • the anti-PD-1 antibody is nivolumab.
  • Nivolumab also known as "OPDIVO®”; BMS-936558; formerly designated 5C4, BMS-936558, MDX-1106, or ONO- 4538
  • S228P fully human IgG4
  • PD-1 immune checkpoint inhibitor antibody that selectively prevents interaction with PD-1 ligands (PD-L1 and PD-L2), thereby blocking the down-regulation of antitumor T-cell functions
  • the anti-PD-1 antibody or fragment thereof binds to the same epitope as nivolumab.
  • the anti-PD-1 antibody has the same CDRs as nivolumab.
  • Anti-PD-1 antibodies useful for the disclosed compositions also include isolated antibodies that bind specifically to human PD-1 and cross-compete for binding to human PD- 1 with nivolumab (see, e.g., U.S. Patent Nos. 8,008,449 and 8,779,105; Int'l Pub. No. WO 2013/173223).
  • the ability of antibodies to cross-compete for binding to an antigen indicates that these antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other cross-competing antibodies to that particular epitope region.
  • These crosscompeting antibodies are expected to have functional properties very similar to those of nivolumab by virtue of their binding to the same epitope region of PD-1.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with nivolumab in standard PD-1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., Int'l Pub. No. WO 2013/173223).
  • antibodies or antigen binding fragments thereof that cross-compete for binding to human PD-1 with, or bind to the same epitope region of human PD-1 as, nivolumab are mAbs.
  • these cross-competing antibodies can be chimeric antibodies, or humanized or human antibodies. Such chimeric, humanized or human mAbs can be prepared and isolated by methods well known in the art.
  • the anti-PD-1 antibody is pembrolizumab.
  • Pembrolizumab also known as "KEYTRUDA®", lambrolizumab, and MK-3475
  • S228P humanized monoclonal IgG4
  • Pembrolizumab is described, for example, in U.S. Patent Nos. 8,354,509 and 8,900,587.
  • Pembrolizumab has been approved by the FDA for the treatment of relapsed or refractory melanoma.
  • the anti-PD-1 antibody is REGN2810. In some aspects, the anti- PD-1 antibody is PDR001. Another known anti-PD-1 antibody is pidilizumab (CT-011). In some aspects, the anti-PD-1 antibody is MEDI0608 (formerly AMP-514), which is a monoclonal antibody. MEDI0608 is described, for example, in U.S. Patent No. 8,609,089. In some aspects, the anti-PD-1 antibody or antigen binding fragment thereof is BGB-A317, which is a humanized monoclonal antibody. BGB-A317 is described in U.S. Publ. No.
  • nnAntibodies or antigen binding fragments thereof that bind the same epitopes or have the same CDRs as any of these antibodies can be used.
  • Anti-human-PD-1 antibodies (or VH and/or VL domains derived therefrom) suitable for uses disclosed herein can be generated using methods well known in the art. Alternatively, art recognized anti-PD-1 antibodies can be used.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab (also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538), pembrolizumab (Merck; also known as KEYTRUDA®, lambrolizumab, and MK-3475; see WO2008/156712), PDR001 (Novartis; see WO 2015/112900), MEDI-0680 (AstraZeneca; also known as AMP-514; see WO 2012/145493), cemiplimab (Regeneron; also known as REGN-2810; see WO 2015/112800), JS001 (TAIZHOU JUNSHI PHARMA; see Si-Yang Liu et al., J.
  • nivolumab also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538
  • pembrolizumab Merck; also
  • Anti-PD-1 antibodies useful for the combination therapy of the disclosed invention also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; and (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody.
  • Anti-PD-1 antibodies suitable for use in the disclosed combination therapies are antibodies that bind to PD-1 with high specificity and affinity, block the binding of PD-L1 and or PD-L2, and inhibit the immunosuppressive effect of the PD-1 signaling pathway.
  • the anti-PD-1 antibody or antigen-binding portion thereof crosscompetes with nivolumab for binding to human PD-1.
  • the anti-PD-1 antibody or antigen-binding portion thereof is a chimeric, humanized or human monoclonal antibody or a portion thereof.
  • the antibody is a humanized antibody. In other embodiments, the antibody is a human antibody.
  • Antibodies of an IgGl, IgG2, IgG3 or IgG4 isotype can be used.
  • the anti-PD-1 antibody or antigen binding fragment thereof comprises a heavy chain constant region which is of a human IgGl or IgG4 isotype.
  • the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody or antigen binding fragment thereof contains an S228P mutation which replaces a serine residue in the hinge region with the proline residue normally found at the corresponding position in IgGl isotype antibodies.
  • the antibody comprises a light chain constant region which is a human kappa or lambda constant region.
  • the anti-PD-1 antibody or antigen binding fragment thereof is a mAb or an antigen-binding portion thereof.
  • the anti-PD-1 antibody is nivolumab.
  • the PD-1 antagonist is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, and dostarlimab.
  • Anti-human-PD-Ll antibodies (or VH and/or VL domains derived therefrom) suitable for use in the combination therapy disclosed herein can be generated using methods known in the art.
  • Examples of anti-PD-Ll antibodies useful in the methods of the present disclosure include the antibodies disclosed in US Patent No. 9,580,507, incorporated herein by reference.
  • 9,580,507 have been demonstrated to exhibit one or more of the following characteristics: (a) bind to human PD-L1 with a KD of 1 x 10' 7 M or less, as determined by surface plasmon resonance using a Biacore biosensor system; (b) increase T-cell proliferation in a Mixed Lymphocyte Reaction (MLR) assay; (c) increase interferon-y production in an MLR assay; (d) increase IL-2 secretion in an MLR assay; (e) stimulate antibody responses; and (f) reverse the effect of T regulatory cells on T cell effector cells and/or dendritic cells.
  • Anti-PD-Ll antibodies useful in the methods disclosed herein include monoclonal antibodies that bind specifically to human PD-L1 and exhibit at least one of the preceding characteristics.
  • a recognized anti-PD-Ll antibodies can be used.
  • human anti-PD-Ll antibodies disclosed in U.S. Pat. No. 7,943,743, the contents of which are hereby incorporated by reference can be used.
  • Such anti-PD-Ll antibodies include 3 G10, 12A4 (also referred to as BMS-936559), 10A5, 5F8, 10H10, 1B12, 7H1, 11E6, 12B7, and 13G4.
  • Other art recognized anti-PD-Ll antibodies which can be used include those described in, for example, U.S. Pat. Nos. 7,635,757 and 8,217,149, U.S. Publication No. 2009/0317368, and PCT Publication Nos.
  • Other examples of an anti-PD-Ll antibody include atezolizumab (TECENTRIQ; RG7446), or durvalumab (IMFINZI; MEDI4736) or avelumab (Bavencio).
  • Antibodies or antigen binding fragments thereof that compete with any of these art-recognized antibodies or inhibitors for binding to PD-L1 also can be used.
  • the anti-PD-Ll antibody is BMS-936559 (formerly 12A4 or MDX-1105) (see, e.g., U.S. Patent No. 7,943,743; WO 2013/173223).
  • the anti-PD-Ll antibody is MPDL3280A (also known as RG7446 and atezolizumab) (see, e.g., Herbst et al. 2013 J Clin Oncol 31 (suppl): 3000; U.S. Patent No. 8,217,149), MEDI4736 (Khleif, 2013, In: Proceedings from the European Cancer Congress 2013; September 27- October 1, 2013; Amsterdam, The Netherlands.
  • antibodies that cross-compete for binding to human PD-L1 with, or bind to the same epitope region of human PD-L1 as the above-references PD-L1 antibodies are mAbs.
  • these cross-competing antibodies can be chimeric antibodies, or can be humanized or human antibodies.
  • Such chimeric, humanized or human mAbs can be prepared and isolated by methods well known in the art.
  • the anti-PD-Ll antibody is selected from the group consisting of BMS-936559 (also known as 12A4, MDX-1105; see, e.g., U.S. Patent No.
  • Atezolizumab (Roche; also known as TECENTRIQ®; MPDL3280A, RG7446; see US 8,217,149; see, also, Herbst et al. (2013) J Clin Oncol 31 (suppl): 3000), durvalumab (AstraZeneca; also known as IMFINZITM, MEDL4736; see WO 2011/066389), avelumab (Pfizer; also known as BAVENCIO®, MSB-0010718C; see WO 2013/079174), STI-1014 (Sorrento; see WO2013/181634), CX-072 (Cytomx; see W02016/149201), KN035 (3D Med/Alphamab; see Zhang et al., Cell Discov.
  • the PD-L1 antibody is atezolizumab (TECENTRIQ®).
  • Atezolizumab is a fully humanized IgGl monoclonal anti-PD-Ll antibody.
  • the PD-L1 antibody is durvalumab (IMFINZITM). Durvalumab is a human IgGl kappa monoclonal anti-PD-Ll antibody. [0274] In some aspects, the PD-L1 antibody is avelumab (BAVENCIO®). Avelumab is a human IgGl lambda monoclonal anti-PD-Ll antibody.
  • the anti-PD-Ll monoclonal antibody is selected from the group consisting of 28-8, 28-1, 28-12, 29-8, 5H1, and any combination thereof.
  • the anti-PD-Ll antibodies of the disclosed methods comprise isolated antibodies that bind specifically to human PD-L1 and cross-compete for binding to human PD-L1 with any anti-PD-Ll antibody disclosed herein, e.g., atezolizumab, durvalumab, and/or avelumab.
  • the anti-PD-Ll antibody binds the same epitope as any of the anti-PD-Ll antibodies described herein, e.g., atezolizumab, durvalumab, and/or avelumab.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with atezolizumab and/or avelumab in standard PD-L1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the antibodies that cross-compete for binding to human PD-L1 with, or bind to the same epitope region of human PD-L1 antibody as, atezolizumab, durvalumab, and/or avelumab are monoclonal antibodies.
  • these cross-competing antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies.
  • Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • Anti-PD-Ll antibodies for use in the methods disclosed herein can include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • Monoclonal antibodies that specifically bind to CTLA-4 include, without limitation, Ipilimumab (Yervoy®; BMS) and Tremelimumab (AstraZeneca/Medlmmune), as well as antibodies disclosed in U.S. Patent Application Publication Nos. 2005/0201994, 2002/0039581, and 2002/0086014, the contents of each of which are incorporated herein by reference, and antibodies disclosed in U.S. Pat. Nos.
  • the immune checkpoint inhibitor is a CTLA-4 antagonist.
  • the CTLA-4 antagonist is selected from the group consisting of ipilimumab and tremelimumab. In some aspects, the CTLA-4 antagonist is ipilimumab.
  • the anti-CTLA-4 antibodies of the disclosed methods comprise isolated antibodies that bind specifically to human CTLA-4 and cross-compete for binding to human CTLA-4 with any anti-CTLA-4 antibody disclosed herein, e.g., ipilimumab.
  • the anti-CTLA-4 antibody binds the same epitope as any of the anti-CTLA-4 antibodies described herein, e.g., ipilimumab.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with ipilimumab in standard CTLA-4 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the antibodies that cross-compete for binding to human CTLA-4 with, or bind to the same epitope region of CTLA-4 as ipilimumab are chimeric antibodies, engineered antibodies, or humanized or human antibodies.
  • Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • Anti-CTLA-4 antibodies for use in the methods disclosed herein can include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • Anti -human-L AG-3 antibodies (or VH/VL domains derived therefrom) suitable for use in the combination therapy disclosed herein can be generated using methods well known in the art. Alternatively, art recognized anti-LAG-3 antibodies can be used. [0286] For example, the anti-human LAG-3 antibody described in US2011/0150892 Al, the teachings of which are hereby incorporated by reference, and referred to as monoclonal antibody 25F7 (also known as "25F7" and "LAG3.1”) can be used.
  • IMP731 H5L7BW
  • MK-4280 28G-10
  • REGN3767 described in Journal for ImmunoTherapy of Cancer, (2016) Vol. 4, Supp. Supplement 1 Abstract Number: Pl 95, BAP050 described in WO2017/019894
  • IMP-701 LAG-525
  • IMP321 eftilagimod alpha
  • Sym022 TSR-033, MGD013, BI754111, FS118, AVA-017 and GSK2831781.
  • anti-LAG-3 antibodies useful in the claimed invention can be found in, for example: WO2016/028672, W02017/106129, WO2017/062888, W02009/044273, WO20 18/069500, WO2016/126858, WO2014/179664, WO2016/200782, WO2015/200119, WO20 17/019846, WO2017/198741, WO2017/220555, WO2017/220569, WO2018/071500, W02017/015560, WO2017/025498, WO2017/087589, WO2017/087901, W02018/083087, WO2017/149143, WO2017/219995, US2017/0260271, WO2017/086367, WO/2017/086419, WO2018/034227, and W02014/140180.
  • the contents of each of these references are herein incorporated by reference.
  • Antibodies that compete with any of the above-referenced antibodies for binding to LAG-3 also can be used.
  • An exemplary anti-LAG-3 antibody is BMS-986016, as described in US Pat. No. 9,505,839, which is herein incorporated by reference. In some aspects, the anti-LAG-3 antibody is BMS-986016.
  • the antibody has the heavy and light chain CDRs or variable regions of BMS-986016.
  • the antibody competes for binding with and/or binds to the same epitope on LAG-3 as the above-mentioned antibodies.
  • the antibody binds an epitope of human LAG-3 comprising the amino acid sequence PGHPLAPG (SEQ ID NO: 82).
  • the antibody binds an epitope of human LAG-3 comprising the amino acid sequence HPAAPSSW (SEQ ID NO: 83) or PAAPSSWG (SEQ ID NO: 84).
  • the anticancer agent is a chemotherapeutic agent selected from the group consisting of taxanes, platinums, adriamycins, cylcophosphamide, topotecan, carmustine (BCNU) or a combination thereof.
  • the anti-cancer therapy is selected from the group consisting of paclitaxel, carboplatin, docetaxel, nab-paclitaxel, doxorubicin and any combination thereof.
  • the chemotherapeutic agent is selected from a group consisting of topoisomerase inhibitors (e.g., irinotecan, topotecan, doxorubicin, epirubicin, idarubicin), anti -microtubule agents (e.g., paclitaxel, docetaxel), alkylating agents (e.g., cyclophosphamide, dacarbizine), platinum-based drugs (cisplatin, carboplatin, oxaliplatin), anti-metabolites (e.g., gemcitabine, methotrexate, 5 -fluorouracil), or combinations thereof.
  • topoisomerase inhibitors e.g., irinotecan, topotecan, doxorubicin, epirubicin, idarubicin
  • anti -microtubule agents e.g., paclitaxel, docetaxel
  • alkylating agents e.g., cyclophos
  • the anticancer agent is doxorubicin.
  • the anti-cancer agent comprises paclitaxel.
  • the anti-cancer agent comprises carboplatin.
  • the anti-cancer agent comprises docetaxel.
  • the anti-cancer agent comprises nab-paclitaxel.
  • the anti-cancer agent comprises.
  • the anticancer agent is administered prior to the interval cytoreductive surgery every three weeks for about 12 weeks to about 18 weeks.
  • the anticancer agent is administered prior to the interval cytoreductive surgery every three weeks for about 12 weeks to about 18 weeks.
  • the anticancer agent is administered at least about 28 days after the interval cytoreductive surgery, every three weeks for about 9 weeks.
  • the anticancer agent is selected from the group consisting of paclitaxel, carboplatin, docetaxel, nab-paclitaxel, and any combination thereof.
  • the nucleic acid vector formulated with the lipopolymer is administered prior to, concurrently with, or after the anticancer agent.
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), and followed by administration of the immune checkpoint inhibitor, (e.g., third).
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), followed by the immune checkpoint inhibitor (e.g. third), and followed by a surgery to remove all or part of a tissue or tumor (e.g., interval cytoreductive surgery) (e.g., fourth).
  • a surgery to remove all or part of a tissue or tumor is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer is administered (e.g., second), and followed by administration of an immune checkpoint inhibitor (e.g., third, fourth, etc. depending on how many immune checkpoint inhibitors are administered).
  • an anticancer agent is administered, followed by the administration of a DNA plasmid, followed by the immune checkpoint inhibitor, followed by an interval cytoreductive surgery.
  • the anticancer agent is administered prior to the interval cytoreductive surgery every three weeks for about 12 weeks to about 18 weeks.
  • the anticancer agent is administered at least about 28 days after the interval cytoreductive surgery (e.g., every three weeks for about 9 weeks).
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV
  • the administration of the anticancer agent comprises administering docetaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of 25 - 350 mg/m 2 , optionally followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about 25 - 250 mg/m
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the nanoparticle prior to the interval cytoreductive surgery begins 15 days after the first administration of the anticancer agent, every week for at least about 12 weeks to about 18 weeks.
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 175 mg/m 2 , followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of about 75 mg/m 2 , followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of about 260 mg/m 2 , followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • Certain aspects of the disclosure are related to a method of treating a subject suffering from cancer comprising administering to the subject a combination of (i) a nanoparticle comprising an IL-2 coding sequence (e.g., a nucleic acid is a plasmid-based gene expression system containing a DNA sequence which encodes an interleukin- 12), and (ii) an immune checkpoint inhibitor.
  • the method further comprises administering a chemotherapeutic agent to the subject.
  • the method further comprises a surgery.
  • the nanoparticle comprises a PEG-PEI-chole sterol (PPC) lipopolymer.
  • This present disclosure also provides a method for treatment of mammalian cancer or hyperproliferative disorders by intratumoral, intraperitoneal, intravenously, intravesicularly, intratracheal, intracranial or systemic administration of pharmaceutical compositions comprising a plasmid-based gene expression system and a gene delivery polymer, without a chemotherapeutic drug.
  • the mammalian cancer is selected from a group consisting of primary or metastasized tumors of the ovary.
  • the nucleic acid is a plasmid-based gene expression system containing a DNA sequence which encodes human interleukin-12.
  • the treatment of tumors with the said pharmaceutical composition results in tumor shrinkage and extension of life span.
  • the combination of gene therapy (nucleic acid and gene delivery polymers) with chemotherapy (chemotherapeutic agents) according to the method of the present disclosure produce additive and/or synergistic efficacy.
  • the efficacy of the method of this invention is defined as but not limited to shrinkage in tumor size or reduction in tumor density, an increase in lymphocyte count or increase in neutrophil count or improvement in survival, or all of the above.
  • the combination of gene therapy (nucleic acid and gene delivery polymers) with chemotherapy (chemotherapeutic agents) lowers the toxicity of the chemotherapeutic agent and reverses tumor resistance to chemotherapy.
  • the toxicity herein is defined as any treatment related adverse effects on clinical observation including but not limited to abnormal hematology or serum chemistry or organ toxicity.
  • the combination of gene therapy (nucleic acid and gene delivery polymers) with a suboptimal dose of chemotherapy (chemotherapeutic agents) according to the method of the present invention enhances the anticancer effect to a level equal to or higher than that of achieved with the optimal dose of the chemotherapeutic agent but with lesser toxicity.
  • New cancer treatment strategies are focused on delivering macromolecules carrying genetic information, rather than a therapeutic protein itself, allowing for the exogenously delivered genes to be expressed in the tumor environment.
  • Methods that utilize non-viral gene delivery systems are considered safer compared to viral delivery systems, but the practical application of current polymeric systems has not been satisfactory due to poor efficiency.
  • a strategy has recently been disclosed whereby the gene transfection efficiency of a low molecular weight PEI was enhanced by covalent attachment of cholesterol forming a water soluble lipopolymer (WSLP). See, Mol. Ther., 2001, 4, 130. IL-12 gene transfer to solid tumors with WSLP was significantly better than by the unmodified PEI and led to more significant tumor inhibition.
  • WSLP water soluble lipopolymer
  • a single treatment strategy against cancer is generally ineffective due to the multi-factorial nature of this disease.
  • the methods disclosed herein provide a benefit from the combination of more than one drug or therapeutic to maximize anticancer response.
  • a chemotherapeutic agent and an immune checkpoint inhibitor with gene delivery of an anticancer IL- 12 expression construct (e.g., GEN-1).
  • an anticancer IL- 12 expression construct e.g., GEN-1
  • Combining safe and efficient delivery of an anticancer gene with a standard chemotherapeutic agent and an immune checkpoint inhibitor can enhance anticancer response.
  • This combination therapy can be used to reduce the chemotherapy dose and increase tumor sensitivity to the chemotherapy.
  • compositions disclosed herein can comprise an anticancer gene complexed with a gene delivery polymer, an immune checkpoint inhibitor, and at least one adjunctive chemotherapeutic drug, wherein the combination is more effective than gene therapy, immune checkpoint inhibitor, or chemotherapy treatment administered alone.
  • the combination therapy disclosed herein can be effective against a wide variety of tumors and given by different routes of administration.
  • CTLA4 blockade can significantly increase CD8 T cell infiltration of the tumor microenvironment (TME).
  • TME tumor microenvironment
  • CTLA4 and PD1 blockade mechanisms are non-redundant leading to enhanced immune and clinical responses in combination.
  • the majority of patients with solid tumors do not respond to these agents where the TME is considered less immunogenic where proinflammatory cytokines such as IL- 12 may play an important role in transforming the TME working synergistically with CTLA4 blockade and PD1 blockade.
  • IL- 12 has been shown to enhance the homeostatic proliferation of CD8+ T cells in vivo, and CD8+ T cells were significantly increased in biopsy specimens obtained from cancer patients treated with IL- 12.
  • IL-12 is a potent pro-inflammatory cytokine that promotes CD4+ cell differentiation into Thl cells and has synergistic proliferative effects on pre-activated NK and T cells. IL-12 also enhances independently and/or synergistically the cytolytic ability of both NK and CD8+ T cells. Antitumor activity of systemic or local administration of IL-12 has been established in preclinical studies against various tumor cell lines. However, the potent pro-inflammatory antitumor immune response mediated by IL- 12 may still be countered through tumor mediated immune suppressive mechanisms where PD-1 : PD-L interaction plays a key role. IL-12 induction of IFN-y and other proinflammatory cytokines is well documented.
  • IFNy is a pleotropic cytokine that has been reported to promote tumor immunity and tumor rejection in many models. In these models, IFNy has anti-proliferative and pro- apoptotic functions and inhibits tumor angiogenesis.
  • IFNy therapy has been associated with melanoma regression in some patients. Nevertheless, clinical trials in patients with melanoma using IFNy alone or in combination with IFNa failed to show improved survival compared with other therapies. Consistent with these findings, several studies have demonstrated that IFNy stimulation can induce up-regulation of PD-L 1 in monocytes, tumor cells and other cell types. In one study, expression of PD-L1 was increased in every cell line and primary cell incubated with IFNy.
  • Certain aspects of the disclosure are related to a combination therapy comprising: a (i) a nucleic acid vector (e.g., a plasmid) comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and (ii) an immune checkpoint inhibitor (e.g., an antibody).
  • a nucleic acid vector e.g., a plasmid
  • an immune checkpoint inhibitor e.g., an antibody
  • Certain aspects of the disclosure are related to a method of treating a subject suffering from cancer comprising administering to the subject a combination therapy comprising: (i) a nucleic acid vector (e.g., a plasmid) comprising a polynucleotide that encodes an interleukin- 12 (IL- 12) formulated with a lipopolymer (e.g., a nanoparticle); and (ii) an immune checkpoint inhibitor (e.g., an antibody).
  • a nucleic acid vector e.g., a plasmid
  • IL- 12 interleukin- 12
  • a lipopolymer e.g., a nanoparticle
  • an immune checkpoint inhibitor e.g., an antibody
  • nucleic acid vector (e.g., a plasmid) comprises a promoter operably linked to a nucleic acid encoding a p35 subunit of IL- 12 and a promoter operably linked to a nucleic acid encoding a p40 subunit of IL12.
  • nucleic acid vector (e.g., a plasmid) comprises an intron, a 3' UTR (e.g., hGH 3' UTR), an antibiotic resistance gene, or any combination thereof (e.g., the elements of FIG. 1).
  • a 3' UTR e.g., hGH 3' UTR
  • an antibiotic resistance gene e.g., the elements of FIG. 1.
  • the lipopolymer comprises of polyethyleneimine (PEI) covalently linked independently to cholesterol and polyethylene glycol (PEG) groups (e.g., the lipopolymer of FIG. 2).
  • PEI polyethyleneimine
  • PEG polyethylene glycol
  • the immune checkpoint inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 (and its ligands PD- L1 and PD-L2), and/or LAG-3.
  • the immune checkpoint inhibitor is an antibody.
  • the immune checkpoint inhibitor is a small molecule inhibitor.
  • the immune checkpoint inhibitor is a PD-1 antagonist selected from the group consisting of nivolumab, pembrolizumab, dostarlimab, and cemiplimab.
  • the immune checkpoint inhibitor is a PD-1 antagonist, wherein the PD-1 antagonist is nivolumab.
  • the immune checkpoint inhibitor is a PD-L1 antagonist selected from the group consisting of atezolizumab, durvalumab, and avelumab.
  • the immune checkpoint inhibitor is a CTLA-4 antagonist is ipilimumab.
  • the immune checkpoint inhibitor is a LAG-3 antagonist, wherein the LAG-3 antagonist is relatlimab.
  • the combination further comprises an anticancer agent.
  • the anticancer agent is a chemotherapeutic agent.
  • the chemotherapeutic agent is selected from the group consisting of doxorubicin, paclitaxel, carboplatin, docetaxel, nab-paclitaxel, olaparib, and any combination thereof.
  • the anticancer agent is doxorubicin.
  • the anticancer agent is paclitaxel.
  • the anticancer agent is carboplatin.
  • the anticancer agent is docetaxel. [0349] In some aspects, the anticancer agent is nab-paclitaxel.
  • the anticancer agent is olaparib.
  • the method further comprises a surgery to remove all or part of a tissue or tumor (e.g., interval cytoreductive surgery) in the subject.
  • a surgery to remove all or part of a tissue or tumor e.g., interval cytoreductive surgery
  • the nucleic acid vector formulated with the lipopolymer is administered intratum orally or intraperitoneally.
  • the nucleic acid vector formulated with the lipopolymer is administered intravenously.
  • the immune checkpoint inhibitor is administered intratumorally intraperitoneally, intravesicularly, or any combination thereof.
  • the immune checkpoint inhibitor is administered intratumorally or intraperitoneally.
  • the immune checkpoint inhibitor is administered intravenously.
  • the nucleic acid vector formulated with the lipopolymer is administered prior to, concurrently with, or after the immune checkpoint inhibitor.
  • the nucleic acid vector formulated with the lipopolymer is administered prior to, concurrently with, or after the anticancer agent.
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), and followed by administration of the immune checkpoint inhibitor (e.g., third).
  • an anticancer agent is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer (e.g., second), followed by the immune checkpoint inhibitor (e.g. third), and followed by a surgery to remove all or part of a tissue or tumor (e.g., interval cytoreductive surgery) (e.g., fourth).
  • a surgery to remove all or part of a tissue or tumor is administered (e.g., first), followed by the administration of the nucleic acid vector formulated with the lipopolymer is administered (e.g., second), and followed by administration of an immune checkpoint inhibitor (e.g., third, fourth, etc. depending on how many immune checkpoint inhibitors are administered).
  • an immune checkpoint inhibitor e.g., third, fourth, etc. depending on how many immune checkpoint inhibitors are administered.
  • an anticancer agent is administered, followed by the administration of a DNA plasmid, followed by the immune checkpoint inhibitor, followed by an interval cytoreductive surgery.
  • the anticancer agent is administered prior to the interval cytoreductive surgery every three weeks for about 12 weeks to about 18 weeks.
  • the anticancer agent is administered at least about 28 days after the interval cytoreductive surgery (e.g., every three weeks for about 9 weeks).
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6
  • the administration of the anticancer agent comprises administering paclitaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of 25 - 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering docetaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV
  • the administration of the anticancer agent comprises administering docetaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of 25 - 350 mg/m 2 , optionally followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of about 25 - 250 mg/m 2 , about 50 - 250 mg/m 2 , about 75 - 250 mg/m 2 , about 100 - 250 mg/m 2 , about 125 - 250 mg/m 2 , about 150 - 250 mg/m 2 , about 175 - 250 mg/m 2 , about 200 - 250 mg/m 2 , about 225 - 250 mg/m 2 , about 25 - 225 mg/m 2 , about 25 - 200 mg/m 2 , about 25 - 175 mg/m 2 , about 25 - 150 mg/m 2 , about 25 - 125 mg/m 2 , about 25 - 250 mg/m 2 , about 25 - 100 mg/m 2 , about 25 - 75 mg/m 2 , or about 25 - 50 mg/m 2 , optionally, followed by administering carboplatin at a dose of about 25 - 250 mg/m
  • the administration of the anticancer agent comprises administering nab-paclitaxel at a dose of about 25 mg/m 2 , about 50 mg/m 2 , about 75 mg/m 2 , about 100 mg/m 2 , about 125 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 200 mg/m 2 , about 225 mg/m 2 , or about 250 mg/m 2 , optionally, followed by administering carboplatin at a dose of about AUC 4-6 IV.
  • the administration of the nanoparticle prior to the interval cytoreductive surgery begins 15 days after the first administration of the anticancer agent, every week for at least about 12 weeks to about 18 weeks.
  • the nanoparticle is administered at least about 28 days following the interval cytoreductive surgery, and administration begins 15 days after the first administration of the anticancer agent, every week for at least about 9 weeks.
  • the interleukin- 12 (IL-12) formulated with a lipopolymer is administered at a dose of about 35 mg/m 2 to about 80 mg/m 2 .
  • the nanoparticle is administered at a dose of about 35 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 40 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 45 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 50 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 55 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 60 mg/m 2 to about 80 mg/m 2 .
  • the nanoparticle is administered at a dose of about 65 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 70 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 75 mg/m 2 to about 80 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 75 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 70 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 65 mg/m 2 .
  • the nanoparticle is administered at a dose of about 35 mg/m 2 to about 60 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 55 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 50 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 45 mg/m 2 . In some aspects, the nanoparticle is administered at a dose of about 35 mg/m 2 to about 40 mg/m 2 .
  • the nanoparticle is administered at a dose of about 35 mg/m 2 , about 40 mg/m 2 , about 45 mg/m 2 , about 50 mg/m 2 , about 55 mg/m 2 , about 60 mg/m 2 , about 65 mg/m 2 , about 70 mg/m 2 , about 75 mg/m 2 , or about 80 mg/m 2 .
  • the nanoparticle is administered at a dose of about 60 mg/m 2 .
  • the immune checkpoint inhibitor is ipilimumab.
  • ipilimumab is administered at a dose of about 0.5 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 1 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 1.5 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 2 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 2.5 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 3 to about 5 mg/kg.
  • ipilimumab is administered at a dose of about 3.5 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 4 to about 5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 4 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 3 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 2.5 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 2 mg/kg. In some aspects, ipilimumab is administered at a dose of about 0.5 to about 1.5 mg/kg.
  • ipilimumab is administered at a dose of about 0.5 mg/kg, about 1 mg/kg, about 1.5 mg/kg, about 2 mg/kg, about 2.5 mg/kg, about 3 mg/kg, about 3.5 mg/kg, about 4 mg/kg, about 4.5 mg/kg, or about 5 mg/kg.
  • ipilimumab is administered at a dose of about 1 mg/kg. [0383] In some aspects, ipilimumab is administered every 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.
  • ipilimumab is administered every 2-8 weeks (e.g., every 6 weeks) during the treatment.
  • ipilimumab is administered intravenously.
  • the immune checkpoint inhibitor is nivolumab.
  • nivolumab is administered at a dose of about 120 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 140 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 160 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 180 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 200 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 220 mg to about 360 mg.
  • nivolumab is administered at a dose of about 240 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 260 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 280 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 300 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 320 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 340 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 340 mg.
  • nivolumab is administered at a dose of about 120 mg to about 320 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 300 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 280 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 260 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 220 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 200 mg.
  • nivolumab is administered at a dose of about 120 mg to about 180 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 160 mg. In some aspects, nivolumab is administered at a dose of about 120 mg to about 140 mg. In some aspects, nivolumab is administered at a dose of about 240 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 260 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 280 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 300 mg to about 360 mg.
  • nivolumab is administered at a dose of about 320 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 340 mg to about 360 mg. In some aspects, nivolumab is administered at a dose of about 140 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 160 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 180 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 200 mg to about 240 mg. In some aspects, nivolumab is administered at a dose of about 220 mg to about 240 mg.
  • nivolumab is administered at a dose of about 120 mg, about 140 mg, about 160 mg, about 180 mg, about 200 mg, about 220 mg, about 240 mg, about 260 mg, about 280 mg, about 300 mg, about 320 mg, about 340 mg, or about 360 mg.
  • nivolumab is administered at a dose of about 240 mg.
  • nivolumab is administered intravenously.
  • nivolumab is administered every 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.
  • nivolumab is administered every 1-4 weeks (e.g., every 2 weeks) during treatment.
  • the method further comprises administering a second immune checkpoint inhibitor.
  • the second immune checkpoint inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 (and its ligands PD-L1 and PD-L2), and/or LAG-3.
  • the second immune checkpoint inhibitor is an antibody.
  • the second immune checkpoint inhibitor is a small molecule inhibitor.
  • the second immune checkpoint inhibitor is a PD-1 antagonist selected from the group consisting of nivolumab, pembrolizumab, dostarlimab, and cemiplimab.
  • the second immune checkpoint inhibitor is a PD-1 antagonist, wherein the PD-1 antagonist is nivolumab.
  • the second immune checkpoint inhibitor is a PD-L1 antagonist selected from the group consisting of atezolizumab, durvalumab, and avelumab.
  • the second immune checkpoint inhibitor is a CTLA-4 antagonist is ipilimumab.
  • the second immune checkpoint inhibitor is a LAG-3 antagonist, wherein the LAG-3 antagonist is relatlimab.
  • the first immune checkpoint inhibitor is nivolumab
  • the second immune checkpoint inhibitor is ipilimumab
  • the nivolumab is administered at about 240 mg every 2 weeks, and the ipilimumab is administered at about 1 mg/kg every 6 weeks.
  • the immune checkpoint inhibitor is administered prior to the interval cytoreductive surgery, at least about 22 days after the first administration of the anticancer agent, every week for at least about 12 weeks to up about 18 weeks.
  • the immune checkpoint inhibitor is administered at least about 28 days after the interval cytoreductive surgery, and at least about 22 days after the first administration of the anticancer agent, every week for at least about 9 weeks.
  • the interval cytoreductive surgery is administered at least about 28 days following the administration of an anticancer agent.
  • the interval cytoreductive surgery is administered at least about 7 days following the administration of the DNA plasmid.
  • interval cytoreductive surgery is administered at least about 7 days following the administration of the DNA plasmid.
  • the interval cytoreductive surgery is administered at least about 28 days before the administration of the immune checkpoint inhibitor.
  • the interval cytoreductive surgery is administered at least about 28 days following the administration of the immune checkpoint inhibitor.
  • the cancer is selected from a group consisting of ovarian cancer, fallopian tube cancer, primary peritoneal cancer, cervical cancer, breast cancer, prostate cancer, colorectal cancer, bladder cancer, brain cancer (e.g., glioblastoma), lung cancer, and any combination thereof, and metastasis of any of the cancers.
  • the cancer is selected from a group consisting of ovarian cancer, fallopian tube cancer, primary peritoneal cancer, and any combination thereof.
  • the subject is a human.
  • the cancer is ovarian cancer.
  • Ovarian cancer is the most lethal gynecologic cancer in the US.
  • the five- year overall survival rate is 20-30% in advanced stage OC, with more than 50% of patients that respond to current therapies experiencing recurrence of their disease.
  • platinum-resistant OC is characterized by only minimal responses to chemotherapy ( ⁇ 10- 15%) and a poor prognosis, with overall survival estimated to be ⁇ 12 months.
  • ICI immune checkpoint inhibitors
  • the impact of immune checkpoint inhibitors (ICI) has been significant, providing durable response rates in several cancers.
  • the response rates in ovarian cancer are low, ranging from 11-15% in platinum-resistant, recurrent settings.
  • dual immunotherapy with nivolumab and ipilimumab was associated with a higher objective response rate (31% vs. 12%) but with limited durability (3.9 vs. 2 months).
  • a combination strategy including IL- 12 gene therapy (GEN-1) and CTLA4-PD-1 blockade will be tested.
  • the mechanisms of action of known sites for activity for IL-12 and CTLA4-PD-1 blockade are non-redundant. Therefore, combination therapy of rhIL-12 and immune checkpoint inhibitors (ipilimumab and nivolumab) will be tested for anti-tumor activity. Further, the study will assess enhanced efficacy of current PD1/PD-L1 -based immunotherapy by targeting pathways that mediate resistance to ICI.
  • IL-12 has been shown to enhance the homeostatic proliferation of CD8+ T cells in vivo.
  • CD8+ T cells were increased in biopsy specimens obtained from regressing lesions of cancer patients treated with IL-12.
  • Correlative assays in phase I studies of IL-12 showed sustained production of IFN-y by immune cells of treated patients.
  • IFN-y a primary inducer of PD-L1 expression, was detected at the interface of PD-L1 (+) tumors and TILs, whereas none was found in PD-L1 (-) tumors.
  • PD-L1 expression has been associated with a higher likelihood of response to anti-PDl monoclonal antibodies and therefore, this effect of IL-12 of upregulating TIL in tumor and enhancing IFN-y secretion will be tested for increased PD- L1 expression and improved response.
  • Interleukin- 12 is one of the more active immunocytokines in the induction of anticancer immunity. It is associated with immunomodulatory properties such as T-lymphocyte and natural killer (NK) cell proliferation, activation of cytotoxic T-lymphocytes (i.e., CD8+ lymphocytes), secretion of IFN-gamma, and inhibition of immunosuppressive regulatory T-cells. IL-12 also inhibits the process of tumor angiogenesis, which leads to tumor death from starvation. These multiple and pluripotent anticancer properties make IL-12 a potentially important immunotherapy.
  • NK natural killer
  • the GEN-1 nanoparticle comprises a DNA plasmid encoding IL-12 gene and a synthetic polymer facilitating plasmid delivery.
  • GEN-1 is designed to be delivered locally (e.g. intraperitoneally), offering the potential for cytokines to be expressed specifically in the tumor micro-environment with the goal of achieving increased efficacy while minimizing potential systemic toxicity.
  • GEN-1 has been studied in subjects with recurrent ovarian cancer as a single agent or combination agent with standard chemotherapy. Additionally, a Phase I clinical trial in newly diagnosed ovarian cancer subjects has been completed where GEN-1 was evaluated as a combination agent with neoadjuvant chemotherapy.
  • IL-12 produced by GEN-1 will be tested to assess whether CTLA4 and/or PD1/PDL1 blocking monoclonal antibodies can overcome the immunosuppressive features of the peritoneal tumors by inducing an inflamed tumor microenvironment thus enhancing the efficacy of these checkpoint inhibitors.
  • Phase I To investigate the safety, tolerability (MTD, DLT) and phase 2 recommended dose (P2RD) of a combined regimen of GEN-1 with anti-PDl/CTLA4 (dual ICI) in subjects with recurrent or persistent ovarian tumors.
  • Phase II To determine the overall objective tumor response rate for the proposed combination regimen.
  • Phase I The safety, tolerability, and recommended phase 2 dose of the regimen will be evaluated by a Data and Safety Monitoring Board (DSMB). Details regarding their responsibilities will be elucidated in a DSMB charter which will be drafted for approval by the DSMB prior to initiation of the study.
  • DSMB Data and Safety Monitoring Board
  • Phase II The overall objective response rate will be determined by using RECIST 1.1
  • PFS progression free survival
  • OS overall survival
  • ORR objective tumor response rate
  • PFS will be measured by time from enrollment to the first date on which one of the following occurs: i) Death by any cause; or ii) Unequivocal progression of target lesions, non-target lesions and/or new lesions by Response Evaluation Criteria in Solid Tumors (RECIST).
  • OS will be measured by time from enrollment to the date of death
  • ORR will be measured according to Response Evaluation Criteria for Solid tumors guidelines version 1.1 (RECIST). This protocol allows for participation of subjects with both measurable and non-measurable disease, and as such, will deviate slightly from the standard RECIST as described later in the protocol.
  • RECIST Response Evaluation Criteria for Solid tumors guidelines version 1.1
  • the dual ICI regimen will be the anti-PDl nivolumab and anti-CTLA4 ipilimumab.
  • Nivolumab 240 mg IV
  • GEN-1 starting dose 60 mg/m 2
  • Ipilimumab will be administered 1 mg/kg IV every 6 weeks. Dosing will continue until disease progression, unacceptable toxicity, or up to 2 years in patients without disease progression.
  • MTD Maximum Tolerated Dose
  • the Phase II portion of the study is a two-stage design to evaluate if the treatment will yield a objective response rate (ORR) of 45% or higher. Twenty (20) patients will be enrolled in stage 1 and evaluate whether six (6) or more subjects achieve a CR or PR. If not, the study will be stopped for futility. If the stage 1 ORR target rate is met then twenty (20) additional subjects will be enrolled to test whether at least 14 of 40 subjects achieve CR or PR to confirm the confirm the response rate in this phase 2 design.
  • ORR objective response rate
  • a 3+3 dose escalation will be employed. All subjects will be monitored for safety from time of signing informed consent. Safety data from subjects who complete 2 cycles of protocol treatment is required to be eligible for dose escalation review. In general, dose escalation may occur at 0/3 DLTs, dose-reduction if >1/3 DLT, and expansion to 6 if 1/3 DLTs. The highest dose level at which ⁇ 2/6 DLTs are observed will be declared the MTD. Data will be reviewed by the study DSMB and recommend dosing modifications and the phase 2 recommended dose. During the phase I portion the dual ICI's will be given at a fixed dose at all dose levels and Gen-1 dose will be escalated according to the table presented below.
  • Nivolumab 240 mg IV will be administered with GEN-1 at (at DSMB recommended dose) every 2 weeks until disease progression, unacceptable toxicity, or up to 2 years in patients without disease progression.
  • Ipilimumab 1 mg/kg IV will be administered every 6 weeks until disease progression, unacceptable toxicity, or up to 2 years in patients without disease progression
  • Human IL-12 plasmid (phIL-12-005) is formulated with lipopolymer PEG-PEI- Cholesterol (PPC) in 10% lactose.
  • PPC lipopolymer PEG-PEI- Cholesterol
  • the phIL- 12-005 plasmid contains hIL-12 gene expression cassette in a plasmid containing a Kanr gene.
  • the hIL-12 gene expression cassette of phIL- 12-005 contains immediate early enhancer and promoter derived from cytomegalovirus (CMV), 5' untranslated region (UTR), synthetic intron, p35 gene, human growth hormone (hGH) 3' UTR and polyadenylation signal sequence, CMV promoter, 5' UTR, synthetic intron, p40 gene, hGH 3' UTR and polyadenylation signal sequence.
  • CMV cytomegalovirus
  • UTR 5' untranslated region
  • synthetic intron p35 gene
  • human growth hormone (hGH) 3' UTR and polyadenylation signal sequence CMV promoter
  • 5' UTR synthetic intron
  • p40 gene hGH 3' UTR and polyadenylation signal sequence.
  • the two hIL-12 subunits are individually under the control of two separate
  • PPC is composed of a PEI backbone to which polyethyleneglycol and cholesterol are independently attached via covalent linkages.
  • the molecular weight of PEI, PEG and cholesterolcarbonyl is 1800, 550 and 414, respectively. See FIG. 2.
  • Reconstituted GEN-1 (in the 50 mL glass vial or IV bag) is stable at room temperature for up to 24 hours. Upon confirmation of catheter patency, an IV bag containing the GEN-1 will be administered through the patient's IP catheter. GEN-1 will be infused via a catheter through gravity from the IV bag with the valve all the way open and free flowing. Typical administrations may take about 1 hour.
  • Subcutaneously implantable IP silicone catheters may be used to administer GEN- 1 to the peritoneal cavity.
  • GEN-1 has been demonstrated to be compatible with silicone catheters in a prior preclinical compatibility study and in prior phase I studies.
  • Port-A-Cath catheter (Deltec, Inc. St. Paul, MN) has been successfully used for IP delivery of GEN-1 with little to no catheter-related serious complications noted. Any other approved catheter with a subcutaneous port for IP delivery may also be used if suitable for aspiration of biological samples for translational studies.
  • Bard 9.6 Fr silicone single lumen catheters for venous access or the equivalent with or without cuff from Bard Access Systems (West Amelia Earhart Drive Salt Lake City Utah) may be used.
  • Port-A-Cath catheter The catheter compatibility studies conducted with Port-A-Cath catheter showed that catheter exposure to GEN-1 does not significantly affect the physico-chemical properties or transfection activity of GEN-1.
  • the IP catheter will be implanted per hospital standard process; the procedure and risks associated with placement of the IP catheter must be explained to the subject and an operative consent form will be obtained from the subject prior to placement of the catheter.
  • the subject will undergo insertion of the IP catheter a minimum of 7 days prior to scheduled study drug administration to allow for sufficient healing and sealing around the catheter site.
  • a semi- permanent subcutaneous access port such as the Port-A-Cath catheter, (SIMS Deltec, Inc, St. Paul MN Inc., 55112) or equivalent device per current institutional clinical practice will be used.
  • Study drug will be infused through this port during the course of the study. Prior to each infusion of study drug, approximately 25 mL of saline will be flushed through the catheter to check for catheter patency; heparin should not be used to flush the catheter at the time of sample collection or drug infusion. At end of study, the catheter may be removed at completion of GEN-1 administration at the clinician's discretion.
  • the calculated dose of GEN-1 is based on the protocol specified dose (in mg/m2) and the subject's body BSA calculated at baseline.
  • the subject's weight should be monitored during treatment (e.g., typically weight is monitored/recorded on the same day of treatment). If the subject's weight changes by 10%, then the BSA is recalculated and the adjusted dose will be administered during the subsequent treatment cycles. The new BSA becomes the baseline for future weight and any dose modifications.
  • nivolumab When nivolumab is administered in combination with ipilimumab, withhold or permanently discontinue both ipilimumab and nivolumab for an adverse reaction meeting these dose modification guidelines.
  • Each cycle of study treatment will be defined as being 6 weeks. On day 1 of each cycle all subjects will receive a fixed dose of the dual ICI (nivolumab and ipilimumab) followed by the prescribed dose of Gen-1. Dosing by cycle is presented in the table below:
  • Subjects will be monitored for safety (with physical exams and assessment of AEs) at every treatment visit from the time of signing informed consent until at least 30 days following their last dose of study drug. Any suspected drug related adverse events may be reported at any time during follow up until resolution to a grade 2 (CTCAE v5.0).
  • the primary end point is objective tumor response (complete or partial) by RECIST, version 1.1,16 prior to progression will be used as a method to evaluate efficacy.
  • Measurable disease Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter to be recorded) as 20 mm by chest x- ray, as 10 mm with CT scan, 10 mm with calipers by clinical exam. All tumor measurements must be recorded in millimeters (or decimal fractions of centimeters).
  • Tumor lesions that are situated in a previously irradiated area will not be considered measurable unless progression is documented, or a biopsy is obtained to confirm persistence at least 90 days following completion of radiation therapy.
  • lymph nodes To be considered pathologically enlarged and measurable, a lymph node must be 15 mm in short axis when assessed by CT scan (CT scan slice thickness recommended to be no greater than 5 mm). At baseline and in follow-up, only the short axis will be measured and followed.
  • CT scan CT scan slice thickness recommended to be no greater than 5 mm.
  • Non-measurable disease All other lesions (or sites of disease), including small lesions (longest diameter ⁇ 10 mm or pathological lymph nodes with 10 to ⁇ 15 mm short axis), are considered non-measurable disease. Leptomeningeal disease, ascites, pleural/pericardial effusions, lymphangitis cutis/pulmonitis, inflammatory breast disease, and abdominal masses (identified by physical exam and not CT or MRI), are considered as non- measurable. [0480] Bone lesions: Lytic bone lesions or mixed lytic-blastic lesions, with identifiable soft tissue components, that can be evaluated by CT or MRI can be considered as measurable lesions if the soft tissue component meets the definition of measurability described above. Blastic bone lesions are non-measurable.
  • Cystic lesions that meet the criteria for radiographically defined simple cysts should not be considered as malignant lesions (neither measurable nor non-measurable) since they are, by definition, simple cysts.
  • 'Cystic lesions' thought to represent cystic metastases can be considered as measurable lesions, if they meet the definition of measurability described above. However, if non-cystic lesions are present in the same patient, these are preferred for selection as target lesions.
  • Target lesions All measurable lesions up to a maximum of 2 lesions per organ and 5 lesions in total, representative of all involved organs, should be identified as target lesions and recorded and measured at baseline. Target lesions should be selected on the basis of their size (lesions with the longest diameter), be representative of all involved organs, but in addition should be those that lend themselves to reproducible repeated measurements. It may be the case that, on occasion, the largest lesion does not lend itself to reproducible measurement in which circumstance the next largest lesion which can be measured reproducibly should be selected. A sum of the diameters (longest for non-nodal lesions, short axis for nodal lesions) for all target lesions will be calculated and reported as the baseline sum diameters. If lymph nodes are to be included in the sum, then only the short axis is added into the sum. The baseline sum diameters will be used as reference to further characterize any objective tumor regression in the measurable dimension of the disease.
  • Non-target lesions All other lesions (or sites of disease) including any measurable lesions over and above the 5 target lesions should be identified as non-target lesions and should also be recorded at baseline. Measurements of these lesions are not required, but the presence, absence, or in rare cases unequivocal progression of each should be noted throughout follow-up.
  • Phase I Safety (DLT's and MTD), 3+3 design in 12 to 18 patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Mycology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une polythérapie comprenant un inhibiteur de point de contrôle immunitaire, un plasmide formulé dans des nanoparticules contenant un acide nucléique codant pour IL-12, et, facultativement, au moins un médicament chimiothérapeutique auxiliaire, ainsi que des méthodes de traitement utilisant de telles polythérapies et/ou compositions.
PCT/US2023/074062 2022-09-13 2023-09-13 Combinaison de thérapie génique utilisant l'il-12 et d'inhibiteurs de point de contrôle immunitaire pour le traitement du cancer WO2024059629A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263375529P 2022-09-13 2022-09-13
US63/375,529 2022-09-13

Publications (1)

Publication Number Publication Date
WO2024059629A1 true WO2024059629A1 (fr) 2024-03-21

Family

ID=90275854

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/074062 WO2024059629A1 (fr) 2022-09-13 2023-09-13 Combinaison de thérapie génique utilisant l'il-12 et d'inhibiteurs de point de contrôle immunitaire pour le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2024059629A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190183936A1 (en) * 2016-08-26 2019-06-20 Baylor College Of Medicine Constitutively active cytokine receptors for cell therapy
US20200102363A1 (en) * 2017-05-18 2020-04-02 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190183936A1 (en) * 2016-08-26 2019-06-20 Baylor College Of Medicine Constitutively active cytokine receptors for cell therapy
US20200102363A1 (en) * 2017-05-18 2020-04-02 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof

Similar Documents

Publication Publication Date Title
CN108368174B (zh) 用于癌症治疗的单独fgfr2抑制剂或与免疫刺激剂的组合
EP3324988B1 (fr) Utilisation d'un virus herpes simplex oncolytique en combinaison avec un inhibiteur de point de contrôle immunitaire, dans le traitement du cancer
US20180128833A1 (en) Methods of treating with tumor membrane vesicle-based immunotherapy and predicting therapeutic response thereto
CN113939309A (zh) 使用sEphB4-HSA融合蛋白治疗癌症
JP2019510769A (ja) チェックポイント阻害剤と組み合わされた腫瘍溶解性ウイルスによるがん療法
KR20210093950A (ko) Il-7 단백질 및 면역 관문 저해제의 조합으로 종양을 치료하는 방법
US20140205609A1 (en) Methods for inducing systemic immune responses to cancer
US20210196744A1 (en) Compositions for cancer therapy and methods
WO2024059629A1 (fr) Combinaison de thérapie génique utilisant l'il-12 et d'inhibiteurs de point de contrôle immunitaire pour le traitement du cancer
US20240101666A1 (en) Lag-3 antagonist therapy for lung cancer
WO2024059630A2 (fr) Thérapie génique de l'il-12 pour le traitement de cancers brca-négatif/aptes à la réparation homologue
EP4090675A1 (fr) Agent de liaison à ccr5 pour le traitement du cancer du sein métastatique ccr5-positif
US20230365701A1 (en) Anti-lymphotoxin beta receptor antibodies and methods of use thereof
WO2024054855A1 (fr) Thérapie génique de l'il-12 et combinaison anti-vegf pour le traitement du cancer
WO2023164638A1 (fr) Polythérapie pour carcinome colorectal
WO2023147371A1 (fr) Polythérapie pour carcinome hépatocellulaire
US20230265188A1 (en) Lag-3 antagonist therapy for hepatocellular carcinoma
KR20240099362A (ko) 혈액암에 대한 lag-3 길항제 요법
CN118251234A (zh) 抗半乳糖凝集素-9抗体及其治疗用途
WO2023076993A1 (fr) Méthodes de traitement d'un lymphome au moyen d'un phagocyte possédant un récepteur antigénique chimérique
WO2024137776A1 (fr) Polythérapie contre le cancer du poumon
WO2021195589A1 (fr) Méthode de traitement d'un cancer muc4+ résistant au traitement
US20220411499A1 (en) LAG-3 Antagonist Therapy for Melanoma
CN116133679A (zh) 白血病衍生细胞在卵巢癌疫苗中的用途

Legal Events

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

Ref document number: 23866429

Country of ref document: EP

Kind code of ref document: A1