WO2024077228A2 - Double-stranded oligonucleotides and methods of use - Google Patents
Double-stranded oligonucleotides and methods of use Download PDFInfo
- Publication number
- WO2024077228A2 WO2024077228A2 PCT/US2023/076223 US2023076223W WO2024077228A2 WO 2024077228 A2 WO2024077228 A2 WO 2024077228A2 US 2023076223 W US2023076223 W US 2023076223W WO 2024077228 A2 WO2024077228 A2 WO 2024077228A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- dna oligonucleotide
- constrained
- nucleotides
- nucleotide
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/113—Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7105—Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [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
- C07K16/2818—Immunoglobulins [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
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/11—DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
- C12N15/117—Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/10—Type of nucleic acid
- C12N2310/11—Antisense
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/30—Chemical structure
- C12N2310/34—Spatial arrangement of the modifications
- C12N2310/341—Gapmers, i.e. of the type ===---===
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2310/00—Structure or type of the nucleic acid
- C12N2310/50—Physical structure
- C12N2310/53—Physical structure partially self-complementary or closed
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/30—Special therapeutic applications
- C12N2320/31—Combination therapy
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2320/00—Applications; Uses
- C12N2320/50—Methods for regulating/modulating their activity
- C12N2320/51—Methods for regulating/modulating their activity modulating the chemical stability, e.g. nuclease-resistance
Definitions
- the immune system can serve as extrinsic tumor suppressor.
- the microenvironment of established tumors is typically characterized by a paucity of tumor-specific CD8 + T cells together with an excess of suppressive regulatory T cells and myeloid-denved suppressor cells that promote tumor immune evasion.
- Myeloid cells and other immune cells in the tumor microenvironment also produce growth factors and angiogenic/metastatic factors critical for tumor progression.
- Signal Transducer and Activator of Transcription 3 STAT3 is an important oncogenic molecule.
- STAT3 plays a critical role in mediating tumor immune evasion.
- STAT3 in myeloid cells inhibits expression of a large number of immunostimulatory molecules related to Thl-type responses, while promoting production of several key immunosuppressive factors as well as angiogenic factors.
- certain cytokines and grow th factors, notably IL-6 STAT3 activation in myeloid cells activates STAT3 in tumor cells, enhancing tumor cell proliferation and survival.
- a phosphorothioated CpG oligodeoxynucleotide (ODN) linked to a first DNA oligonucleotide, wherein the first DNA oligonucleotide is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides, and the second DNA oligonucleotide comprises: (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3‘ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- kits for treating cancer by administering to a patient an effective amount of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor, such as a PD-1 pathway inhibitor.
- FIGS. 1A-1D show serum stability and activity of the double-stranded antisense oligonucleotides.
- FIG. 1A Double-stranded STAT3ASO shows greater serum stability than single-stranded oligonucleotides. Oligonucleotides before and after incubation in 50% human serum were resolved on 15% PAGE.
- FIGS. 1B-1C Double-stranded STAT3- (FIGS. IB, ID) or androgen receptor (AR)-specific ASOs show comparable or improved knockdown of their respective targets compared to their respective single-stranded ASOs within 48h after transfection (FIGS. 1B-1C; 100 ng each) or spontaneous/gymnotic uptake (FIG.
- STAT3ASO is SEQ ID NO:4; STAT3 dsASO is SEQ ID NO:3 hybridized to SEQ ID NO:4; AR-ASO is 5’ accAAGTTTCTTCagc (SEQ ID NO:35, where bold-lower case indicates LNA-modified nucleotide) (described by Zhang et al, Nucleic Acids Research, 46(7):3612-3624 (2016); and ds-AR-ASO (SEQ ID NO:35) hybridized to 5’gctgaagaaacttggt (SEQ ID NO:36)
- FIGS. 2A-2D show design, serum stability and in vitro activity of the CpG-conjugated double-stranded STAT3 antisense oligonucleotides.
- FIG. 2A Schematic design of the doublestranded CpG-STAT3dsASO.
- FIG. 2B Serum stability' of the double-stranded CpG- STAT3dsASO compared to each separate oligonucleotide strand (ASO alone and CpG- passenger strand). Oligonucleotides were incubated in 50% human serum at 37°C for the indicated times and then resolved on 15% PAGE.
- FIGS. 1A Schematic design of the doublestranded CpG-STAT3dsASO.
- FIG. 2B Serum stability' of the double-stranded CpG- STAT3dsASO compared to each separate oligonucleotide strand (ASO alone and CpG- passenger strand). Oligonucleotides were in
- Double-stranded CpG- STAT3dsASO retains target knockdown potency and kinetics comparable with STAT3ASO alone.
- Human A431 (FIG. 2C) and mouse GL261 cells (FIG. 2D) were incubated for various times with 1 pM of the indicated oligonucleotides.
- CpG- STAT3 dsASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; CpG-passenger is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3; and STAT3ASO is SEQ ID NO:4; wherein the moiety of Formula (A)(in this figure and throughout the application) is:
- FIGS. 3A-3F show that CpG-STAT3dsASO shows better potency at target gene knockdown than single-stranded CpG-STAT3ASO variants in different human and mouse target cells.
- Human prostate cancer cells DU145, LAPC4, and ENZR42D
- human U251
- mouse GL261 glioma cells
- mouse macrophages RAW264.7
- CpG-STAT3ASO (2'OMe) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:33; CpG-passenger (LNA) SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO: 3; STAT3ASO (LNA) is SEQ ID NO:4; CpG-STAT3 dsASO (LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and CpG-STAT3 ssASO (LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 4A-4F provide a comparison of time- and dose-dependent ASO uptake by human and mouse target cells.
- Human U251, LN229, T98G
- mouse GL261, K-luc glioma cells
- BV2 mouse microglia cell line
- FIGS. 4A-4F were incubated for 1 h or 4 h with fluorescently-labeled double-stranded CpG-STAT3dsASO Cy3 or with single-stranded CpG-STAT3ssASO Cy3 or unconjugated STAT3ssASO Cy3 at 100 nM or 500 nM concentrations.
- STAT3ASO is SEQ ID NO:4;
- CpG-STAT3 dsASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and
- CpG-STAT3 ssASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 5A-5E show a comparison of time- and dose-dependent ASO uptake by primaryhuman immune cells.
- Human PBMCs were incubated for 1 h or 4 h with fluorescently-labeled double-stranded CpG-STAT3dsASO Cy3 or with single-stranded CpG-STAT3ssASO Cy3 or unconjugated STAT3ssASO Cy3 at 100 nM or 500 nM concentrations.
- the oligonucleotide internalization was assessed using flow cytometry. Shown are histograms indicating oligonucleotide uptake by primary human CDlc + myeloid dendritic cells (mDC) (FIG.
- mDC myeloid dendritic cells
- STAT3ASO is SEQ ID NO:4;
- CpG-STAT3 dsASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and
- CpG-STAT3 ssASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 6A-6D show a comparison of dose-dependent ASO uptake by primary mouse immune cells.
- Mouse splenocytes were incubated for 4 h with fluorescently-labeled doublestranded CpG-STAT3dsASO Cy3 or with single-stranded CpG-STATSssASO 1 ⁇ 3 or unconjugated STAT3ssASO Cy3 at 100 nM or 500 nM concentrations.
- the oligonucleotide internalization was assessed using flow cytometry. Shown are histograms indicating oligonucleotide uptake by CD1 lb + CDl lc- macrophages (FIG.
- STAT3ASO is SEQ ID NO:4; CpG-STAT3 dsASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3. wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and CpG- STAT3 ssASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 7A-7F show biodistribution of the intratumorally injected CpG-STAT3ASOs in mouse brain.
- C57/BL6 mice bearing intracranial GL261 glioma tumors were injected with 1 mg/kg of single- or double-stranded CpG-STAT3ASO Cy3 or with unconjugated STAT3ASO Cy3 .
- Mice were euthanized 3 days or 5 days later to harvest brains.
- Percentages of Cy3 + Ml-like macrophages (CD1 lb'CD45 l "g ll CD86' ) (FIG. 7A).
- M2-like macrophages CD1 lb'CD45 l "g ll CD86'
- CpG-STAT3ASO (2'0Me) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:33;
- STAT3ASO-h/m#3(LNA) is SEQ ID NO:4;
- CpG-STAT3ASO h/m#3(LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 8A-8F show that double-stranded CpG-STAT3dsASO is safer and better tolerated by naive mice than single-stranded CpG-STAT3ssASO.
- FIG. 8A Determination of maximum tolerated dose (MTD) of CpG-STAT3ASO variants in naive mice. C57BL/6 mice were injected intracranially using 0.1, 0.3. or 1 mg/kg of CpG-STAT3 dsASO or CpG- STAT3ssASO twice a week and euthanized 2 weeks later. Body weight measurements for each dosing group; shown are means ⁇ SEM.
- FIGS. 8B-8C Mouse behavior assessed using acute tolerability scoring system (ATSS) (FIG. 8B) is altered more significantly by low doses of the single-stranded but not by double-stranded CpG-STAT3ASO (FIG. 8C). The assessment of acute CNS toxicity based on the mice phenotypic behavior at I and 4 h after IC injection of CpG-STAT3ASOs.
- FIGS. 8D-8F Single- but not double-stranded CpG-STAT3ASO induces significant changes in leukocytes and platelets in treated mice. Hematological parameters such as red blood cells (RBC), white blood cells (WBC) and platelets (PLT) assessed in blood collected from mice treated as described above.
- RBC red blood cells
- WBC white blood cells
- PLT platelets
- dsASO is SEQ ID NO: 15 linked via the moiety' of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and ssASO is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 9A-9B show local administration of LNA-modified single- or double-stranded CpG-STAT3ASOs both lead to similar direct antitumor effects against intracranial human glioma xenotransplants in immunodeficient mice.
- Human U251 glioma-bearing NSG mice were injected intratumorally/IC using 1 mg/kg of 2’O-methyl- or LNA-modified CpG-STAT3ASOs every other day as indicated by red arrows.
- FIG. 9A Tumor progression and occasionally extracranial spread was monitored using bioluminescent imaging (BLI).
- CpG-STAT3ASO (2’0Me) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO: 34;
- CpG-STAT3dsASO (LNA) is SEQ ID NO:43 linked via the moiety of Formula (A) to SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to SEQ ID NO:2;
- CpG-STAT3-ssASO (LNA) is SEQ ID NO:43 linked via the moiety of Formula (A) to SEQ ID NO:2.
- FIGS. 10A-10I show local administration of single- and double-stranded CpG- STAT3ASOs improves animal survival and triggers immune activation in immunocompetent mice.
- C57BL/6 mice bearing established GL261 gliomas were injected intratumorally/IC using 0.25 mg/kg of 2’-O-methyl-or LNA-modified CpG-STAT3ASOs twice weekly.
- FIGS. 10B-10F All three types of CpG-STAT3ASO injections induced maturation/activation of intratumoral DCs.
- FIGS. 1 & 2 Show n are percentages of mature DCs (CD1 lb + CDl lc + MHCII + CD80 + ), mature macrophages (CD1 lb + CDl lc’MHCII+CD80+MHCII + CD80 + ), M2 -like macrophages (CD 1 lb 1 F4/80 1 CD206 1 ), activated microglia (CD 11 b 1 CD45 low MHCII 1 CX3CR1 1 ) and resting microglia (CDl lb + CD45 low MHCirCX3CRl + ).
- 10G-10I CpG-STAT3ASO injections improve the ratio of intratumoral CD8 T cells to Tregs. Shown are percentages of CD8 + T cells (CD3 + CD8 + ), regulatory T cells (CD3 + CD4 + FOXP3 + ), and their ratio. Data are presented as means ⁇ SEM; *P ⁇ 0.05, **P ⁇ 0.01, *** ⁇ 0.001 by one-way ANOVA with Tukey’s multiplecomparison post hoc test. With reference to FIGS.
- CpG-STAT3ASO (2’OMe) is SEQ ID NO:15 linked via the moiety of Formula (A) to SEQ ID NO:33;
- CpG-STAT3dsASO (LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, which is hybridized to SEQ ID NO:4;
- CpG-STAT3-ssASO (LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- FIGS. 11A-11F show the combination of CpG-STAT3dsASO with PD1 blockade results in glioma regression in the majority of treated mice.
- C57BL/6 mice bearing established GL261 gliomas were injected twice weekly using intraperitoneal injections of 200 pg of PD1- specific or control antibodies, using intratumoral/IC injections of 0.25 mg/kg of LNA-modified CpG-STAT3dsASO or both treatments combined.
- FIG. 11A-11E Tumor progression was monitored using BLI.
- IgG is polyclonal rat IgG (Bio X Cell, Cat: BE0094);
- Anti-PD-1 is anti-mouse PD-1 antibody (from Bio X Cell, Clone:29F. lA12, Cat: BE0273);
- CpG-STAT3 dsASO (LNA) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, which is hybridized to SEQ ID NO:4.
- FIG. 12 shows that double-stranded CpG-STAT3dsASO (bottom right panel) has greater potency than single-stranded CpG-STAT3ssASO (top right panel) or CpG-psODN (CpG passenger strand only) (bottom left panel) against RM9 prostate tumors.
- C57BL/6 mice bearing established, dual RM9 tumors were treated using local intratumoral injection of indicated oligonucleotide. Tumor growth progression was monitored by caliper measurement at the secondary, not injected tumor site; (n 4-5/group).
- FIG. 13 shows that local administration of LNA-modified single- or double-stranded CpG-STAT3ASOs improved survival in mice bearing syngeneic intracranial QPP8 glioma.
- C57BL/6 mice bearing established QPP8 gliomas Qk/Tp53/Pten-deY
- the median survival for PBS was 49 days.
- the median survival for LNA-modified STAT3 ASO was 52 days.
- FIG. 14 shows that the combination of CpG-STAT3dsASO ith PD1 blockade resulted in QPP8 glioma regression in the majority of treated mice.
- C57BL/6 mice bearing established QPP8 gliomas were injected twice weekly using Intraperitoneal/IP using 200pg of PDl-specific antibodies, using intracranially /IC of 0.25mg/kg of LNA-modified CpG- STAT3ASOs or both treatment combined.
- FIG. 15 shows that surviving mice were protected from rechallenge with the same tumor.
- Mice surviving after the combination anti-PDl/CpG-STAT3dsASO treatment were rechallenged using intracranial injection with 10 4 GL261 cells and the tumor progression was monitored using bioluminescent imaging (BLI).
- Most of the previously treated survivor mice but not naive mice survived glioma rechallenge; shown are Kaplan-Meier survival curves (n 4- 5/group).
- the median survival for the mice receiving the combination anti-PDl and LNA-modified CpG-STAT3dsASO treatment was more than 152 days.
- LNA-modified STAT3 ASO is SEQ ID NO:4; LNA-modified CpG-STAT3-double stranded ASO (CpG-STAT3dsASO LNA ) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to SEQ ID NO:4; and LNA-modified CpG-STAT3-single stranded ASO (CpG- STAT3SSASO LNA ) is SEQ ID NO: 15 linked via the moiety of Formula (A) to SEQ ID NO:4.
- CpG oligodeoxynucleotide or “CpG ODN” refers to a 5’ C nucleotide connected to a 3’ G nucleotide through a phosphodiester intemucleotide linkage or a phosphodiester derivative intemucleotide linkage.
- a CpG ODN includes a phosphodiester intemucleotide linkage.
- a CpG ODN includes a phosphodiester derivative intemucleotide linkage.
- Class A CpG ODN'’ or “A-class CpG ODN” or “D-type CpG ODN” or “Class A CpG DNA sequence” refers to a CpG motif including oligodeoxynucleotide including one or more of poly-G sequence at the 5’, 3’, or both ends; an internal palindrome sequence including CpG motif; or one or more phosphodiester derivatives linking deoxynucleotides.
- a Class A CpG ODN includes poly-G sequence at the 5’, 3’, or both ends; an internal palindrome sequence including CpG motif; and one or more phosphodiester derivatives linking deoxynucleotides.
- the phosphodiester derivative is phosphorothioate
- Examples of Class A CpG ODNs include ODN D19, ODN 1585, ODN 2216, and ODN 2336, the sequences of which are known in the art.
- Class B CpG ODN or “B-class CpG ODN” or “K-type CpG ODN” or “Class B CpG DNA sequence” refers to a CpG motif including oligodeoxynucleotide including one or more of a 6mer motif including a CpG motif; phosphodiester derivatives linking all deoxynucleotides.
- a Class B CpG ODN includes one or more copies of a 6mer motif including a CpG motif and phosphodiester derivatives linking all deoxynucleotides.
- the phosphodiester derivative is phosphorothioate.
- a Class B CpG ODN includes one 6mer motif including a CpG motif. In embodiments, a Class B CpG ODN includes two copies of a 6mer motif including a CpG motif. In embodiments, a Class B CpG ODN includes three copies of a 6mer motif including a CpG motif. In embodiments, a Class B CpG ODN includes four copies of a 6mer motif including a CpG motif. Examples of Class B CpG ODNs include ODN 1668, ODN 1826, ODN 2006, ODN 2007, ODN BW006, and ODN D-SL01, the sequences of which are known in the art.
- Class C CpG ODN “C-class CpG ODN” “ or “C-type CpG DNA sequence” refers to an oligodeoxynucleotide including a palindrome sequence including a CpG motif and phosphodiester derivatives (phosphorothioate) linking all deoxynucleotides.
- Class C CpG ODNs include ODN 2395, ODN M362, and ODN D-SL03, the sequences of which are known in the art.
- STAT refers to a “Signal transducer and activator of transcription” protein and homologs thereof (e.g. STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, STAT6, STAT7, STAT8, STAT7/8, STAT9).
- STAT transcription factor refers to a human protein. Included in the term “STAT transcription factor” are the w ildtype and mutant forms of the protein.
- STAT transcription factor refers to the wildtype protein.
- STAT transcription factor refers to a mutant protein.
- Phosphorylated STAT refers to a STAT protein that is phosphorylated and activated by the phosphorylation. In embodiments, activation of a STAT transcription factor means the STAT is capable of activating transcription.
- a “STAT3” or “STAT3 protein” refers to any of the recombinant or naturally- occurring forms of the Signal transducer and activator of transcription 3 (STAT3) protein or variants or homologs thereof that maintain STAT3 protein activity (e g. within at least 50%, 80%. 90%. 95%. 96%. 97%. 98%. 99% or 100% activity compared to STAT3).
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% amino acid sequence identity across the w hole sequence or a portion of the sequence (e.g. a 50, 100, 150 or 200 continuous amino acid portion) compared to a naturally occurring STAT3 polypeptide.
- the STAT3 protein is substantially identical to the protein identified by the NCBI reference number GI: 47458820, or a variant or homolog having substantial identity thereto. In embodiments, the STAT3 protein is substantially identical to the protein identified by the NCBI reference number GI: 1610577068, or a variant or homolog having substantial identity thereto. In embodiments, the STAT3 protein is substantially identical to the protein identified by the NCBI reference number GI: 1610577050. or a variant or homolog having substantial identity thereto. “Phosphorylated STAT3” refers to a STAT3 protein that is phosphorylated and activated by the phosphorylation.
- a phosphory lated STAT3 is phosphorylated on tyrosine 705 or the residue corresponding to tyrosine 705 in homologs.
- activation of STAT3 means the STAT3 is capable of activating transcription.
- STAT3 gene'’ or “STAT3 sequence” as used herein refer to the gene or variants thereof that code for an STAT3 polypeptide capable of maintaining the activity of the STAT3 polypeptide (e.g., within at least 50%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or 100% activity compared to the STAT3 polypeptide).
- the variants or homologs have at least 90%, 95%, 96%, 97%, 98%, 99% or 100% nucleic acid sequence identity 7 across the whole sequence or a portion of the sequence (e.g., a 50, 100, 150 or 200 continuous nucleic acid portion) compared to the STAT3 sequence.
- STAT3 is substantially identical to the nucleic acid sequence identified by Accession No. NG007370 or a variant or homolog having substantial identity 7 thereto.
- STAT4 refers to a ‘‘Signal transducer and activator of transcription 4” protein and homologs thereof.
- STAT4 refers to the protein associated with Entrez Gene 6775, OMIM 600558, UniProt Q14765, and/or RefSeq (protein) NP001230764.
- the reference numbers immediately above refer to the protein, and associated nucleic acids, known as of the date of filing of this application.
- STAT5A refers to a ‘'Signal transducer and activator of transcription 5A” protein and homologs thereof.
- STAT5A refers to the protein associated with Entrez Gene 6776, OMIM 601511, UniProt P42229, and/or RefSeq (protein) NP003143.
- the reference numbers immediately above refer to the protein, and associated nucleic acids, known as of the date of filing of this application.
- STAT5B refers to a “Signal transducer and activator of transcription 5B” protein and homologs thereof.
- STAT5B refers to the protein associated with Entrez Gene 6777, OMIM 604260, UniProt P51692, and/or RefSeq (protein) NP036580.
- the reference numbers immediately above refer to the protein, and associated nucleic acids, known as of the date of filing of this application.
- STAT6 refers to a “Signal transducer and activator of transcription 6"’ protein and homologs thereof.
- STAT6 refers to the protein associated with Entrez Gene 6778, OMIM 601512, UniProt P42226, and/or RefSeq (protein) NP001 171549.
- the reference numbers immediately above refer to the protein, and associated nucleic acids, known as of the date of filing of this application.
- the term “linked” or “conjugated” when referring to two moieties means the two moieties are bonded, wherein the bond or bonds connecting the two moieties are covalent or non-covalent.
- the two moieties are covalently bonded to each other (e.g. directly or through a linking group).
- linking groups include a covalent bond, a nucleic acid sequence, a DNA sequence, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, substituted or unsubstituted heteroarylene, or combinations of two or more thereof.
- Nucleic acid refers to nucleotides (e g., deoxy ribonucleotides or ribonucleotides) and polymers thereof in either single-, double- or multiple-stranded form, or complements thereof; or nucleosides (e.g., deoxyribonucleosides or ribonucleosides). In embodiments, “nucleic acid” does not include nucleosides.
- polynucleotide oligonucleotide,” “oligo” or the like refer, in the usual and customary sense, to a linear sequence of nucleotides.
- nucleoside refers, in the usual and customary sense, to a glycosylamine including a nucleobase and a five-carbon sugar (ribose or deoxyribose).
- nucleosides include, cytidine, uridine, adenosine, guanosine, thymidine and inosine.
- nucleotide refers, in the usual and customary sense, to a single unit of a polynucleotide, i.e., a monomer. Nucleotides can be ribonucleotides, deoxyribonucleotides, or modified versions thereof.
- polynucleotides contemplated herein include single and double stranded DNA. single and double stranded RNA, and hybrid molecules having mixtures of single and double stranded DNA and RNA.
- nucleic acid e.g. polynucleotides, contemplated herein include any types of RNA, e.g. mRNA, siRNA, miRNA, and guide RNA and any types of DNA. genomic DNA. plasmid DNA. and minicircle DNA, and any fragments thereof.
- duplex in the context of polynucleotides refers, in the usual and customary sense, to double strandedness. Nucleic acids can be linear or branched.
- nucleic acids can be a linear chain of nucleotides or the nucleic acids can be branched, e.g., such that the nucleic acids comprise one or more arms or branches of nucleotides.
- the branched nucleic acids are repetitively branched to form higher ordered structures such as dendrimers and the like.
- Nucleic acids can include one or more reactive moieties.
- the term reactive moiety includes any group capable of reacting with another molecule, e.g., a nucleic acid or polypeptide through covalent, non-covalent or other interactions.
- the nucleic acid can include an amino acid reactive moiety that reacts with an amio acid on a protein or polypeptide through a covalent, non-covalent or other interaction.
- the terms also encompass nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non- naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides.
- Examples of such analogs include, without limitation, phosphodiester derivatives including, e.g., phosphorami date, phosphorodiamidate, phosphorothioate (also known as phosphorothioate having double bonded sulfur replacing oxygen in the phosphate), phosphorodithioate, phosphonocarboxylic acids, phosphonocarboxylates, phosphonoacetic acid, phosphonoformic acid, methyl phosphonate.
- phosphodiester derivatives including, e.g., phosphorami date, phosphorodiamidate, phosphorothioate (also known as phosphorothioate having double bonded sulfur replacing oxygen in the phosphate), phosphorodithioate, phosphonocarboxylic acids, phosphonocarboxylates, phosphonoacetic acid, phosphonoformic acid, methyl phosphonate.
- nucleotide bases such as, 2’0-methyl, 5 ’fluoro, 2'-deoxy-2'fluoro, 2'-deoxy, a universal base nucleotide, a 5-C methyl nucleotide, an inverted deoxybasic residue incorporation.
- 5-methyl cytidine, or pseudouridine and peptide nucleic acid backbones and linkages.
- Other analog nucleic acids include those with positive backbones; non-ionic backbones, modified sugars, and non-ribose backbones (e.g.
- nucleic acids containing one or more carbocyclic sugars are also included within one definition of nucleic acids. Modifications of the ribose-phosphate backbone may be done for a variety of reasons, e.g., to increase the stability and half-life of such molecules in physiological environments or as probes on a biochip. Mixtures of naturally occurring nucleic acids and analogs can be made; alternatively, mixtures of different nucleic acid analogs, and mixtures of naturally occurring nucleic acids and analogs may be made.
- the intemucleotide linkages in DNA are phosphodiester, phosphodiester derivatives, or a combination of both.
- Nucleic acids can include nonspecific sequences.
- nonspecific sequence refers to a nucleic acid sequence that contains a series of residues that are not designed to be complementary to or are only partially complementary to any other nucleic acid sequence.
- a nonspecific nucleic acid sequence is a sequence of nucleic acid residues that does not function as an inhibitory nucleic acid when contacted with a cell or organism.
- Unmodified nucleotide refers to a nucleotide that is not modified from its natural state.
- Modified nucleotide refers to a nucleotide that is modified from its natural state.
- the modification to the nucleotide can be to the base, the sugar, the phosphate, or two or more thereof.
- Nucleotides can be modified, for example, to include 2'-O-aminopropyl group, a 2’-O- ethyl group, a 2’-fluoro group, a 2’-O-methyl group, 2’-deoxy-2’fluoro group, a 2’-O- methoxy ethyl group, a 2’-O-allyl group, a 2’-O-propyl group, a 2’-O-pentyl group, or a constrained nucleotide.
- a “constrained nucleotide” refers to a nucleotide that is modified to maintain a rigid backbone structure.
- the constrained nucleotide refers to a nucleotide in which the pentose is modified to maintain a rigid backbone structure.
- Exemplary modifications to nucleotides that maintain a rigid structure include locked nucleic acids (e.g., LNA-modified nucleotides), cMOE-modified nucleotides, cEt-modified nucleotides, and the like.
- Constrained nucleotides are described, for example, by Pallan et al, Chem Commun (Camb), 48(66): 8195- 8197 (2012), the disclosure of which is incorporated by reference herein in its entirety.
- LNA-modified nucleotide refers to a locked nucleotide or a bridged nucleotide in which the pentose moiety is modified with an extra bridge connecting the 2’ oxygen and 4’ carbon, as shown in the structure below. where Bx is base. See Braasch et al, Chemistry & Biology. 8: 1-7 (2001).
- cMOE-modified nucleotide or “2 ’,4 ’-constrained 2’-O-methoxyethyl-modified nucleotide” refers to a nucleotide re: where Bx is the base.
- the cMOE-modified nucleotide is a (7?)-cMOE-modified nucleotide.
- the cMOE-modified nucleotide is a (S)-cMOE-modified nucleotide. See Pallan et al, Chem Commun (Camb), 48(66):8195-8197 (2012).
- “2’-O-ethyl-modified nucleotide” or “2’, 4’ -constrained 2'-O-ethyl-modified nucleotide” or ’cEt-modified nucleotide” refers to a nucleotide having the following structure: where Bx is the base.
- the cEt-modified nucleotide is a (/ )-cEt-modified nucleotide.
- the cEt-modified nucleotide is a (5)-cEt-modified nucleotide. See Pallan et al, Chem Commun (Camb), 48(66):8195-8197 (2012).
- a “spacer modification” refers to a moiety that does not include a nucleobase.
- Exemplary spacer modifications include an abasic spacer, a spacer phosphoramidite, abasic phosphoramidite, hexadecane phosphoramidite, octadecane phosphoramidite, a C6 disulfide phosphoramidite, and the like.
- the spacer phosphoramidite is a C3 spacer phosphoramidite, a C6 spacer phosphoramidite, or a C 12 spacer phosphoramidite.
- a “C3 spacer phosphoramidite” is a spacer modification represented by the structure:
- Abasic spacer or “dspacer” is a l’,2’-dideoxyribose without a nucleobase attached.
- an “antisense nucleic acid” as referred to herein is a nucleic acid (e.g., DNA or RNA molecule) that is complementary’ to at least a portion of a specific target nucleic acid and is capable of reducing transcription of the target nucleic acid (e.g. mRNA from DNA), reducing the translation of the target nucleic acid (e.g. mRNA). altering transcript splicing (e.g. single stranded morpholino oligo), or interfering with the endogenous activity of the target nucleic acid.
- synthetic antisense nucleic acids e.g. oligonucleotides
- synthetic antisense nucleic acids are generally between 15 and 25 bases in length.
- antisense nucleic acids are capable of hybridizing to (e.g. selectively hybridizing to) a target nucleic acid.
- the antisense nucleic acid hybridizes to the target nucleic acid in vitro.
- the antisense nucleic acid hybridizes to the target nucleic acid in a cell.
- the antisense nucleic acid hybridizes to the target nucleic acid in an organism.
- the antisense nucleic acid hybridizes to the target nucleic acid under physiological conditions.
- Antisense nucleic acids may comprise naturally occurring nucleotides or modified nucleotides such as, e.g., phosphorothioate, methylphosphonate, and anomeric sugar-phosphate, backbone-modified nucleotides.
- the antisense nucleic acids hybridize to the corresponding RNA forming a double-stranded molecule.
- the antisense nucleic acids interfere with the endogenous behavior of the RNA and inhibit its function relative to the absence of the antisense nucleic acid.
- Antisense nucleic acids may be single or double stranded nucleic acids.
- Non-limiting examples of antisense nucleic acids include siRNAs (including their derivatives or pre-cursors, such as nucleotide analogs), short hairpin RNAs (shRNA), micro RNAs (miRNA), saRNAs (small activating RNAs) and small nucleolar RNAs (snoRNA) or certain of their derivatives or pre-cursors.
- a polynucleotide is typically composed of a specific sequence of four nucleotide bases: adenine (A); cytosine (C); guanine (G); and thymine (T) (uracil (U) for thymine (T) when the polynucleotide is RNA).
- A adenine
- C cytosine
- G guanine
- T thymine
- U uracil
- T thymine
- polynucleotide sequence is the alphabetical representation of a polynucleotide molecule; alternatively, the term may be applied to the polynucleotide molecule itself. This alphabetical representation can be input into databases in a computer having a central processing unit and used for bioinformatics applications such as functional genomics and homology searching.
- Polynucleotides may optionally include one or more non-standard nucleotide(s), nucleotide analog(s) and/or modified nucleo
- “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids that encode identical or essentially identical amino acid sequences. Because of the degeneracy of the genetic code, a number of nucleic acid sequences will encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every' position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations,” which are one species of conservatively modified variations.
- Every' nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
- each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for try ptophan
- TGG which is ordinarily the only codon for try ptophan
- complement refers to a nucleotide (e.g.. RNA or DNA) or a sequence of nucleotides capable of base pairing with a complementary nucleotide or sequence of nucleotides.
- a complement may include a sequence of nucleotides that base pair with corresponding complementary nucleotides of a second nucleic acid sequence.
- the nucleotides of a complement may partially or completely match the nucleotides of the second nucleic acid sequence. Where the nucleotides of the complement completely match each nucleotide of the second nucleic acid sequence, the complement forms base pairs with each nucleotide of the second nucleic acid sequence. Where the nucleotides of the complement partially match the nucleotides of the second nucleic acid sequence only some of the nucleotides of the complement form base pairs with nucleotides of the second nucleic acid sequence. Examples of complementary' sequences include coding and a non-coding sequences, wherein the non-coding sequence contains complementary nucleotides to the coding sequence and thus forms the complement of the coding sequence.
- a further example of complementary sequences are sense and antisense sequences, wherein the sense sequence contains complementary nucleotides to the antisense sequence and thus forms the complement of the antisense sequence.
- the complementarity of sequences may be partial, in which only some of the nucleic acids match according to base pairing, or complete, where all the nucleic acids match according to base pairing.
- two sequences that are complementary to each other may have a specified percentage of nucleotides that are the same (i.e., about 60% identity', preferably 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%. 97%. 98%, 99%. or higher identity' over a specified region).
- the term “gene” means the segment of DNA involved in producing a protein; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
- the leader, the trailer as well as the introns include regulatory' elements that are necessary' during the transcription and the translation of a gene.
- a “protein gene product” is a protein expressed from a particular gene.
- the word “expression” or “expressed” as used herein in reference to a gene means the transcriptional and/or translational product of that gene.
- the level of expression of a DNA molecule in a cell may be determined on the basis of either the amount of corresponding mRNA that is present within the cell or the amount of protein encoded by that DNA produced by the cell.
- the level of expression of non-coding nucleic acid molecules e.g., siRNA
- the specified antibodies bind to a particular protein at least two times the background and more typically more than 10 to 100 times background.
- Specific binding to an antibody under such conditions requires an antibody that is selected for its specificity for a particular protein.
- polyclonal antibodies can be selected to obtain only a subset of antibodies that are specifically immunoreactive with the selected antigen and not with other proteins.
- This selection may be achieved by subtracting out antibodies that cross-react with other molecules.
- a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
- solid-phase ELISA immunoassays are routinely used to select antibodies specifically immunoreactive with a protein.
- nucleic acid, virus, or protein when applied to a nucleic acid, virus, or protein, denotes that the nucleic acid, virus, or protein is essentially free of other cellular components with which it is associated in the natural state. It can be, for example, in a homogeneous state and may be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified.
- Percentage of sequence identity is determined by comparing tw o optimally aligned sequences over a comparison window 7 , w herein the portion of the polynucleotide or polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity 7 .
- nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, preferably 65%, 70%, 75%, 80%, 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity 7 over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms or by manual alignment and visual inspection (see, e.g., http://www.ncbi.nlm.nih.gov/BLAST/ or the like).
- sequences are then said to be “substantially identical.”
- This definition also refers to, or may be applied to, the compliment of a test sequence.
- the definition also includes sequences that have deletions and/or additions, as well as those that have substitutions.
- the preferred algorithms can account for gaps and the like.
- identity exists over a region that is at least about 25 amino acids or nucleotides in length, or more preferably over a region that is 50-100 amino acids or nucleotides in length.
- amino acid or nucleotide base “position” is denoted by a number that sequentially identifies each amino acid (or nucleotide base) in the reference sequence based on its position relative to the N-terminus (or 5'-end). Due to deletions, insertions, truncations, fusions, and the like that must be taken into account when determining an optimal alignment, in general the amino acid residue number in a test sequence determined by simply counting from the N- terminus will not necessarily be the same as the number of its corresponding position in the reference sequence. For example, in a case where a variant has a deletion relative to an aligned reference sequence, there will be no amino acid in the variant that corresponds to a position in the reference sequence at the site of deletion.
- the term “about” means a range of values including the specified value, which a person of ordinary skill in the art would consider reasonably similar to the specified value. In embodiments, about means within a standard deviation using measurements generally acceptable in the art. In embodiments, about means a range extending to +/- 10% of the specified value. In embodiments, about means a range extending to +/- 5% of the specified value. In embodiments, about means a range extending to +/- 1 of the specified value. In embodiments, about includes the specified value.
- Control or “control experiment” is used in accordance with its plain ordinary' meaning and refers to an experiment in which the subjects or reagents of the experiment are treated as in a parallel experiment except for omission of a procedure, reagent, or variable of the experiment.
- the control is used as a standard of comparison in evaluating experimental effects.
- a control is the measurement of the activity 7 of a protein in the absence of a compound as described herein (including embodiments and examples).
- standard controls are most appropriate in a given situation and be able to analyze data based on comparisons to standard control values. Standard controls are also valuable for determining the significance (e.g. statistical significance) of data. For example, if values for a given parameter are widely variant in standard controls, variation in test samples will not be considered as significant.
- alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight (i.e., unbranched) or branched non-cyclic carbon chain (or carbon), or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having the number of carbon atoms designated (i.e., C1-C10 means one to ten carbons).
- saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
- An unsaturated alky l group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include, but are not limited to, viny l. 2-propenyl, croty l.
- An alkoxy is an alkyl attached to the remainder of the molecule via an oxygen linker (-O-).
- alky lene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkyl, as exemplified, but not limited by, -CH2CH2CH2-.
- an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.
- a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
- alkenylene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from an alkene.
- heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable non-cyclic straight or branched chain, or combinations thereof, including at least one carbon atom and at least one heteroatom selected from the group consisting of O, N, P, Si, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quatemized.
- the heteroatom(s) O, N, P, S, and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule.
- heteroalkylene by itself or as part of another substituent, means, unless otherwise stated, a divalent radical derived from heteroalkyl, as exemplified, but not limited by, -CH2-CH 2 -S-CH 2 -CH 2 -, -O-CH2-CH2-NH-CH2-, -O-(CH 2 ) 3 -O-PO 3 -, -O-(CH 2 )-O-PO 3 -, -O-(CH2)2-O-PO 3 -, -O-(CH2)4-O-PO 3 -, and the like.
- heteroatoms can also occupy either or both of the chain termini (e.g.. alkyleneoxy.
- heteroalkyl groups include those groups that are attached to the remainder of the molecule through a heteroatom, such as -C(O)R’, -C(O)NR’, -NR’R", -OR 1 , -SR 1 , and/or -SO2R'.
- heteroalkyd is recited, followed by recitations of specific heteroalky l groups, such as -NR'R" or the like, it will be understood that the terms heteroalky 1 and -NR'R" are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted as excluding specific heteroalkyl groups, such as -NR'R" or the like.
- cycloalkyd and “heterocycloalkyl,” by themselves or in combination with other terms, mean, unless otherwise stated, cyclic non-aromatic versions of “alkyl” and “heteroalkyd,” respectively, wherein the carbons making up the ring or rings do not necessarily need to be bonded to a hydrogen due to all carbon valencies participating in bonds with nonhydrogen atoms. Additionally, for heterocycloalky 1, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.
- Examples of cycloalkyl include, but are not limited to. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1 -cyclohexenyl,
- heterocycloalkyl examples include, but are not limited to, l-(l,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl,
- a “cycloalkylene” and a “heterocycloalkylene,” alone or as part of another substituent, means a divalent radical derived from a cycloalkyl and heterocycloalkyl, respectively.
- halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl” are meant to include monohaloalkyl and polyhaloalkyl.
- halo(Ci-C4)alkyl includes, but is not limited to, fluoromethyl, difluoromethyl, trifluoromethyl. 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
- acyl means, unless otherwise stated, -C(O)R where R is a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyd, substituted or unsubstituted heterocycloalkyd, substituted or unsubstituted ary l, or substituted or unsubstituted heteroaryl.
- ary 1 means, unless otherwise stated, a polyunsaturated, aromatic, hydrocarbon substituent, which can be a single ring or multiple rings (preferably from 1 to 3 rings) that are fused together (i.e., a fused ring aryl) or linked covalently (e.g., biphenyl).
- a fused ring aryl refers to multiple rings fused together wherein at least one of the fused rings is an ary l ring.
- heteroaryl refers to ary 1 groups (or rings) that contain at least one heteroatom such as N, O, or S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quatemized.
- heteroaryl includes fused ring heteroaryl groups (i.e., multiple rings fused together wherein at least one of the fused rings is a heteroaromatic ring).
- a 5,6-fused ring heteroary dene refers to tw o rings fused together, wherein one ring has 5 members and the other ring has 6 members, and w herein at least one ring is a heteroary! ring.
- a 6,6-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 6 members, and wherein at least one ring is a heteroaryl ring.
- a 6.5-fused ring heteroarylene refers to two rings fused together, wherein one ring has 6 members and the other ring has 5 members, and wherein at least one ring is a heteroaryl ring.
- a heteroaryl group can be attached to the remainder of the molecule through a carbon or heteroatom.
- Non-limiting examples of ary l and heteroaryl groups include phenyl,
- aryl and heteroaryl ring systems are selected from the group of acceptable substituents described below.
- Non-limiting examples of heteroary l groups include pyridinyl, pyrimidinyl. thiophenyl, thienyl, furanyl, indolyl.
- benzoxadiazolyl benzodioxolyl, benzodioxanyl, thianaphthanyl, pyrrolopyndinyl, indazolyL quinolinyl, quinoxalinyl, pyridopyrazinyl, quinazolinonyl, benzoisoxazolyl, imidazopyridinyl, benzofuranyl, benzothienyl, benzothiophenyl, phenyl, naphthyl, biphenyl, pyrrolyl, pyrazolyl, imidazolyl, py razinyl, oxazolyl, isoxazolyl, thiazolyl, furylthienyl, pyridyl, pyrimidyl, benzothiazolyl, purinyl, benzimidazolyl, isoquinolyl, thiadiazolyl.
- oxadiazolyl pyrrolyl, diazolyl, triazolyl, tetrazolyl, benzothiadiazolyl, isothiazolyl, pyrazolopyrimidinyl, pyrrolopyrimidinyl, benzotriazolyl, benzoxazolyl, or quinolyl.
- the examples above may be substituted or unsubstituted and divalent radicals of each heteroary l example above are non-limiting examples of heteroarylene.
- a fused ring heterocyloalkyl-aryl is an aryl fused to a helerocycloalk l.
- a fused ring heterocycloalkyl-heteroaryl is a heteroaryl fused to a heleroc cloalkvl.
- a fused ring heterocycloalkyl-cycloalkyl is a heterocycloalkyl fused to a cycloalkyl.
- a fused ring heterocycloalkyl-heterocycloalkyl is a heterocycloalkyd fused to another heterocycloalkyl.
- Fused ring heterocycloalkyl-ary 1, fused ring heterocycloalkyl-heteroaryl, fused ring heterocycloalky 1- cycloalky 1. or fused ring heterocycloalkyl-heterocycloalkyl may each independently be unsubstituted or substituted with one or more of the substitutents described herein.
- oxo means an oxygen that is double bonded to a carbon atom.
- alkylsulfonyl means a moiety having the formula -S(02)-R', where R' is a substituted or unsubstituted alkyl group as defined above. R' may have a specified number of carbons (e.g., “C1-C4 alkylsulfonyl”).
- R, R', R", R'", and R" each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted heteroaryl, substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups.
- aryl e.g., aryl substituted with 1-3 halogens
- substituted or unsubstituted heteroaryl substituted or unsubstituted alkyl, alkoxy, or thioalkoxy groups, or arylalkyl groups.
- each of the R groups is independently selected as are each R', R", R'", and R"" group when more than one of these groups is present.
- R' and R" are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7- membered ring.
- -NR'R includes, but is not limited to, 1 -pyrrolidinyl and 4- morpholinyl.
- alkyd is meant to include groups including carbon atoms bound to groups other than hy drogen groups, such as haloalkyl (e.g., -CF3 and -CH2CF3) and acyl
- R', R", R'", and R" are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
- R groups are independently selected as are each R', R", R'", and R"" groups when more than one of these groups is present.
- Two or more substituents may optionally be joined to form ary l, heteroaryl, cycloalkyl, or heterocycloalkyl groups.
- Such so-called ring-forming substituents are typically, though not necessarily, found attached to a cyclic base structure.
- the ring-forming substituents are attached to adjacent members of the base structure.
- two ringforming substituents attached to adjacent members of a cyclic base structure create a fused ring structure.
- the ring-forming substituents are attached to a single member of the base structure.
- two ring-forming substituents attached to a single member of a cyclic base structure create a spirocyclic structure.
- the ring-forming substituents are attached to non-adj acent members of the base structure.
- Two of the substituents on adj acent atoms of the ary l or heteroaryl ring may optionally 7 form a ring of the formula -T-C(O)-(CRR')q-U-, wherein T and U are independently -NR-, -O-, -CRR'-, or a single bond, and q is an integer of from 0 to 3.
- two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH2) r -B-, wherein A and B are independently -CRR'-, -O-, -NR-, -S-, -S(O) -, -S(O) 2 -, -S(O) 2 NR'-, or a single bond, and r is an integer of from 1 to 4.
- One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
- two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -(CRR')s-X'- (C"R"R"')d-, where s and d are independently integers of from 0 to 3, and X' is -O-, -NR'-, -S-, -S(O)-, -S(O)2-, or -S(O)2NR'-.
- R, R', R", and R' are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
- heteroatom or “ring heteroatom” are meant to include, oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
- a “substituent group,” as used herein, means a group selected from the following moieties: [0083] (A) oxo, halogen, -CF 3 , -CN, -OH, -NH 2 , -COOH, -CONH 2 , -NO 2 , -SH, -SO 2 C1, -SO3H. -SO4H. -SO 2 NH 2 , -NHNH 2 .
- a “size-limited substituent” or “ size-limited substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted Ci-C 2 o alkyd, each substituted or unsubstituted heteroalky l is a substituted or unsubstituted 2 to 20 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C 3 -Cs cycloalkyd, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl, each substituted or unsubstituted ary l is a substituted or unsubstituted Ce-Cw aryl, and each substituted or unsubstituted heteroary
- a “lower substituent’” or “ lower substituent group,” as used herein, means a group selected from all of the substituents described above for a “substituent group,” wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted Ci-Cs alkyl, each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C7 cycloalkyl, each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl, each substituted or unsubstituted aryl is a substituted or unsubstituted Ce-C aryl, and each substituted or unsubstituted heteroaryl is a substituted or un
- each substituted group described in the compounds herein is substituted with at least one substituent group. More specifically, in embodiments, each substituted alkyl, substituted heteroalkyl, substituted cycloalkyl, substituted heterocycloalkyl, substituted aryl, substituted heteroaryl, substituted alkylene, substituted heteroalkyd ene, substituted cycloalky lene, substituted heterocycloalkylene, substituted ary lene, and/or substituted heteroarylene described in the compounds herein are substituted with at least one substituent group. In embodiments, at least one or all of these groups are substituted with at least one size-limited substituent group. In embodiments, at least one or all of these groups are substituted with at least one lower substituent group.
- each substituted or unsubstituted alkyl may be a substituted or unsubstituted C1-C20 alkyl
- each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 20 membered heteroalkyl
- each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted Cs-Cs cycloalkyl
- each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 8 membered heterocycloalkyl
- each substituted or unsubstituted aryl is a substituted or unsubstituted Ce-Cw aryl
- each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 10 membered heteroaryl.
- each substituted or unsubstituted alkylene is a substituted or unsubstituted C1-C20 alkylene
- each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 20 membered heteroalkylene
- each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C3-C8 cycloalkylene
- each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 8 membered heterocycloalkylene
- each substituted or unsubstituted arylene is a substituted or unsubstituted Ce-Cw arylene
- each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 10 membered heteroarylene.
- each substituted or unsubstituted alkyl is a substituted or unsubstituted Ci-Cs alkyl
- each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2 to 8 membered heteroalkyl
- each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C3-C7 cycloalkyl
- each substituted or unsubstituted heterocycloalkyl is a substituted or unsubstituted 3 to 7 membered heterocycloalkyl
- each substituted or unsubstituted aryl is a substituted or unsubstituted C6-C10 aryl
- each substituted or unsubstituted heteroaryl is a substituted or unsubstituted 5 to 9 membered heteroaryl.
- each substituted or unsubstituted alkylene is a substituted or unsubstituted Ci-Cs alkylene
- each substituted or unsubstituted heteroalkylene is a substituted or unsubstituted 2 to 8 membered heteroalkylene
- each substituted or unsubstituted cycloalkylene is a substituted or unsubstituted C3-C7 cycloalkylene
- each substituted or unsubstituted heterocycloalkylene is a substituted or unsubstituted 3 to 7 membered heterocycloalkylene
- each substituted or unsubstituted arylene is a substituted or unsubstituted Ce-Cio arylene
- each substituted or unsubstituted heteroarylene is a substituted or unsubstituted 5 to 9 membered heteroarylene.
- the compound is a chemical species set forth in the Examples section below.
- activation means positively affecting (e.g. increasing) the activity or function of the protein relative to the activity 7 or function of the protein in the absence of the activator.
- activation means positively affecting (e.g. increasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the activator.
- the terms may reference activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein decreased in a disease.
- activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up-regulating signal transduction or enzymatic activity or the amount of a protein associated with a disease (e.g., a protein which is decreased in a disease relative to a non-diseased control).
- Activation may include, at least in part, partially or totally increasing stimulation, increasing or enabling activation, or activating, sensitizing, or up- regulating signal transduction or enzymatic activity or the amount of a protein
- agonist refers to a substance capable of detectably increasing the expression or activity of a given gene or protein.
- the agonist can increase expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the agonist. In certain instances, expression or activity is higher than the expression or activity in the absence of the agonist.
- inhibition means negatively affecting (e.g. decreasing) the activity or function of the protein relative to the activity or function of the protein in the absence of the inhibitor.
- inhibition means negatively affecting (e g. decreasing) the concentration or levels of the protein relative to the concentration or level of the protein in the absence of the inhibitor.
- inhibition refers to reduction of a disease or symptoms of disease. In embodiments, inhibition refers to a reduction in the activity 7 of a particular protein target.
- inhibition includes, at least in part, partially or totally blocking stimulation, decreasing, preventing, or delaying activation, or inactivating, desensitizing, or down-regulating signal transduction or enzymatic activity or the amount of a protein.
- inhibition refers to a reduction of activity of a target protein resulting from a direct interaction (e.g. an inhibitor binds to the target protein).
- inhibition refers to a reduction of activity of a target protein from an indirect interaction (e.g. an inhibitor binds to a protein that activates the target protein, thereby preventing target protein activation).
- inhibitor refers to a substance capable of detectably decreasing the expression or activity of a given gene or protein.
- the antagonist can decrease expression or activity 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more in comparison to a control in the absence of the antagonist. In embodiments, expression or activity is lower than the expression or activity in the absence of the antagonist.
- the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 6 unmodified nucleotides to about 60 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 6 nucleotides to about 60 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5 ’ end.
- the first DNA oligonucleotide comprises from about 6 unmodified nucleotides to about 50 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 6 nucleotides to about 50 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 8 unmodified nucleotides to about 40 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 8 nucleotides to about 40 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 8 unmodified nucleotides to about 30 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 8 nucleotides to about 30 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5‘ end.
- the first DNA oligonucleotide comprises from about 8 unmodified nucleotides to about 26 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 8 nucleotides to about 26 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 30 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 10 nucleotides to about 30 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 28 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 10 nucleotides to about 28 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 24 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 10 nucleotides to about 24 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 22 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 10 nucleotides to about 22 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5' end.
- the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 20 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 10 nucleotides to about 20 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 12 unmodified nucleotides to about 20 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 12 nucleotides to about 20 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises from about 14 unmodified nucleotides to about 18 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) from about 14 nucleotides to about 18 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA- modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different. In embodiments, the constrained nucleotides on the 3 ’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the first DNA oligonucleotide comprises about 10 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 10 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5' end.
- the first DNA oligonucleotide comprises about 11 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 11 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 12 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 12 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 13 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 13 nucleotides, and (ii) a constrained nucleotide on the 3‘ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 14 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 14 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 15 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 15 nucleotides, and (ii) a constrained nucleotide on the 3‘ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 16 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 1 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5‘ end.
- the first DNA oligonucleotide comprises about 17 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 17 nucleotides, and (ii) a constrained nucleotide on the 3‘ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 18 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 18 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 19 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 19 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 20 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 20 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5' end.
- the first DNA oligonucleotide comprises about 21 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 21 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the first DNA oligonucleotide comprises about 22 unmodified nucleotides
- the second DNA oligonucleotide comprises: (i) about 22 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5' end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA- modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different. In embodiments, the constrained nucleotides on the 3 ’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end. In embodiments, the second DNA oligonucleotide comprises two constrained nucleotide on the 3 ’ end and one constrained nucleotide on the 5 ’ end. In embodiments, the second DNA oligonucleotide comprises two constrained nucleotide on the 3 ' end and one constrained nucleotide on the 5‘ end. wherein the two constrained nucleotides are contiguous.
- the second DNA oligonucleotide comprises two constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end, wherein the two constrained nucleotides are alternating. In embodiments, the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and two constrained nucleotide on the 5' end. In embodiments, the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and two constrained nucleotide on the 5’ end, wherein the two constrained nucleotides are contiguous.
- the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and two constrained nucleotide on the 5’ end, wherein the two constrained nucleotides are alternating.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE- modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA-modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide.
- the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different.
- the constrained nucleotides on the 3 ’ end are different than the constrained nucleotides on the 5’ end.
- the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and three constrained nucleotide on the 5’ end. In embodiments, the second DNA oligonucleotide comprises one constrained nucleotide on the 3‘ end and three constrained nucleotide on the 5’ end, wherein the three constrained nucleotides are contiguous. In embodiments, the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and three constrained nucleotide on the 5’ end, wherein the three constrained nucleotides are alternating.
- the second DNA oligonucleotide comprises three constrained nucleotide on the 3' end and one constrained nucleotide on the 5’ end. In embodiments, the second DNA oligonucleotide comprises three constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end, wherein the three constrained nucleotides are contiguous. In embodiments, the second DNA oligonucleotide comprises three constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end, wherein the three constrained nucleotides are alternating.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different.
- the constrained nucleotides on the 3’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification. [0103] In embodiments, the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotides on the 5' end.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3’ end are contiguous and the two constrained nucleotides on the 5’ end are contiguous.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotide on the 5’ end, wherein the two constrained nucleotides on the 3’ end are alternating and the two constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3’ end are contiguous and the two constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3’ end are alternating and the two constrained nucleotides on the 5’ end are contiguous.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different.
- the constrained nucleotides on the 3’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the second DNA oligonucleotide comprises two constrained nucleotide on the 3’ end and three constrained nucleotide on the 5’ end. In embodiments, the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3’ end are contiguous and the three constrained nucleotides on the 5’ end are contiguous.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and three constrained nucleotides on the 5" end, wherein the two constrained nucleotides on the 3’ end are alternating and the three constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3’ end are contiguous and the three constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the two constrained nucleotides on the 3' end are alternating and the three constrained nucleotides on the 5’ end are contiguous.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA-modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different. In embodiments, the constrained nucleotides on the 3’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end. In embodiments, the second DNA oligonucleotide comprises three constrained nucleotides on the 3‘ end and two constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are contiguous and the two constrained nucleotides on the 5‘ end are contiguous.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are alternating and the two constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are contiguous and the two constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are alternating and the two constrained nucleotides on the 5’ end are contiguous.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA- modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide.
- the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different.
- the constrained nucleotides on the 3 ’ end are different than the constrained nucleotides on the 5‘ end.
- the constrained nucleotides on the 3’ end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3‘ end and three constrained nucleotides on the 5’ end. In embodiments, the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are contiguous and the three constrained nucleotides on the 5’ end are contiguous.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are alternating and the three constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3 ’ end and three constrained nucleotides on the 5 ’ end, wherein the three constrained nucleotides on the 3‘ end are contiguous and the three constrained nucleotides on the 5’ end are alternating.
- the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end, wherein the three constrained nucleotides on the 3’ end are alternating and the three constrained nucleotides on the 5’ end are contiguous.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA-modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide.
- the constrained nucleotides in the second DNA oligonucleotide can be the same or different. In embodiments, the constrained nucleotides on the 3’ end are different than the constrained nucleotides on the 5’ end. In embodiments, the constrained nucleotides on the 3' end are the same as the constrained nucleotides on the 5’ end.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the term “on the 3’ end’' means the terminal nucleotide(s) are adjacent the 3‘ end.
- the term “on the 5' end” means the terminal nucleotide(s) are adjacent the 5’ end.
- a constrained nucleotide on the 5’ end with reference to, for example, a nucleotide having the structure: 5'- cTATTTGGATGTCAGC-3’ (SEQ ID NO:38), refers to the terminal nucleotide in lower case letter and underlined on the 5’ end.
- the two or more nucleotides can be contiguous.
- three contiguous constrained nucleotides on the 3’ end refers to the three contiguous nucleotides in lower case letter and underlined on the 3’ end.
- two contiguous constrained nucleotides on the 3’ end refers to the two contiguous nucleotides in lower case letter and underlined on the 5’ end.
- the term '‘contiguous” has the plain and ordinary meaning of “next to” or “together in sequence.”
- the two or more nucleotides can be alternating.
- alternating means that the terminal nucleotide is constrained, the next sequential nucleotide is not constrained, and the next sequential nucleotide is constrained.
- two alternating constrained nucleotides on the 5’ end refers to the two nucleotides in lower case letter and underlined on the 5’ end which alternate between the locked nucleic acid, a nucleic acid that does not have an LNA modification, and a locked nucleic acid.
- three alternating LNA- modified nucleotides on the 3’ end refers to the three nucleotides in lower case letter and underlined on the 3‘ end which alternate betw een the locked nucleic acid, a nucleic acid that does not have an LNA modification, and two locked nucleic acids.
- the term “alternating” can otherwise be described as contiguous, except that the second nucleotide from the 3’ end or 5’ end does not have a LNA modification.
- the first DNA oligonucleotide and the second DNA oligonucleotide do not have the same number of nucleotides.
- the first DNA oligonucleotide can have 18 unmodified nucleotides and the second DNA oligonucleotide can have 16 nucleotides.
- the first DNA oligonucleotide can have 16 unmodified nucleotides and the second DNA oligonucleotide can have 18 nucleotides.
- the first DNA oligonucleotide and the second DNA oligonucleotide have the same number of nucleotides.
- the first DNA oligonucleotide can have 16 unmodified nucleotides and the second DNA oligonucleotide can have 16 nucleotides.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification. In embodiments, at least 10% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 20% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 30% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification.
- At least 40% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 50% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 60% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 70% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, at least 80% of the nucleotides in the second DNA oligonucleotide have a phosphorothioate modification.
- nucleotides in the second DNA oligonucleotide have a phosphorothioate modification. In embodiments, all the nucleotides in the second DNA oligonucleotide have a phosphorothioate intemucleotide linkage.
- the second DNA oligonucleotide further comprises a nucleotide having a modification.
- the modification can be a spacer modification or a nucleotide modification.
- the second DNA oligonucleotide further comprises a nucleotide having a modification to the base or sugar.
- the second DNA oligonucleotide comprises a nucleotide having a modification selected from the group consisting of 2’-O- aminopropyl group, a 2’-0-ethyl group, a 2'-fluoro group, a 2’-O-methyl group, 2'-deoxy- 2’fluoro group, a 2'-O-methoxyethyl group, a 2’-O-allyl group. a 2’-O-propyl group. a 2'-O- pentyl group, and a constrained nucleotide.
- the second DNA oligonucleotide further comprises a nucleotide having a 2’O-methyl group.
- the second DNA oligonucleotide further comprises a nucleotide having a 2’ fluoro group. In embodiments, the second DNA oligonucleotide further comprises a nucleotide having a 2’-deoxy-2’fluoro group. In embodiments, the second DNA oligonucleotide further comprises a nucleotide have a constrained nucleic acid modification, where this constrained nucleic acid modification is in addition to the constrained nucleotide on the 3’ end and 5’ end. In embodiments, this additional nucleotide have a constrained nucleic acid modification is contiguous with the constrained nucleotide on the 3' end or the 5‘ end.
- this additional this additional nucleotide have a constrained nucleic acid modification is separated from the constrained nucleotide on the 3’ end or the 5' end by one or more nucleotides that do not have a constrained modification.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA- modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide.
- the constrained nucleotide is a cET-modified nucleotide.
- the second DNA oligonucleotide further comprises a spacer modification.
- the spacer modification is an abasic spacer, a spacer phosphoramidite, abasic phosphorami dite, hexadecane phosphoramidite, octadecane phosphoramidite, a C6 disulfide phosphoramidite, or a combination of two or more thereof.
- the spacer phosphoramidite is a C3 spacer phosphoramidite, a C6 spacer phosphoramidite, or a C 12 spacer phosphoramidite.
- the second DNA oligonucleotide further comprises an abasic spacer modification.
- the second DNA oligonucleotide further comprises a C3 spacer phosphoramidite.
- the second DNA oligonucleotide is an antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT1 antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT2 antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT3 antisense oligonucleotide.
- the second DNA oligonucleotide is a STAT4 antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT5 antisense oligonucleotide. In embodiments, the second DNA oligonucleotide is a STAT6 antisense oligonucleotide.
- the second DNA oligonucleotide is a STAT3 antisense oligonucleotide.
- the STAT3 antisense oligonucleotide comprises SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO:35, or SEQ ID NO:36.
- the STAT3 antisense oligonucleotide comprises SEQ ID NO:2.
- the STAT3 antisense oligonucleotide comprises SEQ ID NO: 4.
- the STAT3 antisense oligonucleotide comprises SEQ ID NO:6. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO:8. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO: 10. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO: 12. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO: 14. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO:35. In embodiments, the STAT3 antisense oligonucleotide comprises SEQ ID NO:36. [0117] Table 1
- underline refers to a phosphorothioated nucleotide (phosphorothioated intemucleotide linkage); single quote (e g., G’) indicates a 2’0-methyl modification, X 1 is an abasic spacer (“/idSp/”), and X 2 is C3 spacer phosphoramidite (‘7iSpC3/”).
- the first DNA oligonucleotide comprises SEQ ID NO:1, SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO: 11, or SEQ ID NO: 13.
- the first DNA oligonucleotide comprises SEQ ID NO: 1.
- the first DNA oligonucleotide comprises SEQ ID NO:3.
- the first DNA oligonucleotide comprises SEQ ID NO: 5.
- the first DNA oligonucleotide comprises SEQ ID NO:7.
- the first DNA oligonucleotide comprises SEQ ID NO:9.
- the first DNA oligonucleotide comprises SEQ ID NO: 11.
- the first DNA oligonucleotide comprises SEQ ID NO: 13. [0120] Table 2
- the first DNA oligonucleotide comprises SEQ ID NO: 1 and the second DNA oligonucleotide comprises SEQ ID NO:2. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO:3 and the second DNA oligonucleotide comprises SEQ ID NO:4. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO: 5 and the second DNA oligonucleotide comprises SEQ ID NO:6. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO:7 and the second DNA oligonucleotide comprises SEQ ID NO:8.
- the first DNA oligonucleotide comprises SEQ ID NO:9 and the second DNA oligonucleotide comprises SEQ ID NOTO. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO: 11 and the second DNA oligonucleotide comprises SEQ ID NO: 12. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO: 13 and the second DNA oligonucleotide comprises SEQ ID NO: 14. In embodiments, the first DNA oligonucleotide comprises SEQ ID NOT and the second DNA oligonucleotide comprises SEQ ID NO:35. In embodiments, the first DNA oligonucleotide comprises SEQ ID NO: 1 and the second DNA oligonucleotide comprises SEQ ID NO:36.
- a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide is equivalent to the phrase “a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide, wherein the first DNA oligonucleotide is hybridized to a second DNA oligonucleotide.”
- the compounds described herein comprise a phosphorothioated CpG oligodeoxynucleotide (ODN).
- ODN phosphorothioated CpG oligodeoxynucleotide
- the CpG ODN is a CpG-A ODN, a CpG-B ODN, a CpG-C ODN, or a combination of two or more thereof.
- the CpG ODN is a CpG-A ODN.
- the CpG ODN is a CpG-B ODN.
- the CpG ODN is a CpG-C ODN.
- the CpG ODN is CpG ODN 1585, CpG ODN 2216, CpG ODN 2336, CpG ODN 1668, CpG ODN 1826, CpG ODN 2006, CpG ODN 2007, CpG ODN BW006, CpG ODN D-SL01, CpG ODN 2395, CpG ODN M362. CpG ODN D-SL03, CpG ODN DI 9, or a combination of two or more thereof.
- the CpG ODN is CpG ODN 1585.
- the CpG ODN is CpG ODN 2216.
- the CpG ODN is CpG ODN 2336.
- the CpG ODN is CpG ODN 1668. In embodiments, the CpG ODN is CpG ODN 1826. In embodiments, the CpG ODN is CpG ODN 2006. In embodiments, the CpG ODN is CpG ODN 2007. In embodiments, the CpG ODN is CpG ODN BW006. In embodiments, the CpG ODN is CpG ODN D-SL01. In embodiments, the CpG ODN is CpG ODN 2395. In embodiments, the CpG ODN is CpG ODN CpG ODN M362. In embodiments, the CpG ODN is CpG ODN D-SL03. In embodiments, the CpG ODN is CpG ODN DI 9.
- the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43.
- the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43.
- the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43, SEQ ID NO: 16, SEQ ID NO: 17, or SEQ ID NO: 18.
- the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:15, SEQ ID NO: 19, SEQ ID NO:20, or SEQ ID NO:21.
- the CpG ODN is SEQ ID NO:43 or SEQ ID NO: 15. In embodiments, the CpG ODN is SEQ ID NO:43. In embodiments, the CpG ODN is SEQ ID NO: 15. In embodiments, the CpG ODN is SEQ ID NO:16. In embodiments, the CpG ODN is SEQ ID NO:17. In embodiments, the CpG ODN is SEQ ID NO: 18. In embodiments, the CpG ODN is SEQ ID NO: 19. In embodiments, the CpG ODN is SEQ ID NO:20. In embodiments, the CpG ODN is SEQ ID NO:21. In embodiments, the CpG ODN is SEQ ID NO:22.
- the CpG ODN is SEQ ID NO:23. In embodiments, the CpG ODN is SEQ ID NO:24. In embodiments, the CpG ODN is SEQ ID NO:25. In embodiments, the CpG ODN is SEQ ID NO:26. In embodiments, the CpG ODN is SEQ ID NO:27. In embodiments, the CpG ODN is SEQ ID NO:28. In embodiments, the CpG ODN is SEQ ID NO:29. In embodiments, the CpG ODN is SEQ ID NO:30. In embodiments, the CpG ODN is SEQ ID NO:31. In embodiments, the CpG ODN is SEQ ID NO:32.
- the compounds described herein include a phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO:43 covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1 , wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:2; wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO: 15 is covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:2; wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide is covalently bonded to the first DNA oligonucleotide via the moiety of Formula (A).
- the compounds described herein include a phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO:43 covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:35; wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO: 15 is covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:35; wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5‘ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide is covalently bonded to the first DNA oligonucleotide via the moiety of Formula (A).
- the compounds described herein include a phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO:43 covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:36; wherein the 3‘ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO: 15 is covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO: 1, wherein SEQ ID NO: 1 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:36; wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide is covalently bonded to the first DNA oligonucleotide via the moiety of Formula (A).
- the compounds described herein include a phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO: 15 covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:4, wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide comprising SEQ ID NO:43 is covalently bonded to a first DNA oligonucleotide comprising SEQ ID NO:3, wherein SEQ ID NO:3 is hybridized to a second DNA oligonucleotide comprising SEQ ID NO:4, wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- the phosphorothioated CpG oligodeoxynucleotide is covalently bonded to the first DNA oligonucleotide via the moiety of Formula (A).
- underline refers to a phosphorothioated nucleotide (phosphorothioated intemucleotide linkage).
- the phosphorothioated CpG ODN is linked to the first DNA oligonucleotide by any linking group known in the art.
- the linking group comprises a bond, a nucleic acid sequence, a DNA sequence, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, substituted or unsubstituted heteroarylene, or a combination of two or more thereof.
- the linking group comprises a bond, a nucleic acid sequence, unsubstituted alkylene, unsubstituted heteroalkydene, or a combination of two or more thereof. In embodiments, the linking group is a covalent bond. In embodiments, the linking group is a nucleic acid sequence. In embodiments, the linking group is a DNA sequence. In embodiments, the linking group comprises a nucleic acid sequence and a substituted or unsubstituted alkylene. In embodiments, the linking group comprises a nucleic acid sequence and an unsubstituted alkylene. In embodiments, the linking group comprises a nucleic acid sequence and a substituted or unsubstituted heteroalky lene.
- the linking group comprises a nucleic acid sequence and an unsubstituted heteroalkylene. In embodiments, the linking group comprises a substituted or unsubstituted heteroalkylene. In embodiments, the linking group comprises a substituted heteroalkylene.
- the linking group comprises a substituted heteroalkydene. In embodiments, the linking group comprises a substituted 6 to 60 membered heteroalkylene. In embodiments, the linking group comprises a substituted 6 to 54 membered heteroalkylene. In embodiments, the linking group comprises a substituted 12 to 48 membered heteroalky lene. In embodiments, the linking group comprises a substituted 18 to 42 membered heteroalky lene. In embodiments, the linking group comprises a substituted 24 to 36 membered heteroalky lene. In embodiments, the linking group comprises a substituted 30 membered heteroalky lene. In embodiments, the heteroalkylene comprises an oxygen atom, a phosphorous atom, or a combination thereof.
- the substituents on the substituted heteroalkylene comprise oxo, -OH, -O', or a combination of two or more thereof.
- the linking group comprises a substituted 18 to 42 membered heteroalkylene; wherein the heteroalkylene comprises an oxygen atom, a phosphorous atom, or a combination thereof; and wherein the substituents are independently selected from the group consisting of oxo, -OH, and -O'.
- the linking group comprises any one of the following structures: wherein zl, z2, z3 and z4 are independently integers from 0 to 20; and each X is independently - OH or -O’.
- zl is an integer from 0 to 5.
- zl is an integer from 2 to 4.
- z2 is an integer from 0 to 5.
- z2 is an integer from 2 to 4.
- z3 is an integer from 0 to 5.
- zl is an integer from 2 to 4.
- z4 is an integer from 3 to 7.
- z4 is an integer from 4 to 6.
- each X is -OH.
- the linking group comprises the structure: wherein n is an integer from 1 to 10. In embodiments, n is an integer from 2 to 8. In embodiments, n is an integer from 3 to 7. In embodiments, n is an integer from 4 to 6. In embodiments, n is 1. In embodiments, n is 2. In embodiments, n is 3. In embodiments, n is 4. In embodiments, n is 5. In embodiments, n is 6. In embodiments, n is 7. In embodiments, n is 8. In embodiments, n is 9. In embodiments, n is 10.
- the compound further comprises a detectable moiety-.
- the phosphorothioated CpG ODN, the first DNA oligonucleotide, the second DNA oligonucleotide, or any combination thereof comprise a detectable moiety 7 .
- the phosphorothioated CpG ODN comprises a detectable moiety .
- the first DNA oligonucleotide comprises a detectable moiety-.
- the second DNA oligonucleotide comprises a detectable moiety 7 .
- a compound compnsing a detectable moiety' is one that is bound, either covalently, through a linker or a chemical bond, or noncovalently, through ionic, van der Waals, electrostatic, or hydrogen bonds to a detectable moiety' such that the presence of the nucleic acid may be detected by detecting the presence of the detectable moiety bound to the nucleic acid.
- a method using high affinity interactions may achieve the same results where one of a pair of binding partners binds to the other, e g., detectable moiety.
- the phosphorothioate nucleic acid or phosphorothioate polymer backbone includes a detectable agent, as disclosed herein and known in the art.
- a “detectable agent” or “detectable moiety” is a compound or composition detectable by appropriate means such as spectroscopic, photochemical, biochemical, immunochemical, chemical, magnetic resonance imaging, or other physical means.
- a detectable moiety is a monovalent detectable agent or a detectable agent bound (e.g. covalently and directly or via a linking group) with another compound, e.g., a nucleic acid.
- Exemplary 7 detectable agents/moieties for use in the present disclosure include an antibody ligand, a peptide, a nucleic acid, radioisotopes, paramagnetic metal ions, fluorophore (e.g. fluorescent dyes), electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, a biotin-avidin complex, a biotin-streptavidin complex, digoxigenin, magnetic beads (e.g., DYNABEADS® by ThermoFisher, encompassing functionalized magnetic beads such as DYNABEADS® M-270 amine by ThermoFisher), paramagnetic molecules, paramagnetic nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticles, ultrasmall superparamagnetic iron oxide nanoparticle aggregates, superparamagnetic iron oxide nanoparticles, superparamagnetic iron oxide nanoparticle aggregates, monocrystalline iron oxide nanoparticles
- microbubbles e.g. including microbubble shells including albumin, galactose, lipid, and/or polymers; microbubble gas core including air. heavy gases, perfluorcarbon, nitrogen, octafluoropropane, perflexane lipid microsphere, perflutren, etc ), iodinated contrast agents (e.g.
- iohexol iodixanol, ioversol, iopamidol, ioxilan, iopromide, diatrizoate, metrizoate.
- ioxaglate barium sulfate, thorium dioxide, gold, gold nanoparticles, gold nanoparticle aggregates, fluorophores, two-photon fluorophores, or haptens and proteins or other entities which can be made detectable, e.g., by incorporating a radiolabel into a peptide or antibody specifically reactive with a target peptide.
- the disclosure provides pharmaceutical compositions comprising a compound described herein and a pharmaceutically acceptable excipient.
- “Pharmaceutically acceptable excipient” refers to a substance that aids the administration of an active agent to and absorption by a subject and can be included in the compositions of the present disclosure without causing a significant adverse toxicological effect on the patient.
- Non-limiting examples of pharmaceutically acceptable excipients include water, NaCl, normal saline solutions, lactated Ringer's, normal sucrose, normal glucose, binders, fillers, disintegrants, lubricants, coatings, sweeteners, flavors, salt solutions (such as Ringer's solution), alcohols, oils, gelatins, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethycellulose, polyvinyl pyrrolidine, and colors, and the like.
- Such preparations can be sterilized and, if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
- auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
- auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, and/or aromatic substances and the like that do not deleteriously react with the compounds of the disclosure.
- compositions comprising: (1) an immune checkpoint inhibitor and (2) a compound which comprises a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the compound (2) is any compound described herein, including all embodiments of the compounds described herein.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA- modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide.
- the constrained nucleotide is a cET-modified nucleotide.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor. In embodiments, the immune checkpoint inhibitor is a PD-L1 inhibitor. In embodiments, the immune checkpoint inhibitor is a PD-1 inhibitor.
- a “effective amount” is an amount sufficient for a compound to accomplish a stated purpose relative to the absence of the compound (e.g. achieve the effect for which it is administered, treat a disease).
- An example of an “effective amount” is an amount sufficient to contribute to the treatment of a disease which could also be referred to as a “therapeutically effective amount.”
- a “reduction” of a symptom or symptoms means decreasing of the severity' or frequency of the symptoms or elimination of the symptoms. The exact amounts will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, e.g., Lieberman, Pharmaceutical Dosage Forms (vols.
- the therapeutically effective amount can be initially determined from cell culture assays.
- Target concentrations will be those concentrations of active compound that are capable of achieving the methods described herein, as measured using the methods described herein or known in the art.
- therapeutically effective amounts for use in humans can also be determined from animal models. For example, a dose for humans can be formulated to achieve a concentration that has been found to be effective in animals.
- the dosage in humans can be adjusted by monitoring the effectiveness of the compounds or compositions described herein, and adjusting the dosage upwards or downwards. Adjusting the dose to achieve maximal efficacy in humans based on the methods described above and other methods is well within the capabilities of the ordinarily skilled artisan.
- Dosages may be varied depending upon the requirements of the patient and the compound being employed.
- the dose administered to a patient should be sufficient to effect a beneficial therapeutic response in the patient over time.
- the size of the dose also will be determined by the existence, nature, and extent of any adverse side-effects. Determination of the proper dosage for a particular situation is within the skill of the practitioner. Generally, treatment is initiated with smaller dosages which are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under circumstances is reached. Dosage amounts and intervals can be adjusted individually to provide levels of the administered compound effective for the particular clinical indication being treated. This will provide a therapeutic regimen that is commensurate with the severity of the individual's disease state.
- administering means oral administration, administration as a suppository, topical contact, intravenous, parenteral, intraperitoneal, intramuscular, intralesional, intrathecal, intranasal or subcutaneous administration, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to a subject. Administration is by any route, including parenteral and transmucosal (e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal).
- parenteral and transmucosal e.g., buccal, sublingual, palatal, gingival, nasal, vaginal, rectal, or transdermal.
- Parenteral administration includes, e.g., intra-tumoral, intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
- the compounds or pharmaceutical compositions described herein are parenterally administered to a patient.
- the compounds or pharmaceutical compositions descnbed herein are administered intra-tumorally to a patient.
- Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
- the administering does not include administration of any active agent other than the recited active agent.
- the disclosure provides methods of treating cancer in a patient in need thereof.
- the methods of treating cancer comprise administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the cancer is a central nervous system cancer, leukemia, lymphoma, a solid tumor cancer, or an epidermoid carcinoma. In embodiments, the cancer is a central nervous system cancer.
- the cancer is a solid tumor cancer. In embodiments, the cancer is an epidermoid carcinoma. In embodiments, the cancer is leukemia. In embodiments, the cancer is lymphoma. In embodiments, the cancer is B cell lymphoma.
- the disclosure provides methods of treating a central nervous system cancer in a patient in need thereof by administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the central nervous system cancer is glioma, cranial primitive neuroectodermal tumor, ependymal tumor, hemangiopericytoma, germ cell tumor, pineal tumor, or primary central nervous system lymphoma.
- the cancer is glioma.
- the glioma is astrocytoma, glioblastoma, or oligodendroglioma.
- the glioma is astrocytoma.
- the glioma is glioblastoma.
- the glioma is oligodendroglioma.
- the glioma is a brain stem glioma.
- the glioma is a mixed glioma. In embodiments, the glioma is an optic pathway glioma. In embodiments, the cancer is cranial primitive neuroectodermal tumor. In embodiments, the cranial primitive neuroectodermal tumor is medulloblastoma, cerebral neuroblastoma, pineoblastoma, or esthesioneuroblastoma. In embodiments, the cranial primitive neuroectodermal tumor is medulloblastoma. In embodiments, the cranial primitive neuroectodermal tumor is cerebral neuroblastoma. In embodiments, the cranial primitive neuroectodermal tumor is pineoblastoma.
- the cranial primitive neuroectodermal tumor is esthesioneuroblastoma.
- the cancer is ependymal tumor.
- the ependymal tumor is ependymoma, myxopapillary ependymoma, or subependymoma.
- the ependymal tumor is ependymoma.
- the ependymal tumor is myxopapillary ependymoma.
- the ependymal tumor is subependymoma.
- the cancer is hemangiopericytoma.
- the cancer is germ cell tumor. In embodiments, the cancer is pineal tumor. In embodiments, the cancer is primary central nervous system lymphoma. [0148] In embodiments, the disclosure provides methods of treating a glioma in a patient in need thereof by administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA- modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the disclosure provides methods of treating leukemia in a patient in need thereof by administering to the patient in an effective amount of a compound compnsing a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA- modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the cancer is acute myeloid leukemia.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the disclosure provides methods of treating a solid tumor cancer in a patient in need thereof by administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE- modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the solid tumor cancer is prostate cancer, breast cancer, colorectal cancer, bladder cancer, lung cancer, liver cancer, pancreatic caner, renal cancer, stomach cancer, or melanoma.
- the solid tumor cancer is prostate cancer. In embodiments, the solid tumor cancer is breast cancer. In embodiments, the solid tumor cancer is colorectal cancer. In embodiments, the solid tumor cancer is bladder cancer. In embodiments, the solid tumor cancer is lung cancer. In embodiments, the solid tumor cancer is liver cancer. In embodiments, the solid tumor cancer is pancreatic caner. In embodiments, the solid tumor cancer is renal cancer. In embodiments, the cancer is stomach cancer. In embodiments, the solid tumor cancer is melanoma.
- the disclosure provides methods of treating prostate cancer in a patient in need thereof by administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide.
- the constrained nucleotide is a LNA-modified nucleotide.
- the constrained nucleotide is a cMOE-modified nucleotide.
- the constrained nucleotide is a cET-modified nucleotide.
- the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the disclosure provides methods of treating epidermoid carcinoma in a patient in need thereof by administering to the patient in an effective amount of a compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and the second DNA oligonucleotide comprises (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET-modified nucleotide. In embodiments, the constrained nucleotide is a LNA-modified nucleotide. In embodiments, the constrained nucleotide is a cMOE-modified nucleotide. In embodiments, the constrained nucleotide is a cET-modified nucleotide. In embodiments, the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- the compound is any compound described herein, including all embodiments of the compounds described herein.
- the methods further comprise administering to the patient an effective amount of an immune checkpoint inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-L1 inhibitor.
- the immune checkpoint inhibitor is a PD-1 inhibitor.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- the epidermoid carcinoma is thyroid cancer, esophageal cancer, vaginal cancer, anal cancer, cervical cancer, or head and neck cancer. In embodiments, the epidermoid carcinoma is thyroid cancer.
- the epidermoid carcinoma is esophageal cancer.
- the cancer is anal cancer.
- the cancer is cervical cancer.
- the cancer is head and neck cancer.
- the cancer is vaginal cancer.
- the PD-1 inhibitor is pembrolizumab, nivolumab. cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, spartalizumab, retifanlimab, pimivalimab (JTX-4014), AMP-224, or MEDI0680 (AMP-514).
- the PD-1 inhibitor is pembrolizumab, nivolumab, cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, retifanlimab, or spartalizumab.
- the PD-1 inhibitor is pembrolizumab.
- the PD-1 inhibitor is nivolumab.
- the PD-1 inhibitor is cemiplimab.
- the PD-1 inhibitor is dostarlimab.
- the PD-1 inhibitor is camrelizumab.
- the PD-1 inhibitor is sintilimab.
- the PD-1 inhibitor is tislelizumab. In embodiments, the PD-1 inhibitor is toripalimab. In embodiments, the PD-1 inhibitor is spartalizumab. In embodiments, the PD-1 inhibitor is pimivalimab. In embodiments, the PD-1 inhibitor is retifanlimab. In embodiments, the PD-1 inhibitor is AMP-224. In embodiments, the PD-1 inhibitor is MEDI0680.
- the PD-L1 inhibitor is atezolizumab, avelumab, durvalumab, envafolimab, cosibelimab, AUNP12, CA-170, or BMS-986189.
- the PD-L1 inhibitor is atezolizumab, avelumab, durvalumab, envafolimab, cosibelimab, AUNP12, CA-170. or BMS-986189.
- the PD-L1 inhibitor is atezolizumab, avelumab, durvalumab, envafolimab, or cosibelimab.
- the PD-L1 inhibitor is atezolizumab. In embodiments, the PD-L1 inhibitor is avelumab. In embodiments, the PD-L1 inhibitor is durvalumab. In embodiments, the PD-L1 inhibitor is envafolimab. In embodiments, the PD-L1 inhibitor is cosibelimab. In embodiments, the PD-L1 inhibitor is ALJNP12. In embodiments, the PD-L1 inhibitor is CA-170. In embodiments, the PD-L1 inhibitor is BMS-986189. [0155] The term “cancer” refers to all ty pes of cancer, neoplasm or malignant tumors found in mammals (e.g. humans), including leukemias, lymphomas, carcinomas and sarcomas.
- Exemplary cancers that can be treated with the compounds and pharmaceutical compositions described herein include brain cancer, glioma, glioblastoma, neuroblastoma, prostate cancer, colorectal cancer, pancreatic cancer, medulloblastoma, melanoma, cervical cancer, gastric cancer, ovarian cancer, lung cancer, cancer of the head, Hodgkin's disease, and Non-Hodgkin's lymphomas.
- Other cancers that may be treated with the compounds and pharmaceutical compositions described herein include cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head and neck, liver, kidney, lung, ovary, pancreas, rectum, stomach, and uterus.
- cancers that may be treated with the compounds and pharmaceutical compositions described herein include, thyroid carcinoma, cholangiocarcinoma, pancreatic adenocarcinoma, skin cutaneous melanoma, colon adenocarcinoma, rectum adenocarcinoma, stomach adenocarcinoma, esophageal carcinoma, head and neck squamous cell carcinoma, breast invasive carcinoma, lung adenocarcinoma, lung squamous cell carcinoma, non-small cell lung carcinoma, mesothelioma, multiple myeloma, neuroblastoma, glioma, glioblastoma, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, thyroid cancer, neuroblastoma, e
- treating refers to any indicia of clinical success in the therapy or amelioration of a disease (e.g., cancer), including any objective or subjective parameter such as abatement; remission; diminishing of symptoms or making the disease more tolerable to the patient; slowing in the rate of degeneration or decline; making the final point of degeneration less debilitating; improving a patient’s physical or mental well-being.
- a disease e.g., cancer
- the treatment or amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination.
- the term “treating” does not include preventing.
- “Patient” or “subject in need thereof’ refers to a living organism suffering from or prone to a disease n that can be treated by administration of a compound or pharmaceutical composition herein.
- Non-limiting examples include humans, other mammals, bovines, rats, mice, dogs, cats, monkeys, goat, sheep, cows, and other non-mammalian animals.
- a patient is human.
- Cancer model organism is an organism exhibiting a phenoty pe indicative of cancer, or the activity of cancer causing elements, within the organism.
- the term cancer is defined above.
- a wide variety of organisms may serve as cancer model organisms, and include for example, cancer cells and mammalian organisms such as rodents (e.g. mouse or rat) and primates (such as humans).
- Cancer cell lines are widely understood by those skilled in the art as cells exhibiting phenotypes or genotypes similar to in vivo cancers. Cancer cell lines as used herein includes cell lines from animals (e.g. mice) and from humans.
- immune checkpoint inhibitor refers to a compound (e g., an antibody) that is capable of binding to an inhibitory receptor or capable of interfering with the interaction between an inhibitory receptor and its ligand, wherein the inhibitory receptor is essential to balance co-stimulatory receptor activity and limit T-cell activation.
- immune checkpoint inhibitors target immune system checkpoints such as the PD-1 pathway.
- PD-1 pathway inhibitor refers to a substance capable of detectably lowering expression of or activity level of the PD-1 signaling pathway compared to a control.
- An “inhibitor” is a compound or small molecule that inhibits the PD-1 signaling pathway e.g.. by binding, partially or totally blocking stimulation of the PD-1 pathway, decrease, prevent, or delay activation of the PD-1 pathway, or inactivate, desensitize, or down-regulate signal transduction, gene expression or enzymatic activity of the PD-1 pathway.
- the PD-1 pathway inhibitor is a programmed death-ligand 1 (PD-L1) inhibitor or a PD-1 inhibitor.
- a PD-L1 inhibitor is a substance that, at least in part, partially or totally blocks stimulation, decreases, prevents, or delays activation, or inactivates, desensitizes, or down-regulates signal transduction of PD-1.
- a PD-1 inhibitor is a substance that, at least in part, partially or totally blocks stimulation, decreases, prevents, or delays activation, or inactivates, desensitizes, or down-regulates signal transduction of PD-1.
- the methods further comprise administering to the patient an effective amount of an anti-cancer agent.
- Anticancer agent is used in accordance with its plain ordinary meaning and refers to a composition (e.g. compound, drug, antagonist, inhibitor, modulator) having antineoplastic properties or the ability to inhibit the growth or proliferation of cells.
- Exemplary anti-cancer agents include antibodies, small molecules, large molecules, and combinations thereof.
- an anti-cancer agent is a chemotherapeutic.
- an anti-cancer agent is an agent identified herein having utility in methods of treating cancer.
- an anti-cancer agent is an agent approved by the FDA or similar regulatory agency of a country other than the USA. for treating cancer.
- anti-cancer agents include, but are not limited to, MEK (e.g. MEK1, MEK2, or MEK1 and MEK2) inhibitors (e.g. XL518, CI-1040, PD035901, selumetinib/ AZD6244, GSK1120212/ trametmib, GDC-0973, ARRY-162, ARRY-300, AZD8330, PD0325901, U0126, PD98059, TAK-733, PD318088, AS703026, BAY 869766), alkylating agents (e.g., cyclophosphamide, ifosfamide, chlorambucil, busulfan, melphalan.
- MEK e.g. MEK1, MEK2, or MEK1 and MEK2
- MEK inhibitors e.g. XL518, CI-1040, PD035901, selumetinib/ AZD6244, GSK11202
- nitrogen mustards e.g., mechloroethamine, cyclophosphamide, chlorambucil, meiphalan
- ethylenimine and methylmelamines e.g., hexamethlymelamine, thi
- folic acid analog e.g., methotrexate
- pyrimidine analogs e.g., fluorouracil, floxouridine, Cytarabine
- purine analogs e.g., mercaptopurine, thioguanine, pentostatin
- plant alkaloids e.g., vincristine, vinblastine, vinorelbine,
- topoisomerase inhibitors e.g., irinotecan, topotecan, amsacrine, etoposide (VP 16), etoposide phosphate, teniposide, etc.
- antitumor antibiotics e.g., doxorubicin, adriamycin, daunorubicin, epirubicin, actinomycin, bleomycin, mitomycin, mitoxantrone, plicamycin, etc.
- platinum-based compounds or platinum containing agents e.g.
- Taxol.TM i.e. paclitaxel
- Taxotere.TM compounds comprising the taxane skeleton, Erbulozole (i.e. R-55104), Dolastatin 10 (i.e. DLS-10 and NSC-376128), mivobulin isethionate (i.e. as CI-980), vincristine, NSC-639829, Discodermolide (i.e. as NVP- XX-A-296), ABT-751 (Abbott, i.e. E-7010), Altorhyrtins (e.g. Altorhyrtin A and Altorhyrtin C), Spongistatins (e.g.
- Epothilones e.g. Epothilone A, Epothilone B, Epothilone C (i.e. desoxy epothilone A or dEpoA), Epothilone D (i.e. KOS-862
- Epothilone B N-oxide Epothilone A N-oxide, 16-aza-epothilone B, 21-aminoepothilone B (i.e. BMS-310705), 21 -hydroxy epothilone D (i.e. Desoxyepothilone F and dEpoF), 26-fluoroepothilone, Auristatin PE (i.e. NSC-654663), Soblidotin (i.e. TZT-1027), , Vincristine sulfate, Cryptophycin 52 (i.e. LY -355703), Vitilevuamide. Tubulysin A. Canadensol, Centaureidin (i.e.
- NSC-106969 Oncocidin Al (i.e. BTO-956 and DIME), Fijianolide B, Laulimalide, Narcosine (also known as NSC-5366), Nascapine, Hemiasterlin, Vanadocene acetylacetonate, Monsatrol, Inanocine (i.e.
- Eleutherobins such as Desmethyleleutherobin, Desaetyleleutherobin, Isoeleutherobin A, and Z- Eleutherobin
- caribaeoside caribaeolin
- Halichondrin B Diazonamide A
- Taccalonolide A Diozostatin
- (-)-Phenylahistin i.e.
- NSCL-96F0357 Myoseverin B, Resverastatin phosphate sodium, steroids (e.g., dexamethasone), finasteride, aromatase inhibitors, gonadotropin-releasing hormone agonists (GnRH) such as goserelin or leuprolide, adrenocorticosteroids (e.g., prednisone), progestins (e.g., hydroxyprogesterone caproate, megestrol acetate, medroxyprogesterone acetate), estrogens (e.g., diethlystilbestrol, ethinyl estradiol), antiestrogen (e.g., tamoxifen), androgens (e.g., testosterone propionate, fluoxymesterone), antiandrogen (e.g., flutamide), immunostimulants (e.g., Bacillus Calmette-Guerin (BCG), levamisole, interleukin
- anti-CD52, anti- HLA-DR, and anti-VEGF monoclonal antibodies include immunotoxins (e.g., anti-CD33 monoclonal antibody -calicheamicin conjugate, anti-CD22 monoclonal antibody-pseudomonas exotoxin conjugate, etc.), radioimmunotherapy (e.g., anti-CD20 monoclonal antibody conjugated to ni In, 90 Y, or 131 L etc ), triptolide, homoharringtonine, dactinomycin, doxorubicin, epirubicin, topotecan, itraconazole, vindesine, cerivastatin, vincristine, deoxyadenosine, sertraline, pitavastatin, irinotecan, clofazimine, 5 -nonyl oxy tryptamine, vemurafenib, dabrafenib, erlotinib, gefitinib, EGFR inhibitors,
- TAK-285 TAK-285, AST-1306, ARRY334543, ARRY-380, AG-1478, dacomitinib/PF299804, OSI-420/desmethyl erlotinib, AZD8931, AEE788, pelitinib/EKB-569, CUDC-101, WZ8040, WZ4002, WZ3146, AG-490, XL647, PD153035, BMS-599626), sorafenib, imatinib, sunitinib, dasatinib, hormonal therapies, or the like.
- Embodiment 1 A compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein: (a) the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and (b) the second DNA oligonucleotide comprises: (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5‘ end.
- Embodiment 2 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end.
- Embodiment 3 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and two constrained nucleotides on the 5’ end.
- Embodiment 4 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and one constrained nucleotide on the 5’ end.
- Embodiment 5 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotide on the 5’ end.
- Embodiment 6 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises two constrained nucleotides on the 3' end and three constrained nucleotides on the 5’ end.
- Embodiment 7 The compound of Embodiment 1. wherein the second DNA oligonucleotide comprises three constrained nucleotides on the 3 ’ end and two constrained nucleotides on the 5’ end.
- Embodiment 8 The compound of any one of Embodiments 3 to 7, wherein the two constrained nucleotides are contiguous.
- Embodiment 9 The compound of any one of Embodiments 3 to 7, wherein the two constrained nucleotides are alternating.
- Embodiment 10 The compound of Embodiment 1, wherein the second DNA oligonucleotide comprises three constrained nucleotides on the 3' end and three constrained nucleotides on the 5’ end.
- Embodiment 11 The compound of Embodiment 6, 7, or 9, wherein the three constrained nucleotides are contiguous.
- Embodiment 12 The compound of Embodiment 6, 7, or 9, wherein the constrained nucleotides are alternating.
- Embodiment 13 The compound of any one of Embodiments 1 to 12, wherein the constrained nucleotide is a LNA-modified nucleotide.
- Embodiment 14 The compound of any one of Embodiments 1 to 12, wherein the constrained nucleotide is a cMOE-modified nucleotide.
- Embodiment 15 The compound of any one of Embodiments 1 to 12, wherein the constrained nucleotide is a cET-modified nucleotide.
- Embodiment 16 The compound of any one of Embodiments 1 to 12, wherein the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET- modified nucleotide.
- Embodiment 17 The compound of any one of Embodiments 1 to 16, wherein the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 22 unmodified nucleotides; and the second DNA oligonucleotide comprises from about 10 nucleotides to about 22 nucleotides.
- Embodiment 18 The compound of Embodiment 17. wherein the first DNA oligonucleotide comprises from about 14 unmodified nucleotides to about 18 unmodified nucleotides; and the second DNA oligonucleotide comprises from about 14 nucleotides to about 18 nucleotides.
- Embodiment 19 The compound of any one of Embodiments 1 to 18, wherein the first DNA oligonucleotide and the second DNA oligonucleotide comprise the same number of nucleotides.
- Embodiment 20 The compound of any one of Embodiments 1 to 19, wherein the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- Embodiment 21 The compound of any one of Embodiments 1 to 20, wherein the second DNA oligonucleotide further comprises a nucleotide having a modification selected from the group consisting of 2’-O-aminopropyl group, a 2’-0-ethyl group, a 2’-fluoro group, a 2’-O- methyl group, 2’-deoxy-2’fluoro group, a 2’-O-methoxyethyl group, a 2’-O-allyl group, a 2’-O- propyl group, a 2’-O-pentyl group, and a constrained nucleotide.
- Embodiment 22 The compound of any one of Embodiments 1 to 21, wherein the second DNA oligonucleotide further comprises a spacer modification.
- Embodiment 23 The compound of any one of Embodiments 1 to 22, wherein the second DNA oligonucleotide is an antisense oligonucleotide.
- Embodiment 24 The compound of Embodiment 23, wherein the antisense oligonucleotide is a STAT3 antisense oligonucleotide.
- Embodiment 25 The compound of Embodiment 24, wherein the STAT3 antisense oligonucleotide comprises SEQ ID NO:2. SEQ ID NO:4. SEQ ID NO:6, SEQ ID NO:8, SEQ ID NOTO, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO:35, or SEQ ID NO:36.
- Embodiment 26 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO: 1 and the STAT3 antisense oligonucleotide comprises SEQ ID NOT.
- Embodiment 27 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NOT and the STAT3 antisense oligonucleotide comprises SEQ ID NO:4.
- Embodiment 28 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO:5 and the STAT3 antisense oligonucleotide comprises SEQ ID NO:6.
- Embodiment 29 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO:7 and the STAT3 antisense oligonucleotide comprises SEQ ID NO: 8.
- Embodiment 30 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO:9 and the STAT3 antisense oligonucleotide comprises SEQ ID NOTO.
- Embodiment 31 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO: 11 and the STAT3 antisense oligonucleotide comprises SEQ ID NO: 12.
- Embodiment 32 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NO: 13 and the STAT3 antisense oligonucleotide comprises SEQ ID NO: 14.
- Embodiment 33 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NOT and the STAT3 antisense oligonucleotide comprises SEQ ID NO:35.
- Embodiment 34 The compound of Embodiment 24, wherein the first DNA oligonucleotide comprises SEQ ID NOT and the STAT3 antisense oligonucleotide comprises SEQ ID NO:36.
- Embodiment 35 The compound of any one of Embodiments 1 to 34, wherein the phosphorothioated CpG oligodeoxynucleotide is Class A CpG oligodeoxynucleotide, a Class B CpG oligodeoxynucleotide, or a Class C CpG oligodeoxynucleotide.
- Embodiment 36 The compound of any one of Embodiments 1 to 34, wherein the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43, SEQ ID NO: 15, SEQ ID NO:16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO:20, SEQ ID NO 21, SEQ ID NO:22, SEQ ID NO:23. SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26. SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NOTO, SEQ ID NO:31, or SEQ ID NO:32.
- Embodiment 37 The compound of Embodiment 36, wherein the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43.
- Embodiment 38 The compound of Embodiment 36, wherein the phosphoro thioated CpG oligodeoxynucleotide comprises SEQ ID NO: 15.
- Embodiment 39 The compound of any one of Embodiments 1 to 38, wherein the first DNA oligonucleotide is a passenger strand and the second DNA oligonucleotide is a guide strand.
- Embodiment 40 The compound of any one of Embodiments 1 to 39, wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- Embodiment 41 The compound of any one of Embodiments 1 to 39, wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 3’ end of the first DNA oligonucleotide.
- Embodiment 42 The compound of Embodiment 1, wherein the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43; the first DNA oligonucleotide comprises SEQ ID NO: 1; the second DNA oligonucleotide comprises SEQ ID NO:2; and the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- Embodiment 43 The compound of Embodiment 1, wherein the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO: 15; the first DNA oligonucleotide comprises SEQ ID NO:3; the second DNA oligonucleotide comprises SEQ ID NO:4; and the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide.
- Embodiment 44 The compound of any one of Embodiments 1 to 43, wherein the linking group comprises a bond, a nucleic acid, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or unsubstituted cycloalkylene, substituted or unsubstituted heterocycloalkylene, substituted or unsubstituted arylene, substituted or unsubstituted heteroarylene, or a combination of two or more thereof.
- Embodiment 45 The compound of Embodiment 44, wherein the linking group comprises substituted 6 to 60 membered heteroalkylene.
- Embodiment 46 The compound of Embodiment 45, wherein the linking group comprises substituted heteroalkylene of the formula: , wherein n is an integer from 1 to 10.
- Embodiment 47 The compound of Embodiment 46, wherein n is 5.
- Embodiment 48 A pharmaceutical composition comprising the compound of any one of Embodiments 1 to 47 and a pharmaceutically acceptable excipient.
- Embodiment 49 A method of treating cancer in a patient in need thereof, the method comprising administering to the patient an effective amount of the compound of any one of Embodiments 1 to 47 or the pharmaceutical composition of Embodiment 48.
- Embodiment 50 The method of Embodiment 49, wherein the cancer is glioma, cranial primitive neuroectodermal tumor, ependymal tumor, hemangiopericytoma, germ cell tumor, pineal tumor, or primary' central nervous system lymphoma.
- Embodiment 51 The method of Embodiment 50, wherein the central nervous system cancer is glioma, cranial primitive neuroectodermal tumor, ependymal tumor, hemangiopericytoma, germ cell tumor, pineal tumor, or primary’ central nervous system lymphoma.
- Embodiment 52 The method of Embodiment 51 , wherein the glioma is astrocytoma, glioblastoma, or oligodendroglioma; the cranial primitive neuroectodermal tumor is medulloblastoma, cerebral neuroblastoma, pineoblastoma, or esthesioneuroblastoma; and the ependymal tumor is ependymoma, myxopapillary ependy moma, or subependymoma.
- the glioma is astrocytoma, glioblastoma, or oligodendroglioma
- the cranial primitive neuroectodermal tumor is medulloblastoma, cerebral neuroblastoma, pineoblastoma, or esthesioneuroblastoma
- the ependymal tumor is ependymoma, myxopapillary
- Embodiment 53 The method of Embodiment 49, wherein the cancer is glioma.
- Embodiment 54 The method of Embodiment 49, wherein the cancer is leukemia.
- Embodiment 55 The method of Embodiment 49, wherein the cancer is a solid tumor cancer.
- Embodiment 56 The method of Embodiment 49, wherein the cancer is prostate cancer, breast cancer, colorectal cancer, bladder cancer, lung cancer, liver cancer, pancreatic caner, renal cancer, stomach cancer, or melanoma.
- Embodiment 57 The method of Embodiment 49, wherein the cancer is prostate cancer.
- Embodiment 58 The method of Embodiment 49, wherein the cancer is an epidermoid carcinoma.
- Embodiment 60 The method of any one of Embodiments 49 to 59, further comprising administering to the patient an effective amount of an immune checkpoint inhibitor.
- Embodiment 61 The method of Embodiment 60, wherein the immune checkpoint inhibitor is a PD-1 inhibitor.
- Embodiment 62 The method of Embodiment 61, wherein the PD-1 inhibitor is pembrolizumab, nivolumab, cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, spartalizumab, retifanlimab, pimivalimab, AMP -224, or MEDI0680.
- the PD-1 inhibitor is pembrolizumab, nivolumab, cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, spartalizumab, retifanlimab, pimivalimab, AMP -224, or MEDI0680.
- Embodiment 63 The method of Embodiment 60, wherein the immune checkpoint inhibitor is a PD-Ll inhibitor.
- Embodiment 64 The method of Embodiment 63, wherein the PD-L1 inhibitor is atezolizumab, avelumab. durvalumab, envafolimab. cosibelimab, AUNP12, CA-170, or BMS- 986189.
- Embodiment 65 A pharmaceutical composition comprising the compound of any one of Embodiments 1 to 47 and an immune checkpoint inhibitor.
- Embodiment 66 The pharmaceutical composition of Embodiment 65, wherein the immune checkpoint inhibitor is a PD-inhibitor or a PD-L1 inhibitor.
- Embodiment 67 A STAT3 antisense oligonucleotide comprising SEQ ID NO: 4.
- Embodiment Nl A compound comprising a phosphorothioated CpG oligodeoxynucleotide linked to a first DNA oligonucleotide that is hybridized to a second DNA oligonucleotide; wherein: (a) the first DNA oligonucleotide comprises from about 5 unmodified nucleotides to about 70 unmodified nucleotides; and (b) the second DNA oligonucleotide comprises: (i) from about 5 nucleotides to about 70 nucleotides, and (ii) a constrained nucleotide on the 3’ end and a constrained nucleotide on the 5’ end.
- Embodiment N2 The compound of Embodiment N 1 , wherein the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and one constrained nucleotide on the 5’ end. [0233] Embodiment N3.
- the compound of Embodiment N 1 wherein: (i) the second DNA oligonucleotide comprises one constrained nucleotide on the 3’ end and two constrained nucleotides on the 5’ end; (ii) the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and one constrained nucleotide on the 5’ end; (iii) the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and two constrained nucleotide on the 5’ end; (iv) the second DNA oligonucleotide comprises two constrained nucleotides on the 3’ end and three constrained nucleotides on the 5’ end; (v) the second DNA oligonucleotide comprises three constrained nucleotides on the 3’ end and two constrained nucleotides on the 5’ end; or (vi) the second DNA oligonucleotide comprises three constrained nucleot
- Embodiment N4 The compound of Embodiment N3, wherein the two or three constrained nucleotides are contiguous or alternating.
- Embodiment N5 The compound of any one of Embodiments N1 to N4, wherein the constrained nucleotide is a LNA-modified nucleotide, a cMOE-modified nucleotide, or a cET- modified nucleotide.
- Embodiment N6 The compound of any one of Embodiments N 1 to N5, wherein the first DNA oligonucleotide comprises from about 10 unmodified nucleotides to about 22 unmodified nucleotides; and the second DNA oligonucleotide comprises from about 10 nucleotides to about 22 nucleotides.
- Embodiment N7 The compound of any one of Embodiments N1 to N6, wherein the second DNA oligonucleotide comprises a nucleotide having a phosphorothioate modification.
- Embodiment N8 The compound of any one of Embodiments N1 to N7, wherein the second DNA oligonucleotide further comprises a nucleotide having a modification selected from the group consisting of 2’-O-aminopropyl group, a 2’-0-ethyl group, a 2’-fluoro group, a 2’-O- methyl group, 2’-deoxy-2’fluoro group, a 2’-O-methoxyethyl group, a 2’-O-allyl group, a 2’-O- propyl group, a 2’-O-pentyl group, and a constrained nucleotide.
- Embodiment N9 The compound of any one of Embodiments N1 to N8, wherein the second DNA oligonucleotide is an antisense oligonucleotide.
- Embodiment N10 The compound of any one of Embodiments N1 to N8, wherein the second DNA oligonucleotide is a STAT3 antisense oligonucleotide.
- Embodiment N11 The compound of Embodiment N 10, wherein the STAT3 antisense oligonucleotide comprises SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8, SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO 35, or SEQ ID NO:36.
- Embodiment N12 The compound of any one of Embodiments N1 to Ni l, wherein: (a) the first DNA oligonucleotide comprises SEQ ID NO: 1 and the STAT3 antisense oligonucleotide comprises SEQ ID NO:2; (b) the first DNA oligonucleotide comprises SEQ ID NO:3 and the STAT3 antisense oligonucleotide comprises SEQ ID NO:4; (c) the first DNA oligonucleotide comprises SEQ ID NO:5 and the STAT3 antisense oligonucleotide comprises SEQ ID NO:6; (d) the first DNA oligonucleotide comprises SEQ ID NO:7 and the STAT3 antisense oligonucleotide comprises SEQ ID NO: 8; (e) the first DNA oligonucleotide comprises SEQ ID NO:9 and the STAT3 antisense oligonucleotide comprises SEQ ID NOTO; (a) the
- Embodiment N13 The compound of any one of Embodiments N1 to N12, wherein the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43, SEQ ID NO: 15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO:20, SEQ ID NO 21, SEQ ID NO:22, SEQ ID NO:23. SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26. SEQ ID NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO 30, SEQ ID NO:31, or SEQ ID NO:32.
- Embodiment N 14 The compound of any one of Embodiments N1 to N13, wherein the first DNA oligonucleotide is a passenger strand and the second DNA oligonucleotide is a guide strand.
- Embodiment N15 The compound of any one of Embodiments N1 to N14, wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oligonucleotide or wherein the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 3’ end of the first DNA oligonucleotide.
- Embodiment N 16 The compound of Embodiment Nl, wherein: (a) the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO:43; the first DNA oligonucleotide comprises SEQ ID NO: 1; the second DNA oligonucleotide comprises SEQ ID NO:2; and the 3‘ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5‘ end of the first DNA oligonucleotide; or (b) the phosphorothioated CpG oligodeoxynucleotide comprises SEQ ID NO: 15; the first DNA oligonucleotide comprises SEQ ID NO:3; the second DNA oligonucleotide comprises SEQ ID NO:4; and the 3’ end of the phosphorothioated CpG oligodeoxynucleotide is linked to the 5’ end of the first DNA oli
- Embodiment N A pharmaceutical composition comprising the compound of anyone of Embodiments N1 to N16 and a pharmaceutically acceptable excipient.
- Embodiment N 18. A method of treating cancer in a patient in need thereof, the method comprising administering to the patient an effective amount of the compound of any one of Embodiments N1 to N16 or the pharmaceutical composition of Embodiment N17.
- Embodiment N19 The method of Embodiment N18, wherein the cancer is glioma.
- Embodiment N20 The method of Embodiment N18, wherein the cancer is a central nervous system cancer, leukemia, prostate cancer, breast cancer, colorectal cancer, bladder cancer, lung cancer, liver cancer, pancreatic caner, renal cancer, stomach cancer, melanoma, or an epidermoid carcinoma.
- the cancer is a central nervous system cancer, leukemia, prostate cancer, breast cancer, colorectal cancer, bladder cancer, lung cancer, liver cancer, pancreatic caner, renal cancer, stomach cancer, melanoma, or an epidermoid carcinoma.
- Embodiment N21 The method of any one of Embodiments N18 to N20, further comprising administering to the patient an effective amount of an immune checkpoint inhibitor.
- Embodiment N22 The method of Embodiment N21, wherein the immune checkpoint inhibitor is a PD-1 inhibitor or a PD-L1 inhibitor.
- Embodiment N23 The method of Embodiment N22, wherein: (i) the PD-1 inhibitor is pembrolizumab, nivolumab, cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, spartalizumab, retifanlimab, pimivalimab, AMP-224, or MEDI0680, and (ii) the PD-L1 inhibitor is atezolizumab, avelumab, durvalumab, envafolimab, cosibelimab, AUNP12, CA-170, or BMS-986189.
- the PD-1 inhibitor is pembrolizumab, nivolumab, cemiplimab, dostarlimab, camrelizumab, sintilimab, tislelizumab, toripalimab, spart
- Example 1 To augment the STAT3 knockdown in glioma cells, locked-nucleic acid (LNA) chemistn’ was used within ASO part of the molecule.
- LNA locked-nucleic acid
- FIG. 1 double-stranded ASO molecules comprising a gapmer ASO hybridized to a complementary and not phosphorothioate DNA strand are stable in human serum (FIG. 1A) and result in target gene knock down comparable to a single-stranded ASO (FIGS. 1B-1D) in various cancer cells. Similar effects were observed for dsASO targeting STAT3 as well as dsASO specific to a different gene, androgen receptor in prostate cancer cells (FIG. 1C).
- This design was used to create a TLR9-targeted CpG-dsASO conjugate (FIG. 2A).
- the conjugate showed over 2 days half-life in human serum and was still detectable after 5 days of incubation (FIG. 2B).
- the CpG-STAT3dsASO w as confirmed to have similar potency and kinetics of target gene knockdown as a single stranded STAT3ASO alone in cancer cells, such as prostate cancer, epidermoid carcinoma, glioma cells, and mouse macrophages (FIGS. 2C-2D, FIG. 3).
- CpG-STAT3ASO variants were rapidly internalized by human and mouse glioma cells and myeloid cells in vitro (FIGS. 4-6). None of the STAT3ASO variants w as internalized by human T cells, while mouse T cells showed low’ internalization of high oligonucleotide concentrations at longer incubation times only. Biodistribution experiments in mice with intracranial gliomas using intratumoral oligonucleotide injections confirmed very efficient uptake by a variety of myeloid cells in the brain including macrophages, microglia, DCs and MDSCs (FIG. 7).
- CpG-STAT3ASOs Local administration of all tested single- and double-stranded CpG-STAT3ASOs improved animal survival and triggered immune activation in immunocompetent mice. All three types of CpG- STAT3ASO injections induced maturation/ activation of intratumoral DCs, macrophages and microglia, while reducing numbers of tumor-associated M2 macrophages and resting microglia as assessed using flow cytometry (FIG. 10B). Importantly, CpG-STAT3ASO injections improved the ratio of intratumoral CD8 T cells to Tregs (FIG. IOC).
- LNA-modified CpG-STAT3dsASO treatment was combined with systemic administration of PD1 blocking antibodies.
- the combination treatment produced long-term tumor free survival of 5/6 treated mice for over 150 days.
- the locally injected LNA- modified CpG-STAT3dsASO showed efficacy also against RM9 prostate tumors in mice, with evidence of abscopal effects (FIG. 12).
- LNA-modified CpG-STAT3dsASO activity was verified in the new model of immune checkpoint resistant QPP8 (Ok Tp53 len-de ) glioma derived from neural stem cells. As shown in FIG. 13. LNA-modified CpG-STAT3dsASO showed improved antitumor effect as a single agent compared to the single-stranded LNA-modified CpG-STAT3ASO. When combined with PDl-specific antibodies, LNA-modified CpG-STAT3dsASO resulted in complete regression of QPP8 gliomas in the majority of treated mice (FIG. 14).
- the section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents. cited in the application are expressly incorporated by reference herein in their entirety and for all purposes.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Genetics & Genomics (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Organic Chemistry (AREA)
- Veterinary Medicine (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biophysics (AREA)
- General Engineering & Computer Science (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Mycology (AREA)
- Biotechnology (AREA)
- Physics & Mathematics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Plant Pathology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Description
Claims
Priority Applications (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2025519755A JP2025535719A (en) | 2022-10-07 | 2023-10-06 | Double-stranded oligonucleotides and methods of use |
| EP23875846.0A EP4599065A2 (en) | 2022-10-07 | 2023-10-06 | Double-stranded oligonucleotides and methods of use |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263414160P | 2022-10-07 | 2022-10-07 | |
| US63/414,160 | 2022-10-07 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2024077228A2 true WO2024077228A2 (en) | 2024-04-11 |
| WO2024077228A3 WO2024077228A3 (en) | 2024-05-30 |
Family
ID=90608892
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2023/076223 Ceased WO2024077228A2 (en) | 2022-10-07 | 2023-10-06 | Double-stranded oligonucleotides and methods of use |
Country Status (3)
| Country | Link |
|---|---|
| EP (1) | EP4599065A2 (en) |
| JP (1) | JP2025535719A (en) |
| WO (1) | WO2024077228A2 (en) |
Family Cites Families (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2015077657A2 (en) * | 2013-11-22 | 2015-05-28 | City Of Hope | Stat3 inhibitors and uses thereof |
| CN114224907A (en) * | 2015-07-02 | 2022-03-25 | 希望之城 | Compounds and compositions comprising phosphorothioated oligodeoxynucleotides and methods of use thereof |
-
2023
- 2023-10-06 WO PCT/US2023/076223 patent/WO2024077228A2/en not_active Ceased
- 2023-10-06 JP JP2025519755A patent/JP2025535719A/en active Pending
- 2023-10-06 EP EP23875846.0A patent/EP4599065A2/en active Pending
Also Published As
| Publication number | Publication date |
|---|---|
| WO2024077228A3 (en) | 2024-05-30 |
| JP2025535719A (en) | 2025-10-28 |
| EP4599065A2 (en) | 2025-08-13 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| ES2989617T3 (en) | Chemokine receptor modulators and their uses | |
| TWI838350B (en) | Anticancer agents | |
| CN103717214A (en) | Compounds and compositions for stabilizing hypoxia inducible factor-2 alpha as a method for treating cancer | |
| WO2006093991A1 (en) | Compounds which bind psma and uses thereof | |
| WO2019090347A1 (en) | Adenosine pathway inhibitors for cancer treatment | |
| KR20170029565A (en) | Combination therapy for cancer | |
| AU2020368368A1 (en) | Broad spectrum anti-cancer compounds | |
| US20220370434A1 (en) | Methods and compositions for treating cancer | |
| WO2022246597A1 (en) | Imidazopyridine derivatives as sting agonists | |
| WO2015026935A2 (en) | Hdac8 inhibitors for treating cancer | |
| US20210101924A1 (en) | Cyclic dinucleotide compounds as sting agonists | |
| WO2017190107A1 (en) | Sigma receptor binders | |
| WO2020068600A1 (en) | Ubiquitin-specific-processing protease 7 (usp7) modulators and uses thereof | |
| AU2019201104A1 (en) | Compounds and methods for treating cancer, neurological disorders, ethanol withdrawal, anxiety, depression and neuropathic pain | |
| TW202003520A (en) | Biomarkers for TTK inhibition | |
| US12472209B2 (en) | Neural stem cell delivery of therapeutic agents | |
| JP2023550049A (en) | IRE1 alpha inhibitor and its use | |
| US11739121B2 (en) | EPHA2 agonists and uses thereof | |
| US20250376689A1 (en) | Oligonucleotides having 6-thio-2'-deoxyguanosine residues and uses thereof | |
| WO2024077228A2 (en) | Double-stranded oligonucleotides and methods of use | |
| US20230220385A1 (en) | Microrna as a therapeutic agent | |
| JP2020506935A (en) | Compositions and methods for inhibiting reticulon 4 | |
| KR100898330B1 (en) | Novel flavonoid derivatives 7-O- (3-benzyloxypropyl) -5,4'-O-dimethyl-apigenin having anticancer properties, preparations thereof and anticancer compositions comprising the same | |
| CN111587113A (en) | Combination therapy for cancer treatment | |
| WO2022093857A1 (en) | Oncolytic virus compositions and methods for the treatment of cancer |
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: 23875846 Country of ref document: EP Kind code of ref document: A2 |
|
| ENP | Entry into the national phase |
Ref document number: 2025519755 Country of ref document: JP Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025519755 Country of ref document: JP |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2023875846 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2023875846 Country of ref document: EP Effective date: 20250507 |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23875846 Country of ref document: EP Kind code of ref document: A2 |
|
| WWP | Wipo information: published in national office |
Ref document number: 2023875846 Country of ref document: EP |