Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CANCER CHEMOPREVENTION WITH STAT3 BLOCKERS
Document Type and Number:
WIPO Patent Application WO/2019/191493
Kind Code:
A1
Abstract:
Described herein are compositions that inhibit/degrade STAT3, and methods of using such compositions for chemoprevention of non-small cell lung cancer (NSCLC).

Inventors:
SIEGFRIED JILL M (US)
NJATCHA CHRISTIAN NZINKEU (US)
FAROOQUI MARIYA (US)
Application Number:
PCT/US2019/024682
Publication Date:
October 03, 2019
Filing Date:
March 28, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV MINNESOTA (US)
SIEGFRIED JILL M (US)
NJATCHA CHRISTIAN NZINKEU (US)
FAROOQUI MARIYA (US)
International Classes:
C07K14/74; A61K48/00
Domestic Patent References:
WO2011163423A22011-12-29
WO2019036509A12019-02-21
Foreign References:
US20130129675A12013-05-23
Other References:
HE, GL ET AL.: "Inhibition of STAT3- and MAPK-dependent PGE2 synthesis ameliorates phagocytosis of fibrillar [betaj-amyloid peptide (1-42) via EP 2 receptor in EMF-stimulated N9 microglial cells", JOURNAL OF NEUROINFLAMMATION, vol. 13, no. 296, 21 November 2016 (2016-11-21), pages 1 - 14, XP055638656
Attorney, Agent or Firm:
ANDRUS, Alex et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for the prevention of cancer by administering to a patient in need or at risk a therapeutically-effective dose of a STATS Blocker/Degrader.

2. The method of claim 1 wherein the STAT3 Blocker modulates one or more targets selected from STATS mRNA, STATS monomer, phosphorylated STATS, acetylated STATS, activated STATS, STATS homodimer, and STAT3-STAT1 heterodimer.

3. The method of claims 1 or 2 wherein the therapeutically-effective dose of a STATS Blocker is administered to the patient for chemoprevention of non-small cell lung cancer.

4. The method of any one of claims 1 to 3 wherein a tumor of the patient has an EGFR mutation or a KRAS mutation.

5. The method of claim 4 wherein the EGFR mutation is selected from L858R, an exon 19 deletion, T797M, and T790M.

6. The method of any one of claims 1 to 5 wherein the patient is at elevated risk of developing cancer, due to one or more factors selected from a genetic condition,

immunocompromised status, comorbidities, prior treatment for cancer, incidence of cancer, prior treatment with or exposure to one or more chemotherapeutic agents, radiation therapy, exposure to radiation or radon, prior exposure to or treatment with one or more carcinogenic or cytotoxic agents, and social or family history correlated with the incidence of cancer

7. The method of claim 6 wherein the patient is at risk of developing the T790M mutation after prior treatment with EGFR inhibitors.

8. The method of claim 6 wherein the patient has been exposed to asbestos.

9. The method of claim 6 wherein the patient is or has been a smoker

10. The method of any one of claims 1 to 9 wherein the STATS Blocker is selected from AZD-9150, napabucasin, WP1066, WP1120, and WP1732, or an analog thereof.

11. The method of any one of claims 1 to 9 wherein the STAT3 Blocker is an oligonucleotide.

12. Tire method of claim 11 wherein the STATS Blocker comprises an

oligonucleotide having the sequence:

5!-(N6)„CANiTTCN2CN3TN4AN5TC-(N7)s„-3' (SEQ ID NO: 15)

wherein N3 is G: N1, N2, N4, and N3 are A, T, G or C:

and one or more of the following conditions (i)-(v) are met: (i) N’! IS T; N2 is C; N4 is A; N5 is A; and N6 and N7 are independently selected from A,

T, G or C, and n and m are independently selected from an integer from 0 to 50;

(ii) the first end of a first strand is joined to a first end of the second strand by a hexa- ethyleneglycol spacer;

(in) the second end of the first strand is joined to a second end of the second strand by a hexa-eihyleneglycol spacer;

(iv) the oligonucleotide binds to STAT3 protein under physiologic conditions and interferes with STATS binding to its target sequence; and

(v) the oligonucleotide has a serum half-life of greater than about 4 hours.

13. The method of claim 1 1 wherein the oligonucleotide is a linear or partially linear oligonucleotide having any of the following structures (or analog thereof) (SEQ ID NOS 1-4 and 16, respectively, in order of appearance):

14. The method of claim 11 wherein the oligonucleotide comprises one or more sequences selected from SEQ ID NOS.: 1-4 where X is a hexaethyleneglycol, -(CTTCI-EOie spacer.

15. The method of claim 11 wherein the oligonucleotide comprises one or more intemucleotide phosphate analogs, selected from phosphorothioate, phosphoramidate, and alkylphosphonate, or one or more locked nucleic acid (LNA) nucleoside analogs.

16. The method of claim 1 1 wherein the oligonucleotide is CS3D, having the structure (SEQ ID NOS 4 and 16, respectively):

17. The method of claim 1 1 wherein the oligonucleotide has the structure (SEQ ID NOS 17, 4 and 16, respectively):

wherein o is a spacer moiety independently selected from C1-C12 alkylene, and - (CH2CH2OV where n is 1 to 6, and -P EP BORO .)-.

18. The method of claim 11 wherein the oligonucleotide is a cyclic oligonucleotide.

19. The method of claim 1 1 wherein the oligonucleotide comprises SEQ ID NO.:5 or SEQ ID NO.: 6 where (CS18) is a hexaethyleneglycol, -(CEkCEkO^ spacer.

20. The method of any one of the preceding claims further comprising administering a chemotherapeutic agent selected from erlotinib, afatinib, and osimertimb and/or a

chemopreveniion agent selected from the group consisting of aspirin, ibuprofen, an anti-oxidant, Iloprost, a drug of the glitazone class such as pioglitazone, and an agent that acti vates anti-tumor immunity, including pembrolizumab or mvoiumab.

21. A method of measuring one or more markers selected from IL6, COX2, and PGE2 as a means to detect biological activity of a STAT3 Blocker/Degrader m pati ents undergoing therapy or chemopreveniion.

Description:
CANCER CHEMOPREVENTION WITH STATS BLOCKERS

GOVERNMENT FUNDING

This invention was made with government support under F31-CA213982 awarded by the National Institutes of Health. The government has certain rights in the invention.

CROSS REFERENCE TO RELATED APPLICATION

This application claims priority to United States Provisional Application Number 62/650,892 filed on 30 March 2018, which is incorporated by reference herein.

SUBMISSION OF SEQUENCE LISTING

The content of the following submission on ASCII text file is incorporated herein by reference m entirety: a computer readable form (CRF) of the Sequence Listing (file name: 09531_462WOl_SL.txt, created: March 25, 2019, size 5,624).

FIELD OF THE INVENTION

Described herein are compositions that inhibit and/or degrade STATS, and methods of using such compositions for the cancer chemoprevention of STAT3-related disorders, for enhancing the immune system’s ability to reject cancer, and for using biomarkers to monitor the biological activity of compositions that inhibit and/or degrade STAT3.

BACKGROUND

Signal transducer and activator of transcription 3 (STAT3) mediates the expression of a variety of genes in response to cell stimuli and thus plays a key role in several cellular processes such as cell growth and apoptosis. Studies of the Janus kinases (JAKs) and signal transducers and activators of transcription (STATs) signaling have uncovered highly conserved programs linking cytokine signaling to the regulation of essential cellular mechanisms such as proliferation, invasion, survival, inflammation and immunity. Inhibitors of the JAK/STAT pathway are used for treatment of autoimmune diseases, such as rheumatoid arthritis or psoriasis. Aberrant JAK/STAT signaling has been identified to contribute to cancer progression and metastatic development. Deregulation of the STATS activity has been shown in many malignancies, including breast, head and neck, lung, prostate, pancreas, ovarian and brain cancers and melanoma.

Chemoprevention is distinguishable from treatment of cancer; in the former cancerous lesions may not be present. Treatment and chemoprevention may target different patient populations. There is a need to develop preventative treatment options for patients at risk of developing cancer, including non-small cell lung cancer (NSCLC). Safe and effective chemoprevention agents have enormous potential to improve human health. Effective preventive therapy has been demonstrated convincingly in cardiovascular disease, where modifiable risk factors such as high blood pressure and elevated cholesterol levels were identified and targeted for pharmacological management. The result has been the reduction of both the morbidity and mortality associated with heart disease. For some cancers, the promise of chemoprevention has already been realized. Recurrence of breast cancer is being reduced with tamoxifen therapy, and the incidence of colon cancer is being reduced significantly with NSAIDs

Lung cancer remains the leading cause of cancer-related mortality in the United States, representing more than 25% of all cancer deaths, which is largely attributed to the dismal survival of stage IV disease (Siegel RL, et al, Cancer Statistics: 2017. CA Cancer J Clin (2017) 67:7-30; Jemal A, et al JNatl Cancer Inst (2017) 109 (9); Torre LA, et al, Cancer Epidemiol Biomarkers Prev (2016) 25: 16-27). In NSCLC, which accounts for 85% of all lung cancer cases (Chen Z, et al Nat Rev Cancer (2014) 14:535-46), patients are stratified based on genetic alterations in the tumor, which serve as predictive biomarkers for targeted therapy. Patients with somatic epidermal growth factor receptor (EGFR) mutations (such as the L858R point mutation and exon 19 deletions) initially respond to EGFR tyrosine kinase inhibitors (TKis), but eventually develop acquired resistance through new mutations such as the EGFR T790M substitution. The majority of patients without EGFR mutation, includin those who lack alterations in other“drivers’, are intrinsically resistant to EGFR TKis (Gainor JF, et al, J Clin Oncol (2013) 31:3987-96).

Constitutively activated STAT3 plays a critical role NSCLC progression by mediating proliferation and survival. ' The activation of STAT3 in normal cells is transient, making it an attractive target for NSCLC therapy. STATS, a transcription factor that controls important growth-promoting genes, is a key mediator of receptor tyrosine kinase (RTK) downstream signaling. In NSCLC, STAT3 activation is correlated with heightened EGFR signaling, as well as increased c-Met and IL6 receptor signaling. STAT3 inhibition could also potentially be effecti ve when combined w th inhibitors of other RTKs, either in tumors with addiction to a specific pathway like FGFR or c-Met, or in patients with acquired EGFR TKI resistance due to upregulation of those pathways in addition to promoting tumor growth directly, STAT3 also is active in promoting immunosuppression.

STAT3 was often activated during in vitro induction of resistance to the EGFR inhibitor erlotinib (TARCEVA®, Genentech), as the result of feedback up-regulation of multiple other kinases, suggesting that increased STAT3 activity' could commonly occur in response to drugs that target kinases (Lee H-J, et al Cancer Cell (2014) 26:207-221). During lung cancer progression, STAT3 is also often constitutive! }' active, upregulating the expression of important target genes necessary for cellular proliferation, survival, and evasion of cell death (Dutta P, et al, JAKSTAT (2015) 3 (4), e999503. 2015 Jan 20). As a point of convergence for many signaling pathways that are dysregulated during NSCLC progression, STAT3 mediates adaptive mechanisms of resistance to molecular targeted therapy in NSCLC, such as induction of epithelial-to-mesenchymal transition by IL-6 in response to er!otinib (Yao Z, et al, Proc Natl AcadSci USA (2010) 107: 15535-40; Gao SP, et al, J Clin Invest (2007) 117:3846-56). Since STAT3 plays a critical role in malignant cell transformation, but is not essential for normal cell growth (Schlessinger K, et al, Cancer Research (2005) 65: 5828-5834), it is potentially a prime therapeutic target for cancer treatment or cancer chemoprevention.

STAT3 is a critical downstream mediator of tumor progression in NSCLC. As a transcription factor that regulates the expression of multiple target genes, STAT3 also relays signals through multiple receptor and non-receptor tyrosine kinases, including EGFR, IL-6R, and SRC. The key role of STAT3 in mediating proliferation and suppressing apoptosis in NSCLC makes it a prime target for therapeutic intervention. Efforts to apply STAT3 inhibition in the clinical setting have been limited due to a paucity of potent and selective inhibitors (Darnell JE. (2005) Nat. Med. 11:595-6; Chang, N. et ai. Mol. and Cellular Endocrin. (2017) 451:53-65; Pranabananda, D. et al, JAK-STAT. (2014) 3(4): e999503; Everardo, M. et al,

Journal of Skin Cancer, vol. 2013, Article ID 684050, 10 pages, 2013.

doi: 10.1155/2013/684050; Fagard, R., et al (2013) JAK-STAT 2(1) e22882; Furtek SL, et al, ACS Chem Biol (2016) 11 :308-18). Previous approaches to inhibit STAT3 include R A interference, peptidomimetics to prevent dimerization, and blockers of the STAT3 SH2 domain to prevent activation. STATS decoy oligonucleotides have been reported (Sen M, et al, Cancer Discov (2012) 8:694-705; US 2006/0293264; US 8722640; US 9062121). A STATS anti-sense oligonucleotide developed by ISlS/Eii Lilly showed low' clinical activity, due to incomplete downregulation of STAT3 gene expression. Most currently available STAT3 inhibitors either target upstream kinases, lack specificity, or require exceptionally high concentrations to achieve STAT3 inhibition. Previous STATS oligonucleotide decoys were unstable (Castanotto D, et al, Curr Opin Oncol (2014) Nov; 26:584-589; Dean NM, et al, Oncogene (2003) 22:9087-96) and small molecule STATS inhibitors proved relatively ineffective. The ability of non- phosphorylated STAT3 to function as an active dimer (Timofeeva OA, et al, J Biol Chem (2012) 287: 14192-14200; Y ang J, et al, Cancer Res (2005) 65:939-47) also may limit the ability of kinase inhibitors to completely block STAT3 action. SUMMARY

An aspect of the invention is a method for the chemoprevention of cancer by

administering to a patient in need or at risk a therapeutieally-effective dose of a STAT3 Blocker.

An aspect of the invention provides a means for chemoprevention using a STAT3 Decoy oligonucleotide, STATS antibody, STATS antisense, STATS locked nucleic acid, STATS aptamer, STATS small molecule inhibitor (as used herein "Blocker") or STATS degrader (as used herein“Degrader”) following exposure to any generally recognized carcinogenic or cytotoxic agent/agents. Figure 1 shows an exemplary mechanism of action where the STAT Decoy binding to pSTAT3 and blocking STAT3 dimers from binding DNA. In Figure 6B and 6C below, the ability of STATS Decoy to cause p-STAT3 to degrade is demonstrated.

Another aspect of the invention is a means for chemoprevention using a STATS

Blocker/Degrader targeting patient populations otherwise at elevated risk of developing cancer, including without limitation, due to genetic conditions, immunocompromised status, comorbidities, medical history' (including, prior treatment for cancer and presently in remission), prior treatment with or exposure to chemotherapeutic agents, commitment treatment with chemotherapy and/or radiation, prior exposure to or treatment with cytotoxic agents, prior treatment with or exposure to radiation or radon, social history, or family history·.

An aspect of the invention is the use of a cyclic oligonucleotide as a specific and potent STATS inhibitor to inhibit the growth of lung tumors with or without the T790M mutation or with or without intrinsic resistance to EGFR TKIs, by targeting Signal Transducer and Activator of Transcription 3 (STATS).

An aspect of the invention is a STATS Blocker/Degrader oligonucleotide.

The compositions of the invention target STATS.

Two other aspects of the invention are two potential biomarkers (detection of the cytokine mterleukin-6 (IL6) and the enzyme cyclo-oxygenase 2 (COX2) [or products of COX2 activity such as prostaglandin E2 [PGE2]) that can detect biological activity of a STATS inhibitor/blocker/degrader in blood products, in cells, in tissues or in the whole body.

Another aspect of the invention is the use of a STATS inhibitor/blocker/degrader as a means to enhance the activity of cells of the innate and/or adaptive immune system to reject cancerous cells in the body, to augment the activity of any cancer immunotherapies that are developed to increase the activity of cells m the innate and/or acquired immune system to attack and reject cancerous cells in the body.

Another aspect of the invention is a method of measuring one or more markers selected from IL6, COX2, and PGE as a means to detect biological activity of a STATS

Blocker/Degrader in patients undergoing therapy or chemoprevention. BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 shows an exemplary mechanism of action where the STAT Decoy binds to pSTAT3 and blocks STAT3 dimers from binding DNA.

Figure 2 shows CS3D, an exemplary embodiment of a cyclic oligonucleotide STAT Decoy with the forward and reverse STAT3 DNA consensus sequence and the cyclic structure after ligation. CS18 = hexaethyleneglycol - (CH 2 CH 2 0)6.

Figure 3A shows the effects of the cyclic STAT3 decoy CS3D relative to mutant control CS3M on in vitro cell viability 7 . NSCLC cell lines (201T and H1975) were transfected with CS3D or CS3M (cyclic STATS mutant decoy) at concentrations ranging from 0 nM to 600 nM.

Figure 3B shows uptake of fluorescein-labeled CS3D (cyclic STAT3 decoy) by NSCLC in vitro.

Figure 4A shows CS3D inhibits anchorage independent growth. NSCLC cell growth was assessed in colony forming assays in the presence of either 300 nM CS3D or CS3M.

Figure 4B shows detection of apoptosis by examining annexin V and propidium i odide (Pl)-positive cells by flow cytometry: H1975 and 201T cells transfected with lOOnM of CS3M or CS3D for 24hr were stained with apoptosis marker (annexin V) and cell viability dye.

Statistical significance was determined as *P<0.05, n=3.

Figure 5A shows down-regulation of e-Myc and bcl-xL mRNA expression by CS3D in NSCLC ceils. Ceils were treated 24 hours post-transfection with EGF (10 ng/m!) for 1.5 hr and mRNA was harvested from EGFR wild-type 201 T cells. mRNA expression wns assessed by RT- qPCR. Relative mRNA expression was normalized to GAPDH mRNA levels as an internal control. Statistical significance was determined as *P<0.05.

Figure 5B show's down-regulation of c-Myc and bcl-xL mRNA expression by CS3D in NSCLC cells. Cells were treated 24 hours post-transfection with EGF (10 ng/'ml) for 1.5 hr and mRNA w¾s harvested from mutant EGFR T790M HI 975 cells. mRNA expression was assessed by RT-qPCR. Relative mRNA expression was normalized to GAPDH mRNA levels as an internal control. Statistical significance was determined as *P<0 05.

Figure 6A shows down-regulation of c-Myc protein by CS3D. Cells were treated as in Figures 5A and 5B, and 3 hr after EGF treatment, lysates were collected from wild-type 201T (left) and mutant EGFR T797M HI 975 (right) NSCLC cell lines.

Figure 6B show's CS3D degrades phospho~STAT3. HI 975 were stimulated 24 hours post-transfection with either CS3M, CS3D, or lipofectamme alone with IL-6 (50ng/mL) for 1 hr to strongly activate STATS. After ceil lysis, nuclear and cytoplasmic extracts were obtained by subcellulax fractionation using centrifugation, and Western blotting was used to assessed to protein expression. GAPDH was used as a marker of the cytoplasmic fraction and histone H3 was used as a marker of the nuclear fraction. Densitometric quantification of the c-Myc bands are as indicated.

Figure 6C shows evidence that CS3D causes the degradation of phospho-STAT3 through the ubiquitin-proteasome pathway. The proportion of pSTAT3-ubiquitin bound complexes was increased by CS3D compared to control CS3M. 24 hours post-transfection, P-STAT3 was imrnunoprecipitated and the presence of ubiquitin was assessed by immunobl oiling, showing ability of CS3D to activate protein degradation of P-STAT3.

Figures 7A and 7B show CS3D suppresses NSCLC xenograft tumor growth. (A) Intravenous delivery of CS3D inhibits tumor growth in both (7 A) wild-type (201T) and (7B) mutant EGFR T790M (HI 975) xenografts. Approximately 1x10° cells were inoculated subcutaneously m the flanks of nude mice. Following the development of palpable tumors (about 200mm 3 ), mice w¾re randomized and given daily injections of either CS3D or CS3M (5mg/kg/day; 10 tumors/group). Experiments were done twice as biological replicates. Tumor volumes were recorded every other day while animal weights w¾re also monitored during the course of the treatment.

Figure 7C show's c-Myc is suppressed after CS3D treatment in vivo. At the end of the treatments, tumors were harvested, whole cell lysates were prepared and RNA extracted for target gene analysis by Western blotting and RT-qPCR. GAPDH protein was used as internal loading control for immunoblotting. CS3D treated tumors (C4, C5, and C6) from separate animals show 7 a decrease in c-Myc expression level relative to the CS3M-treaied group (C 1 , C2, and C3) in 201T. Immunoblotting analysis of H1975 residual tumors treated with CS3D (C8- C8) also showed reduction in c-Myc expression levels relative to CS3M (C1-C4). The expression of c-Myc w 7 as significantly reduced in response to CS3D, relative to CS3M, in both wild-type (201T, (P<0.042)) and mutant EGFR (H1975, (P<0.05)) derived tumors (*P<0.05). Data are shown as mean +/- SEM.

Figure 7D show's densitometry quantification of c-Myc protein expression levels in 201T and HI 975 from tumor-derived xenografts. The expression of c-Myc was significantly reduced in response to CS3D, relative to ( S3 VI. in both wild-type (20 FT, (PO.042)) and mutant EGFR (H1975, (P<0.05)) derived tumors . Data shown are means +/- SEM between two groups (C S3 Viand CS3D).

Figure 7E show's c-Myc mRNA expression levels in 20 IT and H1975 tumor lysates.

Data are presented as mean +/- SEM. *P<0.05 compared with the mutant control group (CS3M).

Figure 8A show's STAT3 decoy induces caspase-3 cleavage. Representative sections of HI 975 tumors stained for expression of cleaved caspase-3 Hl975-derived xenografts administered daily intravenous injections of either CS3D or CS3M were harvested at the end of treatment (day 14), and expression of cleaved caspase-3 was used as a marker for apoptotic ceils.

Figure 8B shows a bar graph of STATS decoy induced caspase-3 cleavage from sections of HI 975 tumors.

Figure 9A shows short-term CS3D-treatment degrades p-STAT3. Level of p-STAT3 is reduced after five days of CS3D treatment compared to CS3M. Tumors derived from H1975 xenografts were analyzed by immunohistochemical staining for p-STAT3 after 5 daily iv injections of either STATS decoy or mutant decoy.

Figure 9B show's bar graph quantitation of the frequency of low-, medium-, and high- grade p-STATS staining m multiple images; P < 0.001, decoy compared to mutant quantifying the degration of p-STAT3. Bright field images were captured at 40X magnfication.

Figure 10 shows localization of fluorecein-labelled CS3D by confocal imaging. HI 975 cells transfected with 0.25nnM FITC labeled CS3D were fixed after 24hr of transfection and imaged confocal analysis revealed the presence of FITC labeled CS3D (green) within the Dapi stained nucleus (blue) and cytoplasm with a greater proportion detected in the cytoplasm.

Images shown are 40x and 6Qx magnification.

Figure 11 show's long-term CS3D treatment alters tumor eellu!arity. H&E (hematoxylin and eosm) staining of 201T and H1975 harvested tumors following daily treatment with CS3D revealed a decrease in tumor cellularity and significant amount of debris as a result of apoptotic- mediated processes compared to CS3M that showed a more intact tissue achitecture. Images were captured at 20X magnification.

Figure 12A shows short-term CS3D-treatment decreases Ki-67 expression.

Immunohistochemical analysis using a Ki-67 monoclonal antibody revealed a decrease in the proportion of Ki-67 positive cells in CS3D-treated tumors after fi ve days, compared to CS3M.

Figure 12B shows a bar graph of frequency of low-, medium-, and high-grade Ki-67 staining in multiple images; P < 0.001, decoy compared to mutant.Images were captured at 40X magnification.

Figure ISA show's combination treatment results by 201T cell viability assessed at 48 hr by MTS assay. Results from triplicate wells in 96 well plates.

Figure I3B show's combination treatment results by HI 975 cell viability assessed at 48 hr by MTS assay. Results from triplicate wells in 96 well plates.

Figure 14A shows a chemopreventive effect of CS3D by examining histology' of preneoplasias and lung tumors, and bar graphs of counts of preneoplasias and tumors, in the lungs of mice that had previously received the carcinogen NNK. Lungs were examined 1 week after the 8 week treatment with CS3M or CS3D ended, showing a chemopreventive effect. Preneoplasias were reduced by CS3D (P < 0.001) after 1 week. Tumor burden per animal was also reduced.

Figure 14B shows histology of preneoplasias and lung tumors, and bar graphs of counts of preneoplasias and tumors in mice exposed to NNK, 8 w'eeks after the 8 week treatment ended, showing a chemopreventive effect. Tumors were reduced 8 weeks after treatment ended by CS3D, while the number of preneoplasias were increased, compared to CS3M, showing that preneoplasias were inhibited in progression to tumors.

Figure 15 A shows photographs of lungs taken from NNK-exposed mice treated with either inactive CS3M control or active CS3D, 20 weeks after the end of the 8-week dru treatment (6.5 months after NNK exposure), showing a chemopreventive effect with fewer visible tumors in CS3D treated animals.

Figure 15B shows a bar graph of counts of tumors from lungs examined 20 weeks after the end of treatment with CS3M or CS3D, showing a chemopreventive effect. Number of tumors is reduced 42% (P=0.019)

Figure 16 shows a bar graph of average tumor size after treatment with CS3M or CS3D, 20 weeks after the end of treatment, showing a chemopreventive effect, measured using the LAS V4.12 Leica Imaging program to quantify tumor volumes. Tumor size was reduced 50% by CS3D, showing a chemopreventive effect (P=0.011).

Figure 17 shows the proportion of KRAS gene G12D mutations (KRAS+) in tumors from mice treated with CS3M or CS3D, 20 weeks after the end of treatment, showing no difference m incidence of KRAS mutations. Tumor tissue was microdissected from individual slides (n=10 for each group) and Sanger sequencing was used to analyze codon 12 of exon 1 of the KRAS gene. The difference was not significant (P > 0.1)

Figure 18 show's expression of activated phospho-STAT3 protein (Panel A) and activated phospho-NF-Kappa B protein (Panel B) detected by immunohistochemistry in tumors after treatment with CS3M or CS3D, 8 weeks after the end of treatment. Pictures of the stained sections are shown on the left and enumeration of the number of microscopic fields scored low, medium, or high staining is on the right. There is an enrichment of microscopic fields scored low to moderate in CS3D-treated tumors (P < 0.05) for both phospho-STAT3 (panel A) and phospho-NFKB (panel B). This further supports ability of C53D to cause p-STAT3 degradation.

Fi gure 19 show's protein expression of the angiogenic marker VEGF in the lungs 8 weeks after the end of treatment period with either CS3M or CS3D, detected by

immunohistochemistry. Pictures of the stained sections are shown on the left and enumeration of the number of microscopic fields scored low, medium, or high staining is on the right. There is an enrichment of microscopic fields scored low or moderate m CS3D-treated tumors (P < 0.05), showing a reduction in angiogenic stimulation.

Figure 20 show's protein expression of the cytokine interleukin 6 (IL-6) in the lungs detected by immunohistochemistry in tumors after treatment with CS3M or CS3D, 8 weeks after the end of treatment. Pictures of the stained sections are shown on the left and

enumeration of the number of microscopic fields scored low, medium, or high staining is on the right. There is a decrease in the number of fields scored low with CS3D treatment (P< 0.05), indicating IL6 w¾s induced by CS3D.

Figure 21 show's protein expression of the enzyme cyclo-oxygenase 2 (COX2) in the lungs 8 w¾eks after the end of treatment with either CS3M or CS3D, detected by

immunohistochemistry. Pictures of the stained sections are shown on the left and enumeration of the number of microscopic fields scored low, medium, or high staining is on the right. There is a decrease of microscopic fields scored low in CS3D-treated tumors (P < 0.05), indicating COX2 protein was induced by CS3D.

Figure 22 shows proportion of different immune cell types detected by flow' cytometry to separate immune cells removed from lungs of mice treated with NNK for 4 weeks, followed by 1 week of CS3M or CS3D administration. Cells detected are Ml macrophages, M2

macrophages, and MDSCs. The percentage of M2 macrophages and MDSC cells is reduced, while the percentage of Ml macrophages is increased, in CS3D-treated mice, showing a shift towards more anti-tumorigenic immunity with CS3D

DETAILED DESCRIPTION

Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated in the accompanying structures and formulas. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinaiy skill in the art to which this invention belongs.

DEFINITIONS

The term“cancer chemoprevention” refers to the administration of a medication (natural, synthetic, or biological chemical agents) or a naturally occurrin substance such as a nutrient or biological molecule for the purpose of reversing, suppressing, or preventing (including lowering the risk of) carcinogenic progression to invasive cancer. The term“oligonucleotide’" refers to a short DMA or RNA molecule comprised of nucleotides. A, G, C, and T. Nucleotides are organic molecules that serve as the monomer rants for forming the nucleic acid polymers deoxyribonucleic acid (DNA) and ribonucleic acid (RNA), both of which are essential biomolecules in all life-forms. Nucleotides are the building blocks of nucleic acids; they are composed of three subunit molecules: a nitrogenous base, a five-carbon sugar (ribose or deoxyribose), and at least one phosphate group. Oligonucleotides readily bind, in a sequence-specific manner, to their respective complementary oligonucleotides, DMA, or RNA to form duplexes or hybrids of a higher order. Antisense oligonucleotides are single strands of DNA or RNA that are complementary to a chosen sequence. In the case of antisense RNA they prevent protein translation of certain messenger RNA strands by binding to them. Antisense DMA can be used to target a specific, complementary (coding or non-codmg) RNA.

“Carcinogenic” agents are cytotoxic agents that include tobacco smoke, benzene, vaping particulates, asbestos, or some combination thereof, and including an agent on a public list of such agents maintained by the American Cancer Society. The carcinogenic agent may be cisplatin, tamoxifen, other chemotherapeutic, or combination of multiple chemoiherapeutics. In still other embodiments, the carcinogenic agent is any type of radiation exposure, including radon, alone or in combination with other carcinogenic and/or chemotherapeutic agents.

The terms“treat”,“treating”, and“treatment” refer to therapeutic treatment, wherein the object is to slow down (lessen) or reverse an undesired physiological change, disease, condition or disorder, such as the development or spread of an infection, arthritis or cancer. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, including reduction in or reversal of preneoplastic changes, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those with the condition or disorder or those who are at risk for the disorder.

The phrase "therapeutically effective amount" or“effective amount” includes but is not limited to an amount of a compound of the that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (hi) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein, or (rv) prevents or reverses pre-cancerous change or changes (preneoplasia) that do not medically qualify as a malignant change. The term“alkylene” by itself or as part of another substituent means a divalent radical derived from an alkane, such as -CH 2 CH 2 CH 2 CH 2 CH2-.

The term“biomarker” means a measurable biological substance that can be extracted from bodily fluid or tissues that can detect the biological activity of a drug.

The term“cancer immunotherapy” means therapies that are developed to enhance the activity of cells in the innate and/or acquired immune system to attack and reject cancerous cells in the body.

STAT3 Blockers

The STAT3 Blocker can be any type of inhibitor or blocking agent of STAT3, whether targeting STAT3 mRNA, STAT3 monomer, phosphorylated STAT3, acetyl ated STAT3, activated STAT3, STAT3 homodimer, or STAT3-STAT1 heterodimer. It includes any agent that degrades the STATS protein, m either its native or activated state, especially through the ubiquitin proteasome system.

In some embodiments, the STATS Blocker/Degrader comprises an oligonucleotide that is termed a Decoy, or an analog thereof.

In some embodiments, the STAT3 Blocker/Degrader comprises an oligonucleotide having the sequence:

5 , -(N 6 )nCAN 1 TTCN 2 CN 3 TN 4 AN 5 TC-(N 7 ) m -3' (SEQ ID NO: 15)

wherein N 3 is G: N ! , N 2 , N 4 , and N 5 are A, T, G or C;

and one or more of the following conditions (i)-(v) are met:

(i) N ! is T; N 2 is C; N 4 is A; N 5 is A; and N 6 and N 7 are independently selected from A,

T, G or C, and n and m are independently selected from an integer from 0 to 50;

(ii) the first end of a first strand is joined to a first end of the second strand by a hexa- ethyleneglycol spacer;

(iii) the second end of the first strand is joined to a second end of the second strand by a hexa-ethyleneglycol spacer;

(iv) the oligonucleotide binds to STAT3 protein under physiologic conditions and interferes with STAT3 binding to its target sequence and may cause phospho-STAT3 to be degraded; and

(v) the oligonucleotide has a serum half-life of greater than about 4 hours.

In some embodiments, the Decoy is comprised of sense and antisense oligonucleotides forming a double-stranded duplex.

In some embodiments, the Decoy is a single stranded oligonucleotide with complete or partial self-complementarity. In some embodiments, the Decoy is an oligonucleotide having any of the following structures (or analog thereof) (SEQ ID NOS 1-4 and 16, respectively, in order of appearance):

In some embodiments, the Decoy comprises one or more sequences selected from the following:

where X is a hexaethyleneglycol, -(CH 2 CH 2 0) 6 spacer.

In some embodiments, the Decoy is comprised of one or more internucleotide phosphate analogs, selected from phosphorothioate, phosphoramidate, and alkylphosphonate.

In some embodiments, the STATS Decoy is CS3D, a self-complementary, cyclized oligonucleotide having the structure (SEQ ID NOS 4 and 16, respectively):

with two hexaethylene glycol spacers (US 9062121 ; US 8722640; Sen M, et al, Cancer Discov (2012) 8:694-705).

in some embodiments, the STATS Decoy is a cyclized oligonucleotide shown in Figure 2, an exemplary embodiment of a cyclic oligonucleotide STAT Decoy with the forward and reverse STATS DNA consensus sequence. CS 18 = hexaethyleneglycol - (CH 2 CH 2 0) 6 .

In some embodiments, the STATS Decoy structure comprises a carbon spacer and receptor ligand. In some embodiments, the receptor ligand helps to target delivery of the oligonucleotide to a certain receptor. In one embodiment the STATS Decoy structure comprising a carbon spacer and ligand receptor is of the form (or analog thereof) (SEQ ID NOS 17, 4 and 16, respectively):

wherein o is a spacer moiety independently selected from Ci-C l2 alkyl ene, and - (CH 2 CH 2 0)n- where n is 1 to 6, and -CH 2 CH 2 0P(03)-.

In some embodiments, the STATS Blocker is antisense RNA. In some embodiments, the STAT3 Blocker is AZD-9150 (ISIS 481464) or analog thereof (Hong, D. et al, Sci Trans! Med. (2015) Nov 18;7(3l 4):314ral 85) which has the following structure:

In some embodiments, the STATS Blocker is a small molecule inhibitor such as, napabucasin, WP1066, WP1 120, WP1732, or analog thereof.

Not to be limited to a particular mechanism-of-action, Figure i shows a DNA-based STATS decoy binding to pSTATS and blocking STATS dimers from binding DNA. The decoy may dampen these effects by reducing STATS-dependent activity of myeloid-deri ved suppressor cells (MDSCs) and pro-tumor macrophages that promote lung cancer, suggesting STATS blockade could enhance immunotherapies. This approach prevents STATS dimers from binding their DNA promoter elements, effectively blocking the ability of acti vated STATS to induce gene transcription. Complementary oligonucleotides that mimic the DNA sequence found m the promoter regions of STAT3~responsive genes were prepared and tested.

By focusing on inhibiting the active form of STATS rather than blocking all STATS molecules, it may be easier to achieve blockade of STATS signaling. The oligonucleotides serve as a competitive sink for active STATS dimers, reducing transcription of STATS target genes like c-Myc A first-generation linear decoy was very' effective at blocking cancer cell growth m vitro but could not be given systemically. CS3D, an improved, second-generation version of the decoy (Figure 2) has been produced that is thermally stable, by creating complementary oligonucleotides with two hexaethyleneglycol spacers (CS18) placed between one of the complementary sequences to allow for the production of a cyclic double-stranded DNA upon ligation with ligase.

The cyclic STAT3 decoy (CS3D) was shown to be active after systemic administration, and to have anti -tumor effects in human head and neck cancer xenografts after IV injection. Toxicology studies in mice showed no significant effects on body weight, organ histology, or clinical chemistry at repeated decoy doses up to iOOmg/kg. The novel mechanism of action combined with the stability of the circularized STAT3 decoy and its low toxicity are favorable features for clinical application. In all experiments with the STATS decoy, activity is compared to a mutant version with a T-A base pair replacing a G-C base pair at position 9, differing by the two nucleotides in the base pair. The mutant form (CS3M) is unable to bind STAT3 and is biologically inactive (Leong et al., Proc Natl Acad Sci USA (2003) 100:4138-4143).

The precyclized sequences of CS3D and CS3M are as follows:

where (CS18) is a hexaethyleneglycol, -(CH 2 CH 2 0)6 spacer.

A decoy approach was utilized by circularizing (through ligation) a double-stranded oligonucleotide containing a 15 base-pair sequence corresponding to the STAT3 response element (Leong PL, et al, Proc Natl Acad Sci USA (2003) 100:4138-43). The circular molecule was produced through the inclusion of two hexaethyleneglycol spacers that provide flexibility, and upon ligation becomes a thermally stable cyclic double-stranded oligonucleotide (Sen M, et al, Cancer Discov (2012) 8:694-705). The cyclic STATS decoy (CS3D) was compared to a mutant inactive control (CS3M) molecule that differs from the active compound by a single base-pair and lacks binding to the STATS protein. This allows an assessment of the specificity of CS3D to block STAT3's ability to regulate cellular functions. The STATS decoy was previously shown to decrease luciferase activity in cells expressing a iuciferase reporter gene under the control of the STATS consensus sequence, and to competitively bind to activated STATS protein (pSTAT3) in comparison to the mutated version, which showed no affinity for pSTAT3 protein.

The decoy approach allows STAT3 dimers to preferentially interact with the oligonucleotide, acting as a molecular sink to competiti vely inhibit binding of the dimer to the promoters of STAT3 target genes. This approach has demonstrated antitumor efficacy in head and neck squamous cell carcinoma model s (HNSCC) (Sen M, et al, Mol Med (2014) 20:46-56; Klein ID, et al, PLoS ONE (2014) 9(1): e81819. doi: 10.1371/joumal.pone.0081819), and the ligated circularized decoy exhibits greater potency when injected in vivo compared to linear versions.

Wild-type and mutant EGFR NSCLC cells exhibit similar sensitivity to the STAT3 decoy (CS3D)

CS3D (compared to CS3M) is active m NSCLC cell culture, reducing viability and colony formation, and increasing apoptosis (Njatcha et al, Proceedings of ike American

Association for Cancer Research (2017) Volume 58: page 1048, Abstract # 4101, April 2017. Available online March 31, 2017). Figure 3 A shows the effects of the cyclic STAT3 decoy CS3D relative to mutant control CS3M on in vitro cell viability. NSCLC cell lines (201 T and H1975) were transfected with CS3D or CS3M (cyclic STATS mutant decoy) at concentrations ranging from 0 nM to 600 nM. Using MTS assays, cell viabilities were assessed 72 hours later. Three independent experiments were performed, using 24-well plates and 4 wells/concentration. One 4-hr transfection of ligated CS3D using Lipofectamine (Invitrogen) caused significant reduction in viability by MTS assay compared to CS3M in 4 NSCLC cell lines tested (Table 1). The IC50 for CS3D was approximately 300 nM in each NSLCC cell line, regardless of EGFR or KRAS mutation status. CS3M had no appreciable effect. In comparison, the IC5Q for CS3D in normal lung fibroblasts and human bronchial epithelial cells was 30 nM, 100-fold higher. This agrees with toxicology' studies in mice showing no appreciable changes in blood chemistry', weight or organ structure with CS3D dosing. Figure 3B show's uptake of fluorescein-labeled CS3D (cyclic STAT3 decoy) by NSCLC in vitro. Confocal imaging 24 hours post-transfection shows intracellular localization of CS3M or CS3D in 201T and HI 975 cells.

Table 1 Cell viability assay

To assess the therapeutic potential of blocking STAT3 in NSCLC, a decoy approach was utilized by ligating a double-stranded 15-mer oligonucleotide that corresponds to the STAT3 response element within promoter regions of STAT3-target genes, to produce a cyclic STAT3 decoy (CS3D). The decoy was evaluated in vitro using NSCLC cells containing either wild-type (WT) EGF (201T) or mutant EGFR with an additional EGFRi resistance mutation (H1975), as shown in Figure 3 A. These cells are resistant to EGFR inhibitors and require an alternate therapeutic approach. Initial studies showed that transfection of 0.3 mM of CS3D caused a 50% inhibition in proliferation in 201 T (WT) and HI 975 (mutant) cells, relative to CS3M, as well as a 2-fold increase m the percent of apoptotic cells. Toxicity w¾s minimal in normal cells.

Treatment with the active decoy CS3D led to potent anti-tumor effects, while the mutant construct CS3M was largely inactive.

To determine the concentration of CS3D required to inhibit cell viability, increasing concentrations of CS3D was transfected, and after 72 hours, MTS assays were used to assess ceil viability. Data were normalized to ceils treated with lipofectamine alone. The inactive mutant control (CS3M) showed no cytotoxicity (Figure 3 A), and the percent of cy totoxicity induced by CS3D allowed calculation of the ICsos as described in Example 2. The ICso values for CS3D in both the WT (20 IT) and mutant (HI 975) EGFR cell lines wore approximately 0 3mM (Figure 3 A). Other cell lines (H3225 with EGFR activating point L858R mutation and A549 which is EGFR WT) were also assessed in the presence of CS3D and CS3M and showed similar sensitivities (ICsos 0.3 mM). Cell lines that are resistant to EGFR TKls (201T, H1975, and A549) showed the same sensitivity to STAT3 decoy as the EGFR TKI sensitive cell line (H3225). Normal lung fibroblasts and primary bronchial epithelial cells had ICsos that w¾re 100- fold higher (300 mM) than tumor cells.

Uptake of CS3D into NSCLC cell lines

To determine whether CS3D and CS3M were equally taken up by NSCLC cells, both molecules were tagged with a fluorescein dye (iFluorT) to assess transfection efficiency.

Twenty-four hours post-transfection with the iFluorT-labeled oligonucleotides at a

concentration of 25 nM. N SCLC cell lines showed intracellular location of the cyclic molecules (CS3D or CS3M), demonstrating that integrating hexaethyleneglycol linkers into the oligonucleotide, or circularizing the DNA, still allowed intracellular uptake. More importantly, altering the sequence of the CS3D by a singl e base-pair to generate the inactive STAT3 mutant (CS3M) did not affect its uptake, demonstrating that the cytotoxicity of CS3D was not due to preferential uptake compared to CS3M (Fig 3B). The efficiency of transfection was >90% for both CS3M and CS3D Further confoca! microscopy showed clear fluorescent signal in both the nucleus and the cytoplasm of lung cancer cells, with cytoplasmic signal predominating. Figure 10 shows localization of fluorecein-labelled CS3D by confocal imaging. H1975 cells transfected with 0.25nnM FITC labeled CS3D were fixed after 24hr of transfection and imaged confocal analysis revealed the presence of FITC labeled CS3D (green) within the Dapi stained nucleus (blue) and cytoplasm with a greater proportion detected m the cytoplasm. Images shown are 40x and 60x magnification. This suggests that CS3D can interact with its target STAT3 dimers in both compartments.

CS3D inhibits colony-forming ability of NSCLC and induces cell death

The effect of CS3D on the ability of NSCLC cells to grow in an anchorage-independent manner was examined. Cells (20 IT and H1975) were transfected with either CS3D or CS3M (300 nM), then seeded in soft agar. Compared with CS3M, CS3D produced a significant decrease in colony formation. The number of colonies formed was quantified by using a size cutoff (with greater than 35 pixels counted as a colony). A single transfection with CS3D significantly (P<0.051) disrupted anchorage-independent growth of the NSCLC cells by 70% (at 15 days in H1975 cells) and 50% (at 20 days m 201T cells (PO.053)) compared to CS3M. Figure 4A shows CS3D inhibits anchorage independent growth and promotes apoptosis.

NSCLC cell growth was assessed in colony forming assays in the presence of either 300 nM CS3D or CS3M. CS3D significantly blocks the ability of NSCLC to grow in an anchorage independent manner in soft-agar post-transfection. In a soft agar assay, one transfection reduced colony formation by 55% in 201T cells and by 65% in HI975 cells (P< 0.05, Figure 4A) One transfection also increased the number of apoptotic cells by 2-fold in all 4 cell lines and suppressed STAT3 target genes. Using one transfection of 300 nM CS3D or CS3M, followed by stimulation with 10 ng/rnl EOF, expression of Myc and Bcl-xL (STAT3 response genes) was measured by RT-PCR, 1 hr after treatment with EGF. Expression of c-Myc and Bcl-Xl niRNA in response to EGF was reduced 60% or more by CS3D (P < 0.05).

Flow cytometric analysis of Annexin V/propidium iodide-stained cells also demonstrated that transfection with CS3D significantly increased apoptosis in comparison to CS3M. CS3D treatment (100 nM) caused a 2-fold increase (P<0.05) in the number of cells undergoing apoptosis as compared to CS3M at 24 h. Figure 4B shows detection of annexin V and propidium iodide (Pl)-positive cells by flow' cytometry: HI 975 and 20 IT cells transfected with lOOnM of CS3M or CS3D for 24hr were stained with apoptosis marker (annexin V) and cell viability dye. Statistical significance was determined as *P<0.05, n=3.

CS3D suppresses the expression of c-Myc in response to EGF

c-Myc is a STAT3 target gene that is known to be activated by EGF treatment. To investigate the effects of STAT3 blockade on c-Myc expression, cells were first treated with 10 ng/ml EGF for 1.5 hr (deemed the optimal time to observe increased c-Myc RNA), 24 hr after transfection of the cyclic oligonucleotides. RT-qPCR w¾s used to assess differences in mRNA i expression in STAT3 decoy-treated versus mutant decoy-treated ceils (Figures 5A and 5B). A single transfection of CS3D caused a 50% inhibition in mRNA level of c-Myc in 201T cells (Figure 5 A) and a 25-30% inhibition in H1975 cells (Figure 5B). Immimohlotting analysis also showed a 31% reduction in c-Myc protein expression post-CS3D transfection compared to either CS3M or control (lipofectamine aione) in 201T cells. Figure 5 A shows down-regulation of c- Myc and bcl-xL mRNA expression by CS3D in 201 T cells. Cells were treated 24 hours post- transfection with EGF (10 ng/'ml) for 1.5 hr and mRNA was harvested from wild-type (A) and mutant EGFR T790M (B) NSCLC cell lines, and mRNA expression was assessed by RT-qPCR. Relative mRNA expression was normalized to GAPDH mRNA levels as an internal control. Statistical signifi cance was determined as *P<0.05. Figure 5B shows down-regulation of c-Myc and bcl-xL mRNA expression by CS3D in H1975 cells. Cells were treated 24 hours post transfection with EGF (10 ng/ml) for 1.5 hr and mRNA was harvested from mutant EGFR T790M (B) NSCLC cell lines. Similarly, in H1975 cells, a 40% reduction in c-Myc protein expression was found. Figure 6A show's down-regulation of c-Myc protein by CS3D. Cells were treated as in Figures 5A and 5B, and 3 hr after EGF treatment, lysates were collected from wild- type 201T (left) and mutant EGFR T797M H1975 (right) NSCLC cell lines. Protein expression was assessed by Western blot. Relative c-Myc expression was normalized to GAPDH protein levels as an internal control. H3225 cells and A549 cells also showed similar decrease in c-Myc expression. Examination of other STATS genes (for example Bcl-xl as shown in Figures 5A and 5B) also responded with decreased mRNA and/or protein expression across the four cell lines, but not as consistently as c-Myc. Examination of STAT1 target genes (IKBKE, IFT2, 1FT1, 1RF7) and NFKB target genes (IL-Ib and IL-8) that are predominantly regulated by these transcription factors showed no decrease in mRNA expression level by qRT-PCR when comparing CS3D to CS3M treated cells.

Additional immunoblotting analysis of cell extracts showed that after stimulation with IL6, a potent inducer of p-STAT3, there was a 35% reduction in both nuclear pSTAT3 and nuclear total STATS after transfection of CS3D compared to CS3M or control (lipofectamine alone) (Figure 6B). The levels of pSTAT3 were reduced by CS3D, suggesting that the selective binding of pSTAT3 to CS3D alters its stability and causes it to degrade. H 1975 were stimulated 24 hours post-transfection with either ( ' S3 VI. CS3D, or lipofectamine alone with IL-6 (50 ng/niL) for 1 hr to strongly activate STAT3. After cell lysis, nuclear and cytoplasmic extracts were obtained by subcellular fractionation using centrifugation, and Western blotting was used to assessed to protein expression. GAPDH was used as a marker of the cytoplasmic fraction and histone H3 was used as a marker of th e nuclear fraction. CS3D appears to have little effect on the cytoplasmic pool of total STAT3, but caused degradation of p-STAT3. In the nucleus, both the total STAT3 levels and the p-STAT3 levels were reduced showing that interaction with CS3D causes STAT3 to degrade. To test whether CS3D could increase ubiquitination of p- STAT3 protein, an immunoprecipitation was carried out for p~STAT3, followed by

immunoblotting for ubiquitin. After 24 hr, CS3D increased the ratio of p-STAT3 that was ubiquitinated relati ve to amount of STAT3 present (input), compared to CS3M (0.95 vs. 0.74). Figure 6C show's CS3D increases the proportion of pSTATS-uhiquitin bound complexes. 24 hours post-transfection, P-STAT3 was immimoprecipitated and the presence of ubiquitin was assessed by immunoblotting. Greater ratio of pSTAT3-ubiquitin complexes was detected with CS3D treatment. The increase in pSTAT3-unbiquitin complex correlated with decrease in total STAT3 present. Densitometric analysis revealed a greater ratio of pSTAT3 -ubiquitin complex to STAT3 in response to CS3D (0.95) as compared to CS3M (0.74). The input of total cell lysate also showed less STAT3 after CS3D treatment, confirming the results in Figure 6B. This shows that CS3D increases the degradation of p-STAT3 by the ubiquitin proteasome pathway.

Intravenous injection of the cyclic STAT3 decoy (CS3D) inhibits NSCLC tumor growth in vivo

Based on the in vitro effects of CS3D, antitumor effects were evaluated m mice harboring established NSCLC xenografts (10 tumors/group). When tumors reached 200 mm3, mice were given daily intravenous injections (tail vein) of CS3D or CS3M (5mg/kg/d), 5 days per week, and tumor growth was monitored for 14-20 days. CS3D caused a significant and robust tumor growth inhibition in 201T- and H1975-derived xenografts compared to treatment with CS3M. A 96.5% (P0.007) was observed in 201T (Figure 7 A) and 81.7% (P0.0001) reduction in H1975 (Figures 7A and 7B). Tumor growth at 14 days was inhibited over 85% by the active decoy in both KRAS WT cell lines (P < 0.0001) as reported (Njatcha et a!,

Proceedings of the American Association for Cancer Research (2017) Volume 58: page 1048, Abstract # 4101, April 2017. Available online March 31 , 2017).

Figures 7A and 7B show CS3D suppresses NSCLC xenograft tumor growth. Intravenous delivery of CS3D inhibits tumor growth in both wild-type (Figure 7 A, 201T) and mutant EGFR T790M (Figure 7B, H1975) xenografts. Approximately IxlO 6 cells 'ere inoculated

subcutaneously in the flanks of nude mice. Following the development of palpable tumors (about 200mm 3 ), mice were randomized and given daily injections of either CS3D or CS3M (5mg/kg/day; 10 tumors/group). Experiments were done twice as biological replicates. Tumor volumes were recorded every other day while animal weights were also monitored during the course of the treatment. The difference in tumor volume between the CS3D- and CS3M-treated groups w¾s significant for both 201T (P<0.007) and H1975 (PO.OOOl). Figures 7C and 7D show c-Myc is suppressed after CS3D treatment in vivo. At the end of the treatments, tumors were harvested, whole cell lysates were prepared and RNA extracted for target gene analysis by Western blotting and RT-qPCR. GAPDH protein was used as internal loading control for immunoblotting. CS3D treated tumors (C4, C5, and C6) from separate animals show a decrease in c-Myc expression level relative to the CS3M-treated group (Cl , C2, and C3) in 201T. Immunoblotting analysis of H1975 residual tumors treated with CS3D (C8- C8) also showed reduction in c-Myc expression levels relative to CS3M (C1-C4). The expression of c-Myc was significantly reduced in response to CS3D, relative to CS3M, in both wild-type (20 IT, (PO.042)) and mutant EGFR (HI 975, (P<0.05)) derived tumors (*P<0.05). Data are shown as mean +/- SEM.

Figure 7E show's CS3D suppresses c-Myc mRNA expression levels in 201T and H1975 xenografts (Figure 7D). Data are presented as mean +/- SEM. *P<0.05 compared with the mutant control group (CS3M).

Excised tumors showed grossly necrotic areas, as well as greatly increased caspase 3 staining. STATS target gene c-Myc protein w'as significantly reduced (P < 0.01) in lysates from CS3D-treated tumors compared to CS3M (Figure 8A). Staining (H&E, hematoxylin and eosin) of sectioned tumors harvested after treatment showed that the CS3D treated tumors were composed of large areas of debris and infiltration with stroma and lymphocytes, while the CS3M tumors had a high tumor cellularity. Figure 11 show's long-term CS3D treatment alters tumor cellularity. H&E (hematoxylin and eosin) staining of 20 I T and H1975 harvested tumors following daily treatment with CS3D revealed a decrease m tumor cellularity' and significant amount of debris as a result of apoptotic-mediated processes compared to CS3M that showed a more intact tissue achitecture. Images were captured at 20X magnification. The animals showed no significant loss of body weight or decreased activity', and histological examination of the lungs, liver, and spleen showed no signs of toxicity with CS3D.

STAT3 inhibition In Vivo by CS3D down-regulates c-Myc and promotes cell death

To determine how CS3D blocks NSCLC growth, Western blot, cjRT-PCR, and immunohistochemical analysis were performed to determine expression of c-Myc in residual tumors. These analyses identified a substantial downregulation of c-Myc protein (Figure 7C and 7D) and mRNA levels (Figure 7E) in individual CS3D-treated tumors relative to CS3M-treated tumors in both 201T and HI 975 xenografts. Some variability in protein levels among individual xenografts was observed (Figure 7C and 7D); the densitometry units for c-Myc protein after normalizing for GAPDH levels ranged from 0.11 -0.16 units for CS3D-treated xenografts and 0.25-0.46 units for CS3M-treated tumors m H1975. Similarly, in 20lT-derived tumors these measurements ranged from 0.015 to 0.51 units for CS3D and 0.28 to 0.97 units for CS3M. Even with this variability, the effect across the groups in both cell lines was significant (P<0.Q5 for H1975 and PO.042 for 201T xenografts), with a mean decrease of 65% and 70%, respectively.

For mRNA levels m tumor lysates, CS3D produced a 60-70% reduction of c-Myc gene expression relative to GAPDH compared to CS3M (Figure 7E). Figure 8A shows

immunohistochemical (IHC) analysis of tumors harvested after the last day of treatment also revealed a substantial increase in the levels of cleaved caspase-3 (observed predominantly in tumor cells) in response to CS3D. Sections of the H1975 xenografts in response to CS3M showed 10% of fields scoring for high level of cleaved caspase-3 whereas in CS3D-treated xenografts there was an increase to 83% of fields in the high scoring tumor sections (P<0.05). Figure 8B shows a bar graph of STATS decoy induced caspase-3 cleavage from sections of Hl 975 tumors. Some cleaved caspase 3 staining was localized in nuclei, where caspase-3 is known to be active. Some caspase 3 staining was also evident in fibroblasts.

Short-term CS3D Treatment Show's Anti-Tumor Effects including Degradation of Nuclear p- STAT3 in Vivo

Because tumors treated for several weeks with the STAT3 decoy were largely composed of debris (as illustrated in Figure 11), xenografts were examined after only 5 daily tail vein injections to determine what cellular changes were induced prior to collapse of the intracellular tumor structure. Figure 9A shows short-term CS3D-treatment suppresses p-STAT3 protein levels in the xenografts. Tumors derived from HI 975 xenografts were analyzed by

immunohistochemical staining for p-STAT3 after 5 daily iv injections of either STAT3 decoy or mutant decoy. Representative sections of H1975 tumors show a decrease in p-STAT3 protein levels consistant with a degradation of STAT3 in vivo in the xenografts, as was observed in vitro in cells in Figures 6B and 6C. This effect is quantified in Figure 9B.

Figure 10 show's eonfoca! imaging of iF!uor-lab!ed C53D showing the STAT3 decoy/degrader is localized in the nucleus and found in the cytoplasm within structures that appear to be endosomes.

Figure 1 1 shows histological changes (H&E staining) w¾re consistent with

disaggregation of malignant cells in CS3D-treated tumors. Further analysis by

immunohistochemical (IHC) staining revealed a significant decrease in the proliferative capacity ofNSCLC, shown by Ki-67 staining in response to CS3D-ireatment (*P<0.001) compared to CS3M-treated tumors after 5 daily injections. Figure 12A shows short-term CS3D~treatment decreases Ki-67 expression. Immunohistochemical analysis using a Ki-67 monoclonal antibody revealed a decrease in the proportion of Ki-67 positive cells in CS3D-treated tumors after five days, compared to CS3M. Figure 12B shows a bar graph of frequency of low-, medium-, and high-grade staining in multiple images; P < 0.001, decoy compared to mutant.Images were captured at OX magnification. A shift from 41% of cells was observed and scored as high-grade staining with CS3M treatment to 13% with high-grade staining m the presence of CS3D, with corresponding increases in moderate- and low-grade staining in the STAT3 decoy treated group (Figure 12B). Expression levels of p-STAT3 protein (which was mainly observed localized to the nucleus) were likewise significantly suppressed by the STAT3 decoy (*P<0 0001) after 5 days of treatment in vivo compared to mutant decoy. Figure 9B shows bar graph quantitation of the frequency of low-, medium-, and high-grade staining in multiple images; P < 0.001, decoy compared to mutant. Bright field images were captured at 40X magnfi cation. Frequency of high- grade staining for p-STAT3 differed between CS3D and CS3M treatment groups (23% in CS3D-treated and 51% in CS3M treated, with corresponding increase in low-grade staining in presence of the active decoy (Figure 9B). Significant degradation of p-STAT3 protein was detected by IHC in tumors was observed after treatment with CS3D. Percent of high staining fields was reduced from 48% to 22% while percent of fields with low staining increased, P < 0.001 (Figure 9B).

' The p-STAT3 result suggests that multiple doses of the decoy promoted the degradation in p-STAT3 levels in vivo. At this early time point, no induction of apoptosis wus observed (not shown), suggesting that effects on STAT3 and proliferation occur prior to an increase m cell death. No change in NF-kB nuclear protein was observed with the active decoy after five days of treatment, suggesting that the decoy does not induce NK-KB movement to the nucleus.

' The strong activity of CS3D in vivo on reducing p-STAT3 and c- Myc levels, and reducing tumor growth, may reflect greater effect upon repeated in vivo dosing, compared to the single exposure by transfection used in vitro. No differences in weight, food intake or motor activity with CS3D, and no histologic changes in spleen, liver, or lungs were observed.

The combination of CS3D with EGFR TKis showed enhanced effects in both EGFR WT and EGFR mutant/erlotmib resistant NSCLC (Figures 13A and 13B). Figure 13A shows combination treatment results by 20 IT cell viability assessed at 48 hr by MTS assay. Results from triplicate wells in 96 well plates. Figure 13B shows combination treatment results by H1975 cell viability' assessed at 48 hr by MTS assay. Results from triplicate wells in 96 well plates. The effect of the afatinib (GILOTRIF®, Boehringer Ingelheim) 5mM

(micromolar)/CS3D 300nM combination gave the best inhibition compared to other single or double treatments in ceil culture. Afatinib (GILOTRIF®), Boehringer Ingelheim) is an irreversible inhibitor of EGFR/HER2/HER4 with superior clinical activity to erlotinib in NSCLC. This effect was synergistic (P< 0.05) when multiple concentrations were tested, yielding a combination index of 0.4.

CS3D is a novel therapeutic that targets STAT3, a cyclic, double-stranded decoy oligonucleotide with the surprising and unexpected properties of a long half-life, high potency, and limited toxicity and ability to induce degradation of STAT3. The CS3D STATS oligonucleotide decoy specifically and competitively binds activated STATS protein via its similarity to the STATS response element located in the promoter region of the c-fos gene, and to reduce expression of STATS target genes in head and neck cancer models. Head and neck cancer cells also responded to the CS3D STATS decoy in a xenograft model.

' The CS3D STATS decoy produces a robust antitumor effect in NSCLC models that are known to be resistant to FDA-approved EGFRi therapies. In cell culture, CS3D decreased the viability' of NSCLC cells and induced apoptosis, and sensitivity was independent ofEGFR mutation status. These effects were specific compared to a mutant version of the mutant CS3M double-stranded decoy that is unable to recognize STATS protein and were demonstrable after a single transfection. Further in vivo studies illustrated that daily IV injection of the active decoy- had strong anti-tumor effects in mouse xenograft models of NSCLC, accompanied by inhibition of c-Myc protein expression and destruction of the cellul ar integrity of tumor cells within the xenografts. When tumors were analyzed after only 5 daily treatments, nuclear p-STAT3 protein was greatly reduced consistent with degradation by the proteasome and the proliferative state of tumor cells was also greatly decreased. Since the phosphorylation step that activates STAT3 is not affected by CS3L) STATS decoy, this result suggests one effect of the decoy is to reduce accumulation of p-STAT3 in the nucleus. This finding was consistent with the observed decrease in the nuclear pool of STATS and increase in ubiquitination of p-STAT3 protein following CS3D treatment in vitro. The cytoplasmic pool of p-STAT3 was also reduced by CS3D treatment in vitro, which could result from increased degradation of STAT3 dimers when bound to the active decoy. A CS3D-pSTAT3 dimer complex may trigger a ubiquitination process that reduces the amount of active STATS available for signaling in the nucleus.

STATS is primarily activated by modification through phosphorylation at tyrosine and serine residues (Y705 and S727) that effectively initiates dimerization of STATS monomers. However, small molecule inhibitors of STATS that inhibit phosphorylation, might be ineffective in blocking the functional activity of ST AT3 dimers that do not require phosphorylation to initiate dimerization. This strategy' also requires a very efficient inhi bition of the

phosphorylation step, which might limit efficacy . The use of CS3D as a STATS decoy inhibitor provides an opportunity to interrupt the functional activity of STAT3 dimers, without requiring the interruption of phosphorylation. Potentially this strategy' could be more effective, because it focuses on the active dimeric moieties. CS3D specifically downregulates the expression of the STATS target gene c-Myc in NSCLC cell lines m culture and in residual tumors after systemic administration. The transcriptional activity of STAT3 overlaps with other transcription factors such as STAT1 and NF-kB, but genes controlled predominantly by STAT1 or NF-kB such as IRF7 and TL-8 were unaffected by CS3D treatment. Although c-Myc is a direct STAT3 target gene, it is possible that indirect effects on c-Myc mRNA or protein expression could also be occurring in response to the STAT3 decoy.

The cyclic double-stranded oligonucleotide showed enhanced stability compared to linear versions (Sen et al, Cancer Discovery (2012) 2:694-705) and was efficacious after iv injection in a head and neck cancer model. In addition to relative lack of toxicity seen with the STATS decoy against normal lung cells and in mice used for xenograft studies, the decoy effects were also specific since the mutant inactive control oligonucleotide (which differs by a single base-pair mutation) was unable to induce anti-tumor effects. The cellular uptake of fluorescently labeled CS3D and CS3M also did not differ, supporting the specificity of the decoy that is designed to bind with selectivity to dimeric STATS protein. Imaging analysis also showed that the localization of CS3D mostly stained positively in the cytoplasm, although some was present in the nucleus, suggesting the main effect is to bind to pSTAT3 dimers in the cytoplasm. The uptake and anti-tumor effects of CS3D w¾re comparable in both EGFR wild-type and mutant NSCLC. It has been reported that the constitutively active mutant form of STATS, STAT3-C, which is dimerized by cysteine-cysteine residues, mediates epithelial cell transformation by promoting anchorage-independent growth (Bromberg IF, et al, Cell (1999) 99:238-239), suggesting that this phenotype is especially dependent on STATS signaling. After a single transfection, CS3D was very effective in suppressing anchorage-independent growth. The inhibition of c-Myc expression in cultured cells and in xenografts also was a strong indicator of CS3D activity, suggesting that the ability to suppress this STATS target gene may be critical for its anti-tumor mechanism in NSCLC The c-Myc protein binds to promoter regions of genes encoding lung cancer stem cell factors such as SOX2 and nanog, and cooperates with STATS in regulating these genes (Kidder BL, et al FLOS ONE (2008) 3(12): e3932.

doi: l 0 1371 /journal. pone.0003932), further suggesting that c-Myc down-regulation caused by reduction in STATS action may be important in the anti-tumor action of CS3D in NSCLC, especially in reducing stem cell self-renewal.

The cyclic decoy oligonucleotide-based approach exhibited strong therapeutic activity in NSCLC, an effect that was also observed in HNSCC. The non-cyclic version of the STATS decoy injected intratumorally into HNSCC patients in a Phase 0 trial produced remarkable effects by altering the expression of STAT3 target genes critical for the progression of HNSCC. The results support the ability of a systemically administered, double-stranded, cyclic oligonucleotide decoy to attenuate STAT3 signaling in a lung cancer model, which ultimately leads to tumor growth arrest and cell death. No increase in nuclear NF-kB protein in lung tumor xenografts was observed after decoy treatment, suggesting that the STATS decoy does not increase NF-kB nuclear translocation, as has been reported in a STATS knockout KRAS lung cancer model (Grabner B, et al, Nature Communications (2015) 6:6285). The observed effects in NSCLC support the idea that a circularized double-stranded oligonucleotide targeting STATS has promise as a therapeutic agent and may be effective in lung tumors that lack sensitivity to EGFR TKIs. The cyclic STATS decoy lacks toxicity 7 in immunocompetent mice, and no toxic effects were observed in the immunosuppressed animals used here.

METHODS OF CHEMOPREVENTION

Methods of the invention include the chemoprevention of cancer by administering to a patient in need or at risk a therapeutically-effective dose of a STATS Blocker/Degrader. The STATS Blocker/Degrader modulates one or more targets selected from STATS mRNA, STATS monomer, phosphorylated STATS, acetylated STATS, activated STAT3, STATS homodimer, and STAT3-ST ATI heterodimer. Methods of the invention include the chemoprevention of non small cell lung cancer by administering a therapeutically-effective dose of a STATS Blocker is administered to the patient.

A patient to be treated with a STATS Blocker/Degrader according to the methods of the invention may be at elevated risk of developing cancer, due to one or more factors selected from a genetic condition, immunocompromised status, comorbidities, prior treatment for cancer, incidence of cancer, prior treatment with or exposure to one or more chemotherapeutic agents, radiation therapy, exposure to radiation or radon, prior exposure to or treatment with one or more carcinogenic or cytotoxic agents, and social or family history correlated with the incidence of cancer. The factors include, but are not limited to, exposure to asbestos and smoking.

In one embodiment, the S ' TAT ' 3 Blocker/Degrader is given for the purposes of chemoprevention to patients who do not meet clinical guidelines for diagnosis of cancer. In another embodiment, the STATS Blocker is given for the purposes of chemoprevention to patients who have evidence of preneoplastic or dysplastic cells, but do not meet the guidelines for clinical diagnosis of cancer. In another embodiment, the STATS Blocker is given to patients currently m remission from a prior diagnosis of cancer. In another embodiment, the STATS Blocker is given to patients who had a complete resection of a cancer and who have no current evidence of disease. Any method of delivery of the STAT3 Blocker/Degrader to patients can be used including, without limitation, inhalation (including, without limitation, metered dose inhalation, dry powder inhalation, inhalation of aqueous suspension, inhalation of lipophilic suspension, inhalation of emulsion, inhalation of organic-based suspension), IV, topical, locally delivered during invasive procedure, trans dermal, subcutaneous injection, intramuscular injection, sublingual, suppository, eye drops, ear drops, PO, injection, infusion, continuous infusion, or depo injection. As used herein, a patient may be a human or animal of any kind, including, without limitation, a mammal, non-human primate, canine, or feline.

CS3D is active in chemopre vention of l ung cancer without causing toxicity

In a pilot experiment, 6 FVB/N mice were given 24 mg NNK (3 mg by ip injection, twice a wk x 4 wks), followed by 6 wks rest, then IV treatment of 3 mice with either 5 mg/kg CS3D or CS3M 5 days a wk for 18 wks. A considerable reduction in tumor burden was found with CS3D, and CS3D showed no discernable toxic effects, while mice receiving CS3M lost weight and had breathing difficulties. With CS3M, lungs were filled with multiple invasive adenocarcinomas, while m mice receiving CS3D, tumor burden was reduced over 80%. H&E (hematoxylin and eosin) sections were taken of murine lungs exposed to NNK followed by treatment with CS3M or CS3D for 18 wks. Invasive carcinomas were prominent with CS3M and lacking with CS3D.

To confirm these findings, in a second experiment, in which 24 mg NNK was given (3 mg by ip injection, twice a wk x 4 wks), this was followed by one week rest and then immediate treatment with 5 mg/kg CS3D or CS3M, 2 daily injections per week, for 8 weeks. Then 1 week, 8 weeks, or 20 weeks after the end of the treatment course (13 weeks, 21, and 33 weeks after beginning the NNK exposure), necropsy was performed. Mice treated with CS3D were compared to CS3M-treated control group and no body weight changes were observed or organ toxicity (by histological analysis of the liver and spleen) was detectable. Lung tumors were evaluated by inspecting formalin-fixed lungs visually and by histology after H & E staining. Histopathological analysis of the lungs from CS3D treated mice revealed approximately 25% fewer number of preneoplasias on average per animal (detected as thicked epithelial cell layer in the airways) as compared to CS3M at week 1 post-CS3D administration (Figure 14A). Tumor burden (detected by neoplastic growths [adenomas] observed in lung sections) w¾s also assessed at the week 1 timepoint after treatment. Consistent with reduction in preneoplasia, we observed a decrease in the average number of tumors (a 60% reduction m tumors in the CS3D-treated animal) (Figure 14A). To determine if the effects of CS3D is sustainable in a long-term experimental model, sections from lung tissue harvested at the 8 weeks post-treatment were examined and a 52% lower number of adenomas from the CS3D group compared to the CS3M control group was found (Fig MB). But in contrast, the CS3D-treated group showed a 43% higher count of preneoplasia compared to CS3M 8 w eeks after treatment ended, showing an inhibition of tumor transition from prenoplasi a to adenoma by CS3D.

With evidence that CS3D can delay both preneoplasia formation and prenop!asia progression to adenomas after NNK exposure, we assessed tumor burden formation at a later timepoint (20 weeks post-eight weeks of CS3D administration). The average number of tumors per animal was significantly reduced by approximately 40% in CS3D-treated animals as compared to CS3M control group. Figure 15A shows photographs of the wiiole lungs with the tumors visible on the lung surface. Arrow's point to visible tumors. Figure 15B shows enumeration of tumors from lungs of 10 mice per group. This difference was significant at P =0.019. These data show that targeting preneoplasia and the transition towards adenomas by STAT3 inhibition with CS3D suppresses tumor formation in an NNK-induced lung cancer model.

After illustrating that mice treated with CS3D are more resistant to NNK-induced lung preneoplasia and tumor burden formation, tumor size was also determined, by measurements using the LAS V4.12 Leica imaging program measuring individual tumors in each treatment group. CS3D causes 49.8% lower tumor size at 20 weeks post-treatment (Figure 16), further evidence of the chemoprevention effect.

There was no toxicity observed in mice who received CS3D treatment, based on behavior and weight of the mice and examination of the spleens and livers, suggesting it will be suitable for chemoprevention. Addi tional d etail s of the method are found in Example 10.

CS3D Effects on Tumor Burden are Independent of K-RAS Mutations

The potential tumor suppressive role of knocking out STAT3 in a K-RAS mutant lung cancer model was recently suggested, and tins might mean that a select group of patients who develop K-RAS mutations in the lungs could be less likely to benefit from STAT3~targeted therapy [Grabner et al. Nature Comm. 6:6285,2015] This published evidence was generated in a STAT3 knockout mouse model in which no STAT3 protein was expressed. Pharmacological targeting of STAT3 with CS3D w'as not shown to promote the survival of K-RAS driven tumors in a xenograft model, however (Njatcha et al, Mol. Cancer Therapeutics 17: 1917,2018). To evaluate the percent of tumors positive for K-RAS mutations in the NNK experiment, cells from individual tumors w¾re isolated by laser capture microdissection. Exon 1 of the K-RAS gene was sequenced by Sanger sequencing to determine the presence of the G12D missense genetic mutation which is the K-RAS mutation induced in this strain (FVB/N mice) by NNK. This is the only KRAS mutation known to be produced by NNK in mice. After Sanger sequencing analysis, there was no significant difference in the proportion of tumors positive for K-RAS mutation between CS3D and CS3M control group at tumor that developed 20 weeks post treatment (Figure 17), P > 0 1. These sequencing results demonstrate that pharmacological inhibition of STAT3 with C3SD does not promote the growth of K-RAS mutant lung cancer and should not be an impediment to chemoprevention. This further suggests that smokers (w io are likely to have acquired KRAS mutations in their lungs) would be eligible to receive CS3D

chemoprevention.

STAT3-induced Degradation by CS3D During Chemoprevention Reduces NF-kB Activation The selective survival of K-RAS driven tumors in a STATS knockout system as described by Grabner and colleagues (Nature Comm. 6:6285;2015) identified an NF-kB escape signaling mechanism. NF-kB promotes the survival of mutant K-RAS clones by promoting an immunosuppressive phenotype that involves blood vessel formation and macrophage infiltration. With knowledge of this potential drug-resistant mechanism, the effects of CS3D on both STAT3 activation and NF-kB activation were assessed. CS3D caused long-term reduction in activated phosho-STAT3 protein at 8 weeks post-treatment in the NNK model (Figure 18 A, P < 0.05) consistent with the degradation of STATS shown in the previous treatment models. The CS3D-mediated effect is also accompanied by a decrease in phospho-NF-kB activation (Figure 18B, P < 0.05), indicating that CS3D can effectively degrade STAT3 while simultaneously impairing NF-kB ( rather than activating) signaling. This finding reinforces the conclusion that even if KRAS mutations are present, the NF-kB signaling system will not be acti vated by CS3D, producing no effects that would impede chemoprevention in smokers. The long-term loss of pSTAT3 reinforces the claim that CS3D causes STAT3 protein degradation.

CS3D Disrupts the Pro-tumorigenic Phenotype Associated with Lung Cancer Development

STATS as a transcription factor has been widely implicated in several oncogenic events that defines the hallmarks of cancer development. Angiogenesis, cell cycle progression, and chronic inflammation are all STAT3 dependent processes that respectively regulate pro-survival factors such as VEGF, MYC and 1L-6. Biopsies from smokers at high risk of lung cancer shows that preneoplastic changes in the lungs consist of capillary blood vessels and angiogenic protrusions which helps establish a pattern of microvascularization [Jamicki et al Cell Div.5: I4, 2010] Contrary to smokers, biopsies from nonsmokers lack those vascularized lesions, suggesting that VEGF-dependent microvascularization at an early stage of lung carcinogenesis is a clinical feature that can be targeted to prevent tumor formation. VEGF activation in the endothelial compartment is known to be partly regulated by STATS [Chen et al. Cancer Biol. & Therapy 7:1994,2008] Enhanced vascularization in preneoplastic lesions suggests targeting STATS might disrupt the NNK-induced pro-tumorigenic angiogenve phenotype.

Immunohistochemical analysis of airways containing preneoplasias at 8 weeks post-treatment caused a decrease in VEGF staining in the airways (Figure 19, P < 0.05) CS3D also suppresses the endothelial marker CD31 expression (data not shown) suggesting that CS3D impairs the degree of angiogenesis at the premalignant stage. This observation further supports the use of CS3D for chemoprevention.

in addition to vascularization, chronic inflammation is now recognized to be important in the pathogenesis of lung cancer. Patients with lung cancer have elevated levels of 1L-6 which mediates inflammatory signals via canonical activation of STATS. Because IL-6 is a major activator of STATS, inhibition of STAT3 activity by CS3D could deprive cells of downstream IL6 signaling. IL-6 expression was evaluated in response to CS3D and results show' that IL-6 levels the airways are upregulated 8 weeks after completion of therapy with CS3D compared to CS3M, indicating cells may upregulate IL6 production as a rescue mechanism in an attempt to activate STATS (Figure 20, P < 0.05). This rebounding effect can be used as a biomarker of CS3D activity in patients, and IL6 measurement in blood, blood products, or other tissues can be used as a biomarker to monitor increased IL6 production, to show whether CS3D is showing biologic activity in patients.

In addition to IL-6 upregulation, STATS inhibition by CS3D also mediated a rebounding increase in COX-2 expression (P .OOOl) (Figure 21). Increased COX-2 protein or pathway activity (such as measuring PGE2, the product of COX2 enzyme in blood, blood products, or tissues) can also be a biomarker for biological activity of CS3D Because COX2 upregulation can contribute to lung tumorigenesis, combining CS3D with COX inhibitors such as aspirin or ibuprofen might enhance the chemopreventive effect. Both the IL6 and COX2 or COX2 pathway members such as PGE2 can be used as biomarkers for any type of STATS

Blocker/Degrader.

Pharmacological Inhibition of STATS by CS3D Shifts the Immunosuppressive Phenotype towards an Antitumor Immune Response

STATS also plays a central role in immune evasion STATS specifically orchestrates immunosuppression by promoting M2 macrophage and myeloid-derived suppressor cells (MDSCs) activity while suppressing Ml macrophage function [Su et al. Int. J. Mol. Science 19: pll:El 803, 2018] Thus STATS inhibition by CS3D should favor an antitumor immune response by enhancing Ml macrophage activity. Flow cytometry analysis of cells isolated from the lungs of mice that had first received NNK treatment for 4 weeks followed by 1 week of drug administration revealed that CS3D treatment in mice promotes an antitumor response by increasing the proportion of Ml macrophages present in the lung TME and also decreasing M2 macrophages(5.53% vs. 18.3%) and MDSCs (14.2% vs. 4.56%) (Figure 22), compared to control CS3M treatment. This indicates that CS3D may reduce immunosuppression as part of the chemopreventive mechanism, and treatment with CS3D in combination with cancer immunotherapy that enhances the anti-cancer activity of any cells of the innate or acquired immune system could improve both its chemopreventive or therapeutic effect.

PHARMACEUTICAL FORMULATIONS AND DOSAGE

In one embodiment, the STAT3 Blocker/Degrader is given to a patient multiple times a day for ehemoprevention. In another embodiment, the STATS Blocker is given to a patient 1 to 7 times per week for ehemoprevention. In one embodiment, the course of treatment with the STATS Blocker for ehemoprevention is between 1 day and 52 weeks. In another embodiment, the course of treatment with the STATS Blocker for ehemoprevention is 1 to 5 years. In another embodiment, the course of treatment with the STATS Blocker for ehemoprevention is greater than 5 years. In another embodiment, the course of treatment with the STAT3 Blocker for ehemoprevention is for the remaining duration of the patient's natural life. Patients may have multiple courses of treatment. In some embodiments, the patient is given 1 to 100 courses of treatment. In other embodiments the patient is given 101 to 1000 courses of treatment. In still other embodiments, the patient is given more tha 1000 courses of treatment.

Individual dosing typically ranges from O. lmg/kg to lOOOmg/kg for the STAT3 Blocker. In one embodiment, the individual dose ranges from 0.5-100 mg/kg. In a preferred embodiment, the individual dose ranges from 1 -6rng/kg for the STATS Blocker. Dosing may be adjusted for age and cancer risk profile for a given patient.

The STATS Blocker compositions of the invention can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e , orally or parenterally, by intravenous, inhalation, intramuscular, topical or subcutaneous routes.

Thus, the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an edible carri er. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of digestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.114 of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. Tire amount of acti ve compound in such therapeutically useful compositions is such that an effective dosage level will be obtained. The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, com starch or gelatin;

excipients such as dieaicium phosphate; a disintegrating agent such as com starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. The tablet may be coated with a glidant to facilitate passage through the gastrointestinal tract. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propyl parabens as preservatives, a dye and flavoring such as cherry' or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non- toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.

The active compound may also be administered intravenous or intraperitoneal by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of

microorganisms.

The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity' can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many eases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization in the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze dryring techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile- filtered solutions.

For topical administration, the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.

Useful solid earners include finely divided solids such as talc, clay, mierocrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the STATS Blocker compositions can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.

Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.

Examples of useful dermatological compositions which can be used to deliver the compositions to the skin are known to the art (US 4608392; US 4992478; US 4559157; US 4820508).

Useful dosages of the compositions can be determined by comparing the in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see US 4938949. The amount of the compound, or an acti ve salt or derivati ve thereof, required for use m chemoprevention will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.

The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete l oosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.

STATS Blocker/Degrader compositions of the invention can also be administered in combination with other therapeutic agents, for example, other chemotherapeutic agents, including but not limited to selected from erlotinib, afatmih, and osimertinib; and/or other chemoprevention agents such as aspirin, ibuprofen, anti -oxidants, Iloprost, or drugs of the g!itazone class such as piog!itazone; and/or in combination with agents that activate anti-tumor immunity such as immune checkpoint blockers like pembrolizumab or nivolumab.

EXAMPLES

Antibodies and Reagents; Antibodies for STATS, phospho-STATS, p-NFKB, cyclin Dl, cleaved caspase-3 (CCS), c-Myc, Ki67, GAPDH, Ubiquitin and horseradish peroxidase- conjugated secondary antibodies were purchased from Cell Signaling Technology' (Danvers, MA). The CellTiter 96® AQ ue ous One Solution (MTS) Reagent containing a tetrazolium compound [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-( 4-sulfophenyl)-2H- tetrazolium was obtained from Promega (Madison, WI).

Ceil Culture; The wild-type EGFR (201T) ceil line was deri ved from a patient with lung adenocarcinoma as described previously (Siegfried JM, et al, Pulm Pharmacol Ther (1999) 12:291 - 302). H1975 cells with the EGFR intrinsic point mutation L858R and the acquired EGFRi resistance mutation T79GM w-ere purchased from American Type Culture Collection (Manassas, VA). 201T and H1975 w'ere cultured in BME and RPMI respectively containing 10% heat-inactivated fetal bovine serum and lx penicillin/streptomycin (Thermo Fisher, Waltham, MA) and lx GlutaMax (Life Technologies, Carlsbad, CA) maintained at 37°C with 5% C02. Other cell lines w¾re purchased from ATCC (A549 and H3255) cultured in BME and RPMI, respectively. Human bronchial epithelial cells were purchased from ATCC, and primary normal lung fibroblasts were derived from human lung tissue (Stabile LP, et al, Cancer Res (2002) 62:2141-50). Cell lines were authenticated by short tandem repeat DNA profiling.

Example 1 Cyclic STATS Decoy (CS3D) Transfections

D The single-stranded STAT3 decoy and inactive mutant version were generated as previously described (Klein JD, et al, PLoS One PLoS ONE (2014) 9(1): e81819.

doi: l0.137l/joumai.pone.008l819); Siegfried JM, et al, Pulm Pharmacol Ther (1999) 12:291- 302). The inactive single-stranded mutant version differs by a base-pair at position 9 (G to T) as compared to the STAT3 decoy. Both oligonucleotides were synthesized by Integrated DNA Technologies (IDT) by phosphorarnidite chemistry on solid-phase. The single-stranded oligonucleotide (sense and antisense strands) was further modified with two hexaethyleneglycol linkers attached to both strands, allowing the structure to circularize upon enzymatic ligation of the 3’ and 5’ ends of the annealed oligonucleotide. Both molecules were purified by HPLC according to manufacturer’s modifications and reports (IDT). Using T4 DNA ligase (New England Biolabs, Ipswich, MA), ligations were performed overnight to produce the cyclic STAT3 decoy (CS3D) and the inactive circularized mutant control version (CS3M). Ligation efficiency was analyzed on a 15% TBE gel and it ranged between 85 to 95 %, showing that amount of the cyclic form predominates over the linear form of the STAT3 decoy (CS3D) and the mutant construct (CS3M). Tire linear form is also less stable, and is less likely to contribute to the biological effects in the presence of an excess of the cyclic form.

Fluorescently-!abe!ed CS3M and CS3D (with iFluor) were also produced using enzymatic ligation as described above and used to assess uptake efficiency. Transfections of the cyclic oligonucleotides were performed using Lipofectamine 2000 (Life Technologies) in opti- MEM media (Life Technologies) as follows: Cells were plated at 75-80% confluency, and exposed to transfection media containing the respective circularized oligonucleotides for 24 hours at 37°C. Post-transfection, cell recovery was initiated in media containing 10% heat- inactivated fetal bovine serum.

Example 2 MTS Assays and Programmed Cell Death Analysis

Dose-response experiments assessing the relative number of metabolically active viable cells were performed by transfecting cells with increasing concentrations of CS3D or CS3M, followed by performance of MTS assay s. After 72 horns, 5mg/ml of MTS reagent was added in the plates and incubated 37 °C for 20 minutes. The plates were then read at 490 nm in a Synergy microplate reader (BioTek, Winooski, VT) using Gen5 2.05 software. Data obtained following treatment with C3SD or CS3M w¾re normalized to treatment with lipofectamine alone. ICsos determined from these experiments were used to perform independent assays to confirm the effects of CS3D relative to CS3M. To assess induction of apoptosis in response to CS3D, H1975 and 201T cells were transfected with lOOnM of CS3M or CS3D. After twenty four hours of transfection, cells were resuspended in annexin V -binding buffer, incubated with 5pL of propidium iodide (BD Biosciences) and subjected to flow cytometric analysis, using a BD FACS Canto II Flow Cytometer.

Example 3 Colony Forming Assays

Following the transfection protocol described above, cells transfected with either CS3D or CS3M were detached from the plate using Trypsm-EDTA (Life Technologies) and seeded at a density of 2 x 10 4 cellsAvell (in a 6-well plate) in soft agar to determine the ability of cells to grow in an anchorage-independent setting. Sea Plaque Agarose (Lonza - Rockland, ME) was used to prepare 0.8% base agarose layer and a 0.48% upper agarose layer containing the transfected cells. 1ml of appropriate growth media was added into each well and incubated at growth conditions of 37°C with 5% CO2. Media was replaced every 3-4 days and colony formation was monitored. After 15-20 days, the media was aspirated, and wells were incubated with crystal violet in 10% formalin for 1 h. Using a dissecting scope, colonies were

photographed m a 6-well plate (with 4 quadrantsAvell) using Image! software analysis to automatically count colonies, using 35 pixels as a cut-off.

Example 4 Immunoblotting

Cells were seeded at 0.5 x 10 6 cells/well in six-well plates in BME/RPMi containing 10% FBS and transfected with 0. ImM CS3D/CS3M for 24 hours as described above. Post- transfection, cell lysates were extracted, and protein concentrations quantified using DC assay reagents. Whole cell lysates (20 micrograms/sample) were electrophoresed on 7.5% SDS- polyacrylamide gels for 1 h and transferred onto Trans-Blot polyvinyhdene difluonde (PVDF) membranes (Bio-Rad Laboratories) for 1 h at 100 V. The membranes were blocked using 5% nonfat dry milk, 0.1% Tween 20 in lx phosphate-buffered saline (TEST) for 1 h. Membranes w¾re incubated m primary antibody diluted at 1: 1000 at 4°C overnight, and washed three times with TEST (15 min/wash). The membranes w¾re then incubated with secondary antibody for 1 h (1 :2000) at room temperature, followed by three washes in TEST. Blots were later developed using a super-enhanced chemiluminescence substrate according to the manufacturer’s protocol (ThermoScientific). Image) l.X software w ? as used to quantify the results and assess changes in protein expression patterns.

Example 5 Confocal Microscopy

Twenty-four hours post-transfection with fluorescein-tagged CS3D, HI 975 cells were fixed in 4% paraformaldehyde and prolong antifade reagent (CST) w¾s applied directly to the slides. Fixed cell imaging was performed using a Nikon Eclipse Ti Confocal Microscope System. Images were captured and analyzed using the imaging software NIS Elements and Image J. Fluorescence detection of fluorescein-labelled CS3D was detected at 488 nm. Images were taken and processed at 40X and 60X.

Example 6 Immunoprecipitation (IP)

Post-transfection with either lOQnM CS3D or CS3M, HI 975 were lysed in IP lysis buffer (Thermo Scientific, Rockford, IL) supplemented with protease inhibitor cocktail (Roche, Basel, Switzerland) and phosphatase inhibitors (NaF and Na3V04). Immunoprecipitations were performed using the appropriate antibody (pSTAT3) at 1 :500 and Protein A Magnetic Beads (Thermo Scientific) at 4°C. Immunopredpitated lysates were electrophoresed on 7.5% SDS- polyaciydarmde gels followed by exposure to ubiquitin (1: 1000) at 4°C overnight and the appropriate secondar' antibody while following the immunoblotting method described above.

Example 7 Real-Time qPCR

Post transfection, cells were treated with 10 ng/m! EGF for 1.5 hr to activate STAT3. Trizol (Invitrogen, Carlsbad, CA) was then used to extract total RNA. One microgram of total RNA was reverse-transcribed using a cDNA synthesis kit (Quanta Biosciences) according the manufacturer’s instructions using a T100 Thermal Cycler (BioRad). Real-time qPCR

(polymerase chain reaction) was performed using a SYBR Green Super Mix kit on a CFX connect Real-Time System (BioRad). Gene-specific DNA oligonucleotide primers for STAT3 target genes were used to assess rnRNA levels normalized to GAPDH mRNA levels as internal control, and the ratio of normalized mRN A to the control conditions was de termined using the comparative ACT method for analysis. The primers used for real-time qPCR are as follows:

Example 8 In Vivo Tumor Xenograft Studies

All studies were approved by the University of Minnesota Institutional Animal Care and Use Committee and were carried out in accordance with institutional guidelines for animal care. Female nude mice (4 - 6 weeks old) were injected with lxlG 6 ceils in the flanks. After 3 weeks, animals with palpable tumors (200mm 3 ) were randomized into two treatment groups with 10 tumors/group. Daily tail vein injections (5 days per week) of the cyclic STATS decoy (CS3D) or mutant STATS control (CS3M) (lOOpg in 200m1 per injection) was delivered for 2 - 3 weeks. Tumor measurements were made with calipers every 2 days. Volumes were calculated from the formula V= (Length x Width 2 )/2. After treatment, the animals were sacrificed according to IACUC guidelines.

immunohistochemical analysis: Tumors wure excised for immunohistochemical (IHC) staining; 5-pm (micron) sections were cut from formalin-fixed paraffin-embedded tissue blocks, deparaffinized and rehydrated using successive wushes of xylene followed by ethanol. Antigen retrieval was performed in a microwuve oven (for 20 minutes) in an unmasking solution containing sodium citrate buffer followed by peroxidase blocking in 3% hydrogen peroxide. The sections were incubated in ABC blocking buffer (Vector laboratories Inc, Burlingame, CA) for 1 hour at room temperature. Sections were then stained with H&E (hematoxylin and eosm) or subjected to sequential incubations with different primary antibodies and peroxidase-conjugated goat anti-rabbit secondary antibodies. The following primary antibodies were used (at the indicated dilutions): cleaved caspase-3 (#9664, Cell Signaling, 1 :300), p-STAT3 (#9145, Ceil Signaling, 1 :400), Ki67 (#abl 5580, Abeam, 1 : 1000). Sections were developed with DAB and counterstained with hematoxylin. Bright field microscopy was performed using Leica DM 4000 B LED microscope and images were captured at 20X and 40X magnification using LASv4.7 software. IHC analysis was done as a blinded study with 100 images from each treatment group graded as low, moderate, or high. Staining was graded as low (<30% positive cells per field, scored as 1 ), moderate (30-60% positive cells per field, scored as 2) or high (> 60% positive cells per field, scored as 3).

Statistical Analyses: For statistical analyses, data were reported as mean ± standard deviation (SD) or standard error of the mean (SEM) To assess significance between different treatment groups, a student’s t test (two tailed) was used to determine significance with p values at least < 0.05 categorized as statistically significant. To analyze IHC scoring data, Chi-Squared test w¾s used to compare the frequency of scores of 1,2, or 3 in treatment groups.

Example 9 In vivo orthotopic NSCLC tumor growth NSCLC cells are injected by tail vein into scid (severe combined immunodeficiency) mice, using cells tagged with a LUC vector that are suspended in saline with 15% Matrigel. NSCLC cell lines to be examined are 20 IT, HI 975 and A549 from Table 1, as well as H23, which is EGFR WT/KRAS G12C mutant. Following 1 wk (week) for engraftment, tumor growth wall be monitored by bioluminescence. Placebo or HER TKI is then given by gavage and STATS decoys (CS3D and CS3M) are administered by IV injection 5 days per week, for 14-21 days, depending on response. Groups will be vehicle alone, afatinib (GILOTRDF®, Boehringer Ingelheim) alone (25 mg/kg [standard dose]), CS3D alone (5 nigy'kg), CS3M alone (5 mg/kg), afatinib combined with CS3D and afatinib combined with CS3M. In addition to imaging, lung tumors are also assessed at sacrifice histologically and by IHC (in formalin fixed paraffin embedded tissues [FFPE]) and by immuno blotting of frozen unfixed lysates after sacrifice (2 lung lobes are set aside from each mouse to prepare ly sates). All animal procedures are in accordance with IACUC guidelines and approval. Doses will be adjusted if needed for each cell line. Mice are monitored for weight, activity level, and breathing difficulty. For therapy studies,

8 animals per group will give 80% power at p :::: 0.05 to detect a mean >25% effect size, based on 20% variation among animals. 10 animals/group will be injected to account for average 80% tumor engraftment. An equal number of male and female mice will be used as the host animal.

After determining which orthotopic model responds to CS3D and whether combination with an EGFR TKI is efficacious, lung cancer PDXs, available from Jackson Laboratories, may be used. PDXs with and without EGFR mutation (including #TM002Q4 with an T790M resistance mutation) and with KRAS mutations if applicable (such as J000095635) will be propagated as subcutaneous xenografts (8 per group) and optimal drug regimen will be tested compared to controls, with tumor size monitored with calipers and signaling evaluation.

Monitoring uptake of CS3D into orthotopic tumors will be done with spiking of F1TC- conjugated CS3D (0.5 mg/kg) along with unlabeled CS3D in a separate set of 10 tumor-bearing animals. Accumulation of CS3D is examined by fluorescent microscopy of lungs, livers and spleens from 2 animals collected at 0 time and 1 hr after daily injections on days 1 -5 to estimate tissue accumulation after 1-5 doses.

Assessment of signaling molecules: total/phospho-STAT3, total/phospho-EGFR/HERs, total/phospho-NFicB (NF-kappa B), total/phospho MAPK and AKT, as well as protein levels of cyclin Dl, Myc, VEGF, IL6 and IL8, and caspase 3 (for apoptosis) will be measured by IHC and/or immunoblotting in tumor lysates. Proliferation index will be assessed by Ki67 labeling. Angiogenesis will be assessed by CD31 IHC staining. EMT/invasion: ECadherin and Snail will be assessed by IHC for EMT; CD44 and focal adhesion kinase (FAK) activation using p-FAK antibody wall be assessed to determine invasion. If reduced effects on EMT or invasion are seen this will then be examined after 5 days of CS3D, to determine if reduced EMT/mvasion precedes growth effects.

Mean and median tumor volumes based on bioluminescence will be calculated over time of treatment and compared by ANOVA followed by t-test. Significance will be at P < .05. For 1HC. scoring will be done on a 1-3 scale (low, medium, high) as previously published (Siegfried et al , Oncotargel (2017) 8:24063-76). FFPE sections from 5 tumors/treatment group will be analyzed, with 20x fields/section (100 total) scored blindly and compared by ANOVA followed by t-test. Positive controls (tumors with known staining) and negative controls (leaving out primary antigen) will be done. For western blotting, individual lysates for 5 tumors/group will be run in duplicate with results normalized for GAPDH or actin. Positive control will be cell lysates known to express protein studied.

Alternate approach: Osimertinib (TAGRJSSi)®, AstraZeneca) is a HER TKI developed to treat NSCLC with the EGFR T790M resistance mutation and overcomes the inability of erlotinib to bind to the ATP binding pocket with the T790M alteration. Data suggests that EGFR WT cells are also more sensitive to osimertinib than to erlotinib. Combination therapy by in vitro and in vivo combination index studies with osimeritinib and CS3D may be conducted in selected cell lines to detect and measure synergy.

Example 10 In vivo NNK-induced lung tumor model for chemoprevention

The chemopreventive effect of CS3D compared to CS3M on lung tumor development was studied in an NNK-induced immunocompetent lung tumor model using the tobacco carcinogen, 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone, CAS Reg. No. 64091-91-4 (NNK) in mice. The advantages of this model are: NNK is an inducer of KRAS mutations, it is known to induce lung cancers m mice and humans, and the model is immunocompetent. In addition, this is a useful model for studyin chemoprevention after exposure to a carcinogenic agent (Hecht SS, et al, Cancer Res. (1980) 40(2): 298-302).

After four weeks of NNK exposure, mice are then dosed IV 2 times per week with CS3D or CS3M (5 mg/kg) for eight weeks. The mice are sacrificed after at one week, 8 weeks, and 20 weeks after the end of oligonucleotide dosing to examine lungs. Lungs are formalin-inflated and surface tumors are enumerated visually under a dissecting microscope. The area of each tumor is measured with a analysis tool built m the imaging software LAS v4.7 (Leica). Lungs are also sectioned and examined histologically by hematoxylin and eosin staining for counting the preneoplasia and tumors.

Immunohistoehemistry is performed on the lung section to determine the expression levels of p-STAT3, P-NFkB, VEGF, IL-6 and COX2. For this 5pm thick FFPE sections are deparaffinized in xylene and rehydrated in alcohol. Heat mediated antigen retrieval is performed in citrate buffer pH 6.0. The tissue is blocked for nonspecific binding by incubating in 10% goat serum. The tissues sections are incubated with primary antibodies (pSTA ' 13; 1 :400, C OX2; 1 :600 Cell Signaling), 1L-6(1:600; Thermoscientific) VEGF(1:400 Abeam), pNFkB (1 :500) Abeam). The primary antibody binding is determined by incubating with HRP conjugated secondary antibody and color developed by incubating the samples in DAB and counter stained with haemotoxylin. The bright field images are captured at 20X or 40X using Leica DM 4000B LED microscope equipped with LASv4G7 software. Staining is graded as low (< 30% positive cells per field score 1, moderate (30%-60% positive cells per field, scored as 2), or high (>60% positive cells per field, scored as 3).

In order to determine the KRAS status of individual tumor, 10 um FFPE lung sections are mounted on special Leica foiled glass slides. Sections are stained with H&E and slides are mounted on Laser captured Microdissection (LCM) stage of LMD6500 laser microdissection microscope. Desired tumor sections are microdissected and captured in labeled tubes. Total DNA is isolated using DNA isolation kit (Qiagen). To amplify exon 1 that contains codon 12/13, 50ng of DNA sample was used for the PCR amplification reaction in a final 25 pL volume containing IX GoTaq Green Master Mix (12.5pL) (Promega), 2mM each of the pair of primers (forward: 5'-GACATGTTCTAATTTAGTTG-3' (SEQ ID NO: 18); reverse: 5'- AGGCGCTCTTGCCTACGGCA-3' (SEQ ID NO: 19)) [153], and nuclease-free water to 25pL. The mixture was heated at 95°C for 9 minutes then subjected to 40 cycles (94°C/1 min, 53°C/1 min, 72°C/1 min). For the 2 nd round PCR, lpL of the first round PCR products was then diluted into a final 25 pL with same reaction buffer but amplified with a different set of primers (forward:

5’GCCGCCTGCAGCCCGCGCCCCCCGTGCCCCCGCCCCGCCGCCGGCCCGGCGCCCT ATTGTAAGGCCTGCTGAAAAT-3' (SEQ ID NO: 20); reverse: 5'- AGGCGCTCTTGCCT ACGGC A-3 ' (SEQ ID NO: 19)) [153] The 2 nd reaction was heated at 95°C for 9 min then subjected to 25 PCR cycles (94°C/l min, 60°C/1 min, 72°C/l min). The PCR products containing the 126 hp exon 1 fragment were then sequenced (Sanger) and analyzed for the G12D substitution mutation using ExPASy Translate tool.

Example 11 Flow' cytometry on the cells isolated from the lungs from CS3M and CS3D mice.

Lungs from mice were digested with RPMI media containing collagenase and DNase to obtained single cell suspension. The cells were counted and lxl 0 6 cells were staining with fluorescent labeled CD45, CDl lb, MHC II, Ly6C and F4/80. The labeled cells were gated and data acquired on LSR fortessa (BD Biosciences) and further analysed by Flowjo.

Example 12 Representative pharmaceutical formulations and dosage forms as tablets, capsules, parental injection, and aerosol, containing a compound of formula I, for therapeutic or prophylactic use in humans.

(i) Tablet 1 mg/tablet

STAT3 compound 100.0

Lactose 77

Povidone 15.0

Croscarmeiiose sodium 12.0

Mi crocrystalline cellulose 92.5

Magnesium stearate 0

300.0 total mg/tablet

STAT3 compound 20.0

Microcrystalline cellulose 410.0

Starch 50 0

Sodium starch g!ycolate 15.0

Magnesium stearate 5.0

500.0 total

(hi) Capsule mg/capsule

STAT3 compound 10.0

Colloidal silicon dioxide 1.5

Lactose 465.5

Pregelatinized starch 120.0

Magnesium stearate TO

600.0 total

(iv) Injection 1 (1 u nil) mg/ ' ml

STAT3 compound 1.0

Dibasic sodium phosphate 12.0

Monobasic sodium phosphate 0.7

Sodium chloride 4.5

1.0 N Sodium hydroxide solution

(pH adj ustment to 7.0-7.5) q.s.

Water for injection q.s. ad 1 ml

(v) Injection 2 (10 mg/'ml) mg/ml

STAT3 compound 10.0

Monobasic sodium phosphate 0.3 Dibasic sodium phosphate 1.1

Polyethylene glycol 400 200.0

LO N Sodium hydroxide solution

(pH adjustment to 7.0-7.5) q.s.

Water for injection q.s. ad 1 mL

(vi¾ Aerosol mg/can

STATS compound 20.0

Oleic acid 10.0

Trichloromonofluoromethane 5,000.0

Diehl orodi fl uorometha e 10,000.0

Dichi orotetrafluoroethane 5,000.0

The above formulations are only representative and may be obtained by conventional procedures well known in the pharmaceutical art.

All publications, patents, and patent documents are incorporated by reference herein, as though individually incorporated by reference. The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention.