Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 May 29.
Published in final edited form as: Nat Rev Drug Discov. 2013 Aug;12(8):611–629. doi: 10.1038/nrd4088

Therapeutic modulators of STAT signalling for human diseases

Gabriella Miklossy 1,*, Tyvette S Hilliard 1,*, James Turkson 1
PMCID: PMC4038293  NIHMSID: NIHMS580732  PMID: 23903221

Abstract

The signal transducer and activator of transcription (STAT) proteins have important roles in biological processes. The abnormal activation of STAT signalling pathways is also implicated in many human diseases, including cancer, autoimmune diseases, rheumatoid arthritis, asthma and diabetes. Over a decade has passed since the first inhibitor of a STAT protein was reported and efforts to discover modulators of STAT signalling as therapeutics continue. This Review discusses the outcomes of the ongoing drug discovery research endeavours against STAT proteins, provides perspectives on new directions for accelerating the discovery of drug candidates, and highlights the noteworthy candidate therapeutics that have progressed to clinical trials.


The signal transducer and activator of transcription (STAT) protein family is composed of seven members: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6. Structurally they share five domains, which are an amino-terminal domain, a coiled-coil domain, a DNA-binding domain, an SH2 domain and a carboxy-terminal transactivation domain (FIG. 1). The transactivation domain contains one or two amino acid residues that are crucial for the activity of the STAT protein; that is, phosphorylation of a particular tyrosine residue promotes dimerization, whereas phosphorylation of a particular serine residue enhances transcriptional activation13.

Figure 1. A schematic representation of the structures of the STAT proteins.

Figure 1

Linear representations of the domain structures of the seven members of the signal transducer and activator of transcription (STAT) protein family: STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B and STAT6. The transactivation domain contains a crucial tyrosine (Y) residue, the phosphorylation of which initiates STAT activation and dimerization between two monomers through a reciprocal phosopho-Tyr-SH2 domain interaction. The serine (S) residue present in the transactivation domain of certain STAT proteins is thought to enhance transcriptional activity when it is phosphorylated.

The STAT proteins were discovered as cytoplasmic transcription factors that mediate cellular responses to cytokines and growth factors1,2 (FIG. 2). Once a ligand interacts with its receptor, STAT activation is induced by the phosphorylation of the key tyrosine residue in the STAT transactivation domain by growth factor receptors, Janus kinases (JAKs), SRC family kinases and other tyrosine kinases. This leads to numerous events including STAT–STAT dimerization through a reciprocal phospho-tyrosine (pTyr)-SH2 domain interaction, nuclear translocation, DNA binding and the transcriptional induction of genes in the nucleus. Physiological negative regulators, such as suppressors of cytokine signalling (SOCS) and protein tyrosine phosphatases (PTPs), ultimately downregulate the active STAT signalling.

Figure 2. STAT signalling pathway, functions and associated diseases.

Figure 2

Activation of signal transducer and activator of transcription (STAT) is promoted when a ligand (L) binds its receptor (R). The ligand–receptor interaction induces receptor phosphorylation (P), which recruits the STAT proteins to the phospho-motifs of the receptor. Phosphorylation of the critical tyrosine residue in the STAT protein is then initiated by tyrosine kinases, such as growth factor receptors, Janus kinases (JAKs) and SRC family kinases. Two phosphorylated STAT monomers dimerize through reciprocal pTyr-SH2 domain interactions, and the STAT–STAT dimers translocate to the nucleus where they bind to specific STAT-response elements in the target gene promoters and regulate transcription. The STAT-dependent induction of genes is essential for many physiological functions. The activation of normal STAT signalling is controlled by physiological negative modulators, such as suppressors of cytokine signalling (SOCS) and protein tyrosine phosphatases (PTPs), in accordance with normal cellular functions. Although the STAT proteins (STAT1 to STAT6) are differentially activated and promote varying cellular processes depending on the ligand and the context, collectively, their normal induction regulates cell growth, differentiation, survival and apoptosis. Their normal induction also regulates inflammatory and immune responses, embryonic development and mammary gland development. By contrast, defective or abnormal STAT signalling is associated with various human diseases, including susceptibility to infection, immune disorders, many types of cancer, asthma and allergic diseases. IFN, interferon; IL-2Rα, interleukin-2 receptor-α; TH, T helper.

STAT proteins promote fundamental cellular processes, including cell growth and differentiation, development, apoptosis, immune responses and inflammation1,2 (FIG. 2). STAT1 signalling is activated in response function partly by controlling the growth and apoptosis of immune cells4. STAT1 signalling regulates T helper type 1 (TH1) cell-specific cytokine production that alters both immune function and inflammatory responses by shifting the balance between TH1 and TH2 cells5. Indeed, STAT1 deficiency abrogates IFN responsiveness, leading mice to succumb to bacterial and viral infections6. Furthermore, the loss of responsiveness to IFNγ due to STAT1 deficiency provides malignant cells with a growth advantage and leads to increased tumour formation4. This outcome suggests that STAT1 has a tumour-suppressive function; although recent data indicate that the protein has a more complex role in carcinogenesis4. Moreover, in STAT1-null mouse models of atherosclerosis-susceptible bone-marrow transplantation, these mice have reduced foam cell formation and atherosclerosis, which suggests that STAT1 has a pro-atherogenic function7. By contrast, gain-of-function mutations in the STAT1 gene — which lead to STAT1 hyperactivation and defective nuclear dephosphorylation — affect TH1 and TH17 cell responses and cause chronic mucocutaneous candidiasis8,9.

STAT2 signalling is important for the induction of antiviral effects. STAT2-null mice and STAT2-null cell lines have defective antiviral responses to IFNα and IFNβ, as well as blunted apoptotic effects to IFNα and IFNβ10. Evidence further suggests that altered STAT2 signalling may partly contribute to carcinogenesis through the upregulation of interleukin-6 (IL-6) production, which promotes STAT3 activation10. Notably, the colon of STAT2-deficient mice showed markedly lower levels of IL-6, which was associated with diminished tumour progression10. By contrast, the reconstitution of STAT2 in the null background upregulated IL-6 production and increased the levels of pSTAT3.

STAT3 function is essential for early embryonic development, which is clearly demonstrated by the death on day 8.5 of STAT3-deficient mouse embryos11. The biological importance of STAT3 was further studied using tissue-specific STAT3-deficient mice12. In vitro cultured STAT3-deficient T cells did not respond to IL-6 stimulation and could not be rescued by IL-6 from apoptotic cell death, indicating that STAT3 functions are essential for IL-6-mediated anti-apoptotic responses13. Furthermore, STAT3-deficient keratinocytes showed a poor wound-healing response in vitro and in vivo, which was due to the limited migration of the cells14. In addition, the dys-regulation of the role of STAT3 in keratinocyte physiology is thought to contribute to the induction of skin carcinogenesis15. STAT3 function is often aberrant in the context of cancer, and this abnormality represents an underlying mechanism of STAT3 for promoting malignant transformation and progression. Of clinical significance, constitutively active STAT3 is detected in numerous malignancies, including breast, melanoma, prostate, head and neck squamous cell carcinoma (HNSCC), multiple myeloma, pancreatic, ovarian, and brain tumours1623. Aberrant STAT3 signalling promotes tumorigenesis and tumour progression partly through dysregulating the expression of critical genes that control cell growth and survival, angiogenesis, migration, invasion or metastasis1619,2123. These genes include those that encode p21WAF1/CIP2, cyclin D1, MYC, BCL-X, BCL-2, vascular endothelial growth factor (VEGF), matrix metalloproteinase 1 (MMP1), MMP7 and MMP9, and survivin1619,2123. Evidence also supports a role of STAT3 in the suppression of tumour immune surveillance24,25. Consequently, the genetic and pharmacological modulation of persistently active STAT3 was shown to control the tumour phenotype and to lead to tumour regression in vivo1823,2630. Of further clinical significance, mutations within the SH2 domain-coding sequence in STAT3 occur in patients with a rare primary immunodeficiency, hyper-immunoglobulin E syndrome31, and in large granular lymphocytic leukaemia32. These mutations reportedly increase the stability of the functional STAT3–STAT3 dimers and are associated with the disease pathogenesis32,33.

STAT4 is a crucial mediator of IL-12 function that regulates the differentiation of TH1 cells and their inflammatory responses5. Accordingly, STAT4 signalling is associated with autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE) induction, an animal model of multiple sclerosis34. Indeed, mice deficient in the Stat4 gene were protected from developing EAE35. Additionally, STAT4-null lymphocytes showed decreased proliferation and had an impaired response to IL-12 (REF. 36). Treatment with a STAT4-specific antisense oligonucleotide (ASO) led to an improvement in the systemic lupus erythematosus (SLE) phenotype, severe lupus nephritis in mouse models, which suggests that STAT4 also has an important role in SLE, even though STAT4 knockout had no effect on the clinical presentation of the disease in mouse models36.

STAT5 has two isoforms, STAT5A and STAT5B, which are encoded by distinct genes and share 96% sequence homology, with notable differences occurring in the transactivation domain37. Normal STAT5 signalling is important in mammary gland development and milk production, and in haematopoiesis38. Mice deficient in both STAT5A and STAT5B show defects in IL-2 receptor-α expression in T lymphocytes12. By contrast, mice that are deficient in STAT5B only showed loss of sexual dimorphism of body growth rate, which was accompanied by a decreased expression in male-specific liver genes39 and the attenuation of the cytolytic activity of natural killer cells40. Constitutive STAT5 activation is also implicated in the pathogenesis of HNSCC, chronic myelogenous leukaemia (CML)41, breast42, prostate and uterine cancers19. The aberrant STAT5 activation by BCR–ABL in CML43 is particularly noteworthy. It is widely recognized that STAT5 and STAT3 share similar functions in promoting cancer, including the induction of pro-proliferative and anti-apoptotic genes44,45.

STAT6 signalling is induced by IL-4 and IL-13 and supports immune function, notably regulating the balance between inflammatory and allergic immune responses4649. Beyond the immune system, STAT6 signalling promotes luminal mammary epithelium development and is implicated in the pathology of lung and airway disease, including the promotion of airway hyperactivity and mucus production in the lung epithelium, and the regulation of allergic skin inflammation46,5052.

Mutations in JAKs, as observed in certain haematological disorders, immunodeficiencies and cancers, lead to dysregulation of STAT signalling that contributes to these disease phenotypes53,54. The clinical significance of the JAK–STAT pathway in diseases is exemplified by an ongoing clinical study of patients with primary immunodeficiency disorders to identify mutations in the JAK3, STAT1 and STAT4 genes, among others (ClinicalTrials. gov identifier: NCT00001788).

Therapeutic approaches that target STAT proteins

It has been more than a decade since the first peptide inhibitor of a STAT protein was reported30 and efforts to target STAT signalling for therapeutic purposes continue. Extensive drug discovery activities targeting STAT3 are well underway, as evident by the numerous publications related to this protein. This is in contrast to the moderate research efforts for inhibitory modulators of STAT1, STAT2, STAT4, STAT5 and STAT6, as demonstrated by the few published reports. Various inhibitory strategies against STAT signalling and function are being pursued (FIG. 3). Most of these focus on inhibiting STAT dimerization using peptides or peptidomimetics generated through structure-based design, small molecules identified by molecular modelling, virtual or library screening, or natural products. STAT dimerization and signalling can also be blocked by inhibiting tyrosine kinases that phosphorylate STAT proteins, or by inducing phosphatases that dephosphorylate STAT proteins. Other approaches, discussed in BOX 1 and BOX 2, include the use of oligodeoxynucleotide (ODN) decoys as specific STAT DNA-binding domain inhibitors and ASOs that interfere with STAT mRNA.

Figure 3. Models of inhibition of STAT signalling.

Figure 3

a | Small-molecule dimerization disruptors (SMDDs) or phospho-peptidomimetic inhibitors (PPMIs) target the phospho-Tyr-SH2 domain interaction at the interface of dimers of signal transducer and activator of transcription (STAT) proteins. This leads to the disruption of STAT–STAT dimers and the formation of STAT–SMDD or STAT–PPMI heterocomplexes, leading to a suppression of STAT signalling and function. b | The binding of ligands, such as growth factors and cytokines, to their cognate receptors on the cell surface induces STAT tyrosine phosphorylation and activation, and this process is competitively inhibited by antibody-based therapeutics. Tyrosine kinases that mediate STAT phosphorylation, leading to STAT–STAT dimer formation, are the targets of small-molecule tyrosine kinase inhibitors. The overall effect of these modulators is to block the induction of STAT phosphorylation and signalling. c | Decoy oligdeoxyonucleotides (ODNs) compete against the DNA-response elements in the target gene promoters for the binding of the active STAT–STAT dimers. The interaction of ODNs with STAT–STAT dimers precludes the binding of the dimers to target gene promoters, thereby preventing the induction of STAT-responsive genes. d | The induction of protein tyrosine phosphatases has been noted to occur in response to certain STAT inhibitors, including sorafenib analogues and natural products. This promotes the dephosphorylation of the phospho-STAT proteins leading to the suppression of their signalling and function.

Box 1. Oligodeoxynucleotide decoy approach to inhibit STAT functions.

One approach to modulate STAT signalling is to use oligodeoxynucleotide (ODN) decoys as specific DNA-binding domain inhibitors28,197 (FIG. 3c; TABLE 1). ODN decoys compete with endogenous promoter sequences for binding to the target transcription factor, thereby preventing gene expression. In a rat cardiac allograft model of atherosclerosis, a 25-bp STAT1 ODN decoy based on the γ-activated sequence (GAS) element198 specifically downregulated the protein and mRNA levels of CD40. CD40 is a STAT1 target gene that is involved in T-cell activation, and suppression of its expression by the STAT1 ODN may account for the protective effects of the STAT1 ODN against atherosclerosis development199. Additionally, in a myelogenous leukaemia cell line (K562), treatment with a 21-bp ODN element containing a modified version of the GAS element that binds STAT5 suppressed STAT5 transcriptional activity, which led to cell cycle arrest and apoptosis200. This result suggests that blocking STAT5, which is downstream of the constitutively activated BCR–ABL, with an ODN decoy can be a promising therapeutic approach to overcome imatinib resistance in chronic myelogenous leukaemia200.

The use of ODN decoys to target STAT3 is further advanced. A 15-bp STAT3 ODN decoy (5′-CATTTCCCGTTAATC-3′) was initially designed on the basis of the sequence of the high-affinity cis-inducible element of the FOS promoter28. A modified version of this STAT3 ODN decoy (5′-CATTTCCCGTAAATC-3′) was evaluated in mice bearing glioblastoma xenografts201. Intratumoural injection of this modified version downregulated STAT3 target genes and decreased tumour growth201. Furthermore, the inhibition of STAT3 function by the same ODN decoy sensitized resistant head and neck squamous cell carcinoma (HNSCC) and bladder cancer cells to the epidermal growth factor receptor (EGFR) inhibitors cetuximab and erlotinib202. Although this STAT3 ODN decoy disrupted STAT1 signalling in vitro, its inhibitory effect on tumour cell growth is reportedly independent of the STAT1 activation status203.

To evaluate the potential clinical application of ODN decoys, a single-dose intramuscular injection of the STAT3 ODN decoy with the sequence 5′-CATTTCCCGTTAATC-3′ was evaluated for toxicities in a non-human primate model28. Encouragingly, this ODN decoy showed no systemic or organ-specific toxicity when it was given over a 2-week period. There was no observable dose–response effect on STAT1 activation in peripheral blood cells, whereas pSTAT3 levels decreased28. A Phase 0 clinical trial of the STAT3 ODN decoy 5′-CATTTCCCGTTAATC-3′ (ClinicalTrials. gov identifier: NCT00696176) assessed the safety of a single dose intratumoural injection in patients with HNSCC196. This decoy showed a reasonable safety profile and suppressive effects on the expression of STAT3 target genes196.

More stable cyclic versions of the STAT3 ODN decoy 5′-CATTTCCCGTAAATC-3′, which are resistant to serum nucleases, also decreased the expression of STAT3-regulated genes and suppressed the viability of HNSCC and bladder cancer cells196. A hairpin ODN version with a modified consensus sequence containing two STAT3-binding sites that achieves a STAT3/STAT1 discriminating property was efficacious in SW480 colon cancer cells197. Together, these studies suggest that the clinical development of STAT3 ODN decoys has great promise196.

Box 2. Antisense oligonucleotide approach to inhibit STAT functions.

The use of antisense oligonucleotides (ASOs) to target aberrant signal transducer and activator of transcription 3 (STAT3) signalling in cancer has yielded promising results. Preclinical studies have shown that STAT3-specific ASOs inhibited STAT3 expression and downregulated the STAT3 target gene vascular endothelial growth factor (VEGF) in melanoma and mammary carcinoma models in vitro204. Additionally, they suppressed the malignant phenotype in prostate cancer205 and hepatocellular carcinoma cells206.

Moving towards a therapeutic application of ASOs to inhibit STAT functions, a phosphorothioate-modified chimeric ASO sequence that targets human STAT3 mRNA has been designed by ISIS Pharmaceuticals (ISIS 481464). Studies in cynomolgus monkeys showed that ISIS 481464 given at a dose of 10 mg per kg per week for 6 weeks was sufficient to decrease STAT3 protein levels by up to 90%207. Importantly, the agent was well tolerated when given at doses of up to 30 mg per kg per week for 6 weeks, with no treatment-related changes in clinical signs or treatment-related deaths207.

In the following section, we discuss direct inhibitors reported for each member of the STAT protein family. There are no significant discoveries of STAT2 inhibitors, and so these are not discussed here, while inhibitors of STAT1 and STAT4 are discussed together owing to the low volume of reports and their relatively similar functional roles. Tyrosine kinase inhibitors (TKIs), which broadly affect the induction of multiple STAT proteins by inhibiting the kinases that phosphorylate and thereby activate the STAT proteins, are discussed at the end of the section.

Inhibitors of STAT3

There are several human cancers that could benefit from therapeutics that target aberrantly active STAT3. Owing to its therapeutic significance, STAT3 is the target in many drug discovery research efforts (TABLE 1). Approaches to target STAT3 also include TKIs, ODN decoys (BOX 1) and ASOs (BOX 2), which are discussed separately. STAT3 drug discovery research has mostly focused on targeting the pTyr-SH2 domain interaction2123 given its importance in promoting STAT3 dimerization and function, and these efforts have generated various inhibitory agents. However, there are many reported STAT3 inhibitors that may induce their effects through multiple mechanisms.

Table 1.

STAT signalling modulators, their targets and effects, and clinical indications

Molecule Target Effect Indication (clinical trial phase) Refs
Peptide and peptidomimetics
PY*LKTK STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 30
Ac-pTyr-Leu-Pro-Gln-Thr-Val-NH2 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 56,59
ISS-610 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 7,30,55,58
PM-73G STAT3 SH2 domain Inhibits phosphorylation of STAT3 Not applicable 62,63
CJ-1383 STAT3 SH2 domain Inhibits phosphorylation of STAT3 Not applicable 61
ISS-840 STAT1 SH2 domain Inhibits STAT1–STAT1 dimerization Not applicable 58
Non-peptide small molecules
S3I-M2001 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 27,55
STA-21 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Psoriasis (Phase I/II) 64
Catechol moiety STAT3 SH2 domain Inhibits STAT3 DNA binding Not applicable 66
Stattic STAT3 SH2 domain Inhibits phosphorylation of STAT3 Not applicable 72
LLL12 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 83,84
FLLL32 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 80
S3I-201 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 26
BP-1-102 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 68
S3I-201.1066 STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Not applicable 67
Pravastatin STAT1 Inhibits phosphorylation of STAT1, decreases levels of IFNγ Schizophrenia (Phase IV), pre-eclampsia (Phase I), hyperlipidaemia (Phase I/II/III/IV), cirrhosis (Phase II/III), gastroesophageal cancer (Phase IV), myeloid leukaemia (Phase I/II), pneumonia (Phase 0) 143,144
Pimozide STAT5 Inhibits phosphorylation of STAT5 Schizophrenia (Phase II/IV), Tourette’s syndrome (Phase II), psychosis (Phase III), post-operative nausea (phase not provided) 41
Chromone nicotinyl hydrazone STAT5 DNA-binding domain Inhibits phosphorylation of STAT5 Not applicable 149
AS151749951 STAT6 Inhibits phosphorylation of STAT6 Not applicable 51
Leflunomide Various protein kinases Inhibits phosphorylation of STAT6 Rheumatoid arthritis (Phase I/II/III/IV), lupus nephritis (Phase II/III), uveitis (Phase II), melanoma (Phase I/II), prostate cancer (Phase II/III), glioblastoma multiforme (Phase III) 50
Niflumic acid JAK2, STAT6 Inhibits phosphorylation of STAT6 Not applicable 157
YM-341619 STAT6, IL-4 Inhibits phosphorylation of STAT6 Not applicable 155
AS1810722 STAT6, IL-4 Inhibits phosphorylation of STAT6 Not applicable 156
Natural products
Capsaicin STAT3, gp130 Inhibits STAT3 expression, depletes gp130 Chronic obstructive pulmonary disease (Phase 0/I/II), psoriasis (Phase IV), chronic neck pain (Phase II), rhinitis (Phase I/II/IV), pulmonary hypertension (Phase II), HIV infections (Phase II/III), peripheral nervous system diseases (Phase II/III), migraine (Phase I), burning mouth syndrome (Phase 0) 104
Curcumin JAK1, JAK2, JAK3, STAT3 Inhibits phosphorylation of STAT3 Pancreatic cancer (Phase II/III), colon cancer (Phase I/ II/III), breast cancer (Phase II), head and neck cancer (Phase 0), osteosarcoma (Phase I/II), multiple myeloma (Phase II), atopic asthma (phase not provided), dermatitis (Phase II/III), type 2 diabetes (Phase IV), schizophrenia (Phase I/II), Alzheimer’s disease (Phase I/II), multiple sclerosis (Phase II), rheumatoid arthritis (Phase 0) 78,79
Avicin D JAK1, JAK2, STAT3 Inhibits phosphorylation of STAT3 Not applicable 109
Resveratrol JAK1, STAT3 Inhibits phosphorylation of STAT3 Colorectal cancer (Phase I), follicular lymphoma (Phase II), cardiovascular diseases (Phase I/II), type 2 diabetes (Phase I/II/III), obesity (Phase II), Alzheimer’s disease (Phase II/III), memory impairment (phase not provided) 93
Piceatannol JAK1, STAT3 Inhibits phosphorylation of STAT3 Not applicable 9698
Sanguarine JAK2, SRC, STAT3 Inhibits phosphorylation of STAT3 Not applicable 137
Celastrol JAK2, SRC, STAT3 Inhibits phosphorylation of STAT3 Not applicable 107,108
Withaferin A JAK2, STAT3 Inhibits phosphorylation of STAT3 Schizophrenia (phase not provided) 110,111
Cucurbitacin I JAK2, STAT3 Inhibits phosphorylation of STAT3 Not applicable 119122
Cucurbitacin B JAK2, STAT3 Inhibits phosphorylation of STAT3 Not applicable 123
3,3′-diindolyl-methane JAK2, STAT3 Inhibits phosphorylation of STAT3 Breast cancer (Phase I/II/III), prostate cancer (Phase I/II), cervical dysplasia (Phase III), laryngeal papilloma (Phase II), thyroid disease (Phase 0) 141
Emodin JAK2, STAT3 Inhibits phosphorylation of STAT3 Polycystic kidney disease (phase not provided) 128
Cucurbitacin E JAK2, VEGFR2, STAT3 Inhibits phosphorylation of STAT3 Not applicable 124126
Oleanolic acid/CDDO-Me JAK2, SRC, EGFR, STAT3 Inhibits phosphorylation of STAT3 Solid tumours and lymphomas (Phase I), chronic kidney disease and type 2 diabetes (Phase I/II/III), diabetic nephropathy (Phase II), hepatic dysfunction (Phase I/II) 116118
Caffeic acid JAK2, SRC, STAT3 Inhibits phosphorylation of STAT3 Not applicable 100
Vinorelbine STAT3 Inhibits STAT3 phosphorylation, disrupts STAT3–tubulin interaction Non-small cell lung cancer (Phase I/II/III/IV), breast cancer (Phase I/II/III/IV), prostate cancer (Phase I/ II), rhabdomyosarcoma (Phase I/II/III), gastric cancer (Phase II), melanoma (Phase II), low-grade gliomas (Phase II), Hodgkin’s lymphoma (Phase I/II/III) 106
Paclitaxel STAT3 Inhibits STAT3 phosphorylation, disrupts STAT3–tubulin interaction Peritoneal cavity carcinoma (Phase I/II/III), breast cancer (Phase I/II/III/IV), ovarian cancer (Phase I/II/III/ IV), cervical cancer (Phase I/II/III), non-small cell lung cancer (Phase I/II/III/IV), bladder cancer (Phase I/II/III), melanoma (Phase I/II/III), oesophageal cancer (Phase I/II/III), thyroid cancer (Phase I/II/III), gastric cancer (Phase I/II/III) 106
Evodiamine JAK2, SHP1, STAT3 Decreases JAK2 phosphorylation, increases SHP1 expression leading to inhibition of activated STAT3 Not applicable 134
Cryptotanshinone STAT3 SH2 domain Inhibits STAT3–STAT3 dimerization Polycystic ovary syndrome (phase not provided) 105
Honokiol EGFR, SHP1, STAT3 Decreases EGFR expression, increases SHP1 expression leading to inhibition of activated STAT3 Not applicable 131133
Berbamine JAK2, SRC, STAT3 Inhibits phosphorylation of STAT3, decreases level of STAT4, induces STAT6 activation Not applicable 145,146
Cinnamon bark STAT4 Inhibits phosphorylation of STAT4, decreases levels of IFN γ Polycystic ovary syndrome (Phase I), hypercholesterolaemia and type 2 diabetes (Phase II) 147
Indirubin JAK, STAT3 DNA-binding domain, STAT5 DNA-binding domain Inhibits phosphorylation of STAT3, inhibits phosphorylation of STAT5 Not applicable 89
TMC-264 STAT6 DNA-binding domain, Inhibits phosphorylation of STAT6, decreases IL-4 signal transduction Not applicable 154
Tyrosine kinase inhibitors
Tofacitinib JAK3 Inhibits phosphorylation of STAT1, STAT3, STAT4, STAT5 and STAT6 Rheumatoid arthritis (Phase I/II/III), juvenile idiopathic arthritis (Phase I/II/III), psoriasis (Phase I/II/III), ankylosing spondylitis (Phase II), keratoconjunctivitis sicca (Phase II), ulcerative colitis (Phase III) 183
Sorafenib JAK2, STAT3 Inhibits phosphorylation of STAT3 Hepatocellular carcinoma (Phase I/II/III/IV), head and neck squamous cell carcinoma (Phase I/II), gastric cancer (Phase I/II), breast cancer (Phase I/II/III), prostate cancer (Phase I/II), thyroid cancer (Phase II/ III), non-small cell lung cancer (Phase I/II/III), pancreatic cancer (Phase I/II/III), bladder cancer (Phase I/II), colorectal cancer (Phase I/II), kidney cancer (Phase I/II/III/IV), liver cancer (Phase I/II/III), glioblastoma multiforme (Phase I/II), leukaemia (Phase I/II/III), melanoma (Phase I/II/III) 169173
SC-1, SC-49 STAT3, SHP1 Increases SHP1 activity leading to inhibition of activated STAT3 Not applicable 174,175
AZD1480 JAK1, JAK2 Inhibits phosphorylation of STAT1, STAT3, STAT5 and STAT6 Hepatocellular carcinoma, lung carcinoma and gastric cancer (Phase I), essential thrombocythaemia myelofibrosis and post-polycythaemia vera (Phase I) 186189
Atiprimod JAK2 Inhibits phosphorylation of STAT3, inhibits phosphorylation of STAT5 Neuroendocrine carcinoma (Phase II), multiple myeloma (Phase I/II) 160,190,191
Auranofin JAK1 Inhibits phosphorylation of STAT3 Chronic lymphocytic leukaemia (Phase II), squamous cell lung cancer (Phase II), ovarian cancer (phase not provided) 192,193
DNA-binding modifier
Oligodeoxy-nucleotide decoy DNA-binding domain of STAT1, STAT3 and STAT5 Inhibits transcription activity of STAT1, STAT3 and STAT5 Head and neck cancer (Phase 0) 28,196203

EGFR, epidermal growth factor; gp130, glycoprotein 130; IFN, interferon; IL, interleukin; JAK, Janus kinase; pTyr, phosphotyrosine; STAT, signal transducer and activator of transcription; VEGFR2, vascular endothelial growth factor receptor 2.

Peptides and peptidomimetics

The STAT3 SH2 domain (FIG. 1) is required for promoting dimerization (FIG. 2). One of the limitations of targeting protein dimerization is the practicality of targeting the dimer interface, which is challenging owing to the planarity of the large surface area. Nevertheless, novel peptide inhibitors that target the pTyr-SH2 domain interaction were the first STAT3 inhibitors to be established23,30,5557 (FIG. 3a; TABLE 1).

A semi-rational, structure-based design approach identified the first SH2 domain-binding peptides and peptidomimetics that disrupt the STAT3 pTyr-SH2 domain interactions and STAT3–STAT3 dimerization21,30,55,58. The native, parent pTyr peptide, PY*LKTK (where Y* stands for pTyr) and its modified forms blocked the DNA-binding and transcriptional activities of STAT3 at high micromolar concentrations30. Peptidomimetic and non-peptide analogues, including ISS-610 and S3I-M2001, exhibited improved potency against STAT3 activity in vitro and against diverse malignant cells harbouring aberrantly active STAT3 (REFS 27,30,55,58). S31-M2001 inhibited the growth of human breast tumour xenografts, while ISS-610 inhibited cell growth and induced apoptosis in vitro27,55.

Furthermore, phosphopeptide binding sequences with the primary structure pTyr-Xxx-Xxx-Gln (where Xxx represents any amino acid) derived from leukaemia inhibitory factor (LIF), IL-10 receptor, epidermal growth factor receptor (EGFR), granulocyte colony-stimulating factor (GCSF) receptor or glycoprotein 130 (gp130), similarly inhibited STAT3 activation57,59. In these studies, the peptidomimetic Ac-pTyr-Leu-Pro-Gln-Thr-Val-NH2 was derived, which inhibited STAT3 activity (IC50 values of 150 nM)56,59. Moreover, a 28-mer native peptide identified as SPI, derived from the STAT3 SH2 domain, inhibited the STAT3 pTyr-SH2 domain interaction and signalling. The activity of SPI was moderate, but it did suppress cell viability and induced apoptosis of human breast, pancreatic, prostate and non-small cell lung cancer (NSCLC) cells in vitro60.

Other studies identified CJ-1383 as a SH2 domain-binding peptidomimetic inhibitor that induced anti-tumour cell effects against human breast cancer cells61, as well as a phosphatase-stable, cell-permeable SH2 domain-targeting phosphopeptide mimetic prodrug, PM-73G63. PM-73G potently inhibited both constitutive and IL-6-stimulated STAT3 phosphorylation in vitro in breast cancer cells62, and in in vivo tumour xenografts further inhibited VEGF expression, angiogenesis and tumour growth63. However, at low concentrations PM-73G did not inhibit the STAT3 target genes that encode cyclin D1, BCL-2 or survivin, or promote apoptosis62,63. When the concentration of PM-73G was increased 50-fold cell proliferation inhibition was observed62. The lack of effects on cell growth and survival and on the related target genes at the lowest concentration of PM-73G tested in these studies are in contrast to the large body of data on the inhibition of STAT3 activity by other modalities that resulted in tumour cell growth blockade and apoptosis. Together, these data suggest that the role of STAT3 and the impact of STAT3 inhibitors in tumour cell biology are more complex than currently known. Despite the prolific research activity into peptide inhibitors, metabolic instability, poor cell permeability and other peptide-associated liabilities have precluded their clinical development as therapeutics.

Small molecules

Many small-molecule inhibitors of STAT3 activity have been identified through computational modelling, docking studies and virtual screening of chemical libraries (TABLE 1). In most cases, these agents function as disruptors of STAT3–STAT3 dimerization (FIG. 3a). STA-21 (also known as NSC628869) was identified from the screening of the National Cancer Institute (NCI) chemical library as an inhibitor of STAT3 dimerization, DNA-binding activity and transcriptional function in breast cancer cells, with a potency of 20 M64. Its structural analogue, LLL-3 (which showed improved membrane permeability), decreased cell viability in vitro and intracranial tumour size in vivo in glioblastoma animal models65.

A catechol (1,2-dihydroxybenzene) compound was identified from Wyeth’s proprietary small-molecule collection as a STAT3 SH2 domain inhibitor, and this agent was active at 106 μM against a multiple myeloma cell line66. Furthermore, a structure-based, virtual screen of the NCI chemical library targeting the STAT3 SH2 domain discovered S3I-201 (also known as NSC74859) as a STAT3–STAT3 dimerization disruptor, with a potency of 60–110 μM26. S3I-201 inhibited STAT3 DNA-binding and transcriptional activities, induced growth inhibition and apoptosis of tumour cells harbouring constitutively active STAT3, and suppressed the growth of human breast cancer xenografts26. Several of its derivatives, including S3I-201.1066, BP-1-102 and S3I-1757, showed improved potencies, with IC50values of 35 μM (S3I-201.1066), 6.8 μM (BP-1-102) and 13.5 μM (S3I-1757), and inhibited cell growth, malignant transformation, survival, migration and invasiveness in vitro of malignant cells harbouring aberrantly active STAT3 (REFS 6769). In particular, S3I-201.1066 and BP-1-102 inhibited growth of human breast or NSCLC xenografts in mice, and notably BP-1-102 is orally bioavailable.

Virtual ligand screening identified Cpd30 (4-(5-((3-ethyl-4-oxo-2-thioxo-1,3-thiazolidin-5-ylidene) methyl)-2-furyl)benzoic acid), which selectively inhibited STAT3 with moderate potency, blocked STAT3 nuclear translocation upon IL-6 stimulation, and induced apoptosis in breast cancer cells harbouring constitutively active STAT3 (REF. 70). The related agent, Cpd188 (4-((3-((carboxymethyl)thio)-4-hydroxy-1-naphthyl)amino)sulphonyl)benzoic acid), in combination with docetaxel, decreased tumour growth in chemotherapy-resistant human breast cancer xenograft models71.

Stattic, a non-peptide small molecule discovered through virtual screening of chemical libraries, targets the STAT3 SH2 domain and inhibits STAT3 signalling at 10 μM72. It induced apoptosis of STAT3-dependent breast cancer and HNSCC cells and inhibited growth of orthotopic HNSCC tumour xenografts72. STX-0119, another STAT3 SH2 domain antagonist, induced antitumour cell effects in vitro and antitumour effects in vivo in a human lymphoma model73, possibly by disrupting STAT3–STAT3 dimerization74, with little effect on STAT3 phosphorylation73. Fragments of STX-0119 and stattic were chemically fused to generate HJC0123 (REF. 75), which suppressed STAT3 phosphorylation and transcriptional activity in breast cancer cells and induced antitumour cell effects against breast and pancreatic cancer cells in vitro at IC50 values of 0.1–1.25 μM. The oral administration of HJC0123 led to inhibition of growth of human breast cancer xenografts75.

Compound 6, which was identified through structure-based virtual screening, inhibited breast cancer cell viability in vitro, with an IC50 value of 20 μM, and decreased STAT3 phosphorylation and transcriptional activity in hepatocellular carcinoma (HCC) cells76. Furthermore, an agent identified as OBP-31121 was reported to inhibit STAT1, STAT3 and STAT5 phosphorylation. OBP-31121 induced loss of viability and apoptosis in vitro and inhibited tumour growth in vivo in gastric cancer models, and it further sensitized gastric cancer cells to cisplatin and 5-fluorouracil77. OBP-31121 has progressed to a Phase I clinical trial to determine the maximum tolerated dose (ClinicalTrials. gov identifier: NCT00955812).

Natural product inhibitors and their derivatives

Natural products have been an important resource in STAT3 inhibitor discovery and these efforts have yielded several lead candidates (TABLE 1). In many of these cases, however, the mechanism of action of these candidates with regard to STAT3 activity are unclear. It is possible that they inhibit STAT3 indirectly and are likely to block several targets.

Curcumin, a phenolic compound derived from the perennial herb Curcuma longa was shown to suppress JAK–STAT signalling at 15 μM, induce cell cycle arrest and inhibit cell invasion in vitro in a small cell lung cancer model78. In mice bearing gastric cancer xenografts, treatment with curcumin inhibited IL-6 production by IL-1β-stimulated myeloid-derived suppressor cells, which was associated with decreased activation of STAT3 and nuclear factor-κB (NF-κB)79.

Curcumin analogues with improved bioavailability and stability, such as FLLL32, had enhanced potency (IC50 values of 0.75–1.45 μM) in suppressing both pSTAT3 and total STAT3, and they also induced STAT3 ubiquitylation and possible proteasomal degradation in canine and human osteosarcoma cells in vitro80. Another curcumin analogue, HO-3867, similarly downregulated STAT3 signalling in cisplatin-resistant human ovarian cancer cells, thereby promoting enhanced sensitivity to cisplatin81. HO-3867 also induced apoptosis in BRCA1-mutated human ovarian cancer cells that harbour aberrantly active STAT3 (REF. 82).

Studies suggest that LLL12 (TABLE 1), another small-molecule inhibitor of STAT3 signalling that is based on curcumin83,84, might suppress STAT3 activation by blocking its recruitment to the receptor and thereby preventing phosphorylation by tyrosine kinases, and by interfering with dimerization85. LLL12 suppressed cell viability, induced apoptosis, and repressed colony formation and migration in vitro in studies of glioblastoma, osteosarcoma and breast cancer cells84,86. It also inhibited angiogenesis, tumour vasculature development, and tumour growth in vivo in osteosarcoma xenograft models84,85. Except for the finding that the analogues FLLL32 and FLLL62 may interact with the STAT3 SH2 domain87, questions remain regarding how curcumin or its analogues inhibit STAT3 signalling.

Melanoma cells harbouring aberrantly active STAT3 were shown to be responsive to BBMD3, a derivative of the natural bis-benzylsioquinoline alkaloid berbamine from Berberis amurensis, with an IC50 value of 2.9 μM. Melanoma cells treated with BBMD3 had decreased levels of pJAK2, pSRC and pSTAT3, and reduced cell viability88. In vitro kinase assays showed that BBMD3 directly inhibited the autophosphorylation of the mutant JAK2V617F (REF. 88).

The bis-indole alkaloid indirubin, which is derived from a mixture of plants used in the traditional Chinese medicine called Danggui Longhui Wan, was reported to inhibit VEGF receptor-mediated JAK–STAT3 signalling89. Indirubin regulated angiogenesis in both chick embryo chorioallantoic membrane and mice corneal micropocket assays89. Indirubin derivatives, including IRD E804 and MLS-2488, directly inhibited SRC activity in vitro (IC50 value of 0.43 μM), suppressed pJAK, pSTAT3 and pAKT activity, suppressed STAT3 DNA-binding activity, repressed myeloid cell leukaemia sequence 1 (MCL1) and survivin expression, and induced apoptosis in human breast cancer cells90,91. Recently, a more water-soluble indirubin derivative, E738, was identified that inhibits pJAKs (IC50 values of 10.4 nM (JAK1), 74.1 nM (JAK2) and 0.7 nM (TYK2)) and SRC kinases (IC50 value of 10.7 nM). Studies of E738 showed that it downregulated pJAK2, pSRC and pSTAT3 activity, and STAT3 transcriptional activity in pancreatic cancer cells at 1–2 μM92.

Resveratrol (3,5,4′-trihydroxystilbene), a polyphenolic compound found in red grapes and several other plants, was reported to inhibit STAT3 signalling at high micromolar concentrations, and it is likely that this effect contributes to the antitumour cell responses to this agent. Constitutive and IL-6-induced STAT3 activation in multiple myeloma, leukaemia and other tumour cell types were inhibited on resveratrol treatment, leading to a decreased expression of BCL-2 and other anti-apoptotic proteins, and apoptosis induction in vitro93. In leukaemia-bearing mice, treatment with resveratrol prolonged their life span and this was associated with decreased levels of pSTAT3 in liver tissue lysates94. The resveratrol derivative LYR71 suppressed breast cancer cell viability (IC50 value of 20 μM), and inhibited STAT3-mediated MMP9 expression95. Reports also showed that the inhibition of the JAK–STAT3 pathway by resveratrol or its analogue, piceatannol (3,3′,4,4′-transtrihydroxystilbene), decreased BCL-XL and BCL-2 expression and sensitized lung carcinoma, multiple myeloma, prostate and pancreatic cancer and the glioblastoma multiforme patient-derived CD133-positive cells to radiation or chemotherapy in vitro9698. A combination of resveratrol and 5-fluorouracil induced synergistic cytotoxicity in human colon cancer cells, which was accompanied by decreased STAT3 activity99. Despite these observations, the mechanisms for the inhibition of STAT3 signalling by resveratrol or its analogues remain poorly understood.

Caffeic acid, a natural phenolic acid compound present in fruits, wine and coffee100, and its synthetic derivative, CAPDE, were reported to inhibit tumour growth and angiogenesis in a renal carcinoma mouse xenograft model. Its effects are induced partly by suppressing active STAT3 and the expression of hypoxia-inducible factor 1α (HIF1α) and VEGF101. Another caffeic acid analogue, WP1193, inhibited STAT3 phosphorylation and blocked the growth of murine melanoma102 and human glioma xenograft tumours103. However, the mechanistic details of the inhibition of STAT3 phosphorylation remain undefined.

Treatment with 100 μM capsaicin (trans-8-methyl-N-vanillyl-6-nonenamide), which is present in chilli peppers, inhibited cell proliferation and induced apoptosis of multiple myeloma cells by inhibiting both constitutively active STAT3 and inducible STAT3 expression104. Apart from promoting gp130 depletion, the molecular mechanisms of how capsaicin modulates IL-6, gp130 and STAT3 signalling are not fully understood.

Studies also indicate that cryptotanshinone, the active component in the herbal plant Salvia miltiorrhiza Bunge (danshen), has a moderate inhibitory effect on STAT3 phosphorylation and its functions, possibly by binding to the STAT3 SH2 domain and interfering with dimerization, although this remains to be confirmed105. Prostate cancer cells treated with cryptotanshinone showed decreased expression of known STAT3-regulated genes, including those that encode cyclin D1, BCL-XL and survivin105.

Two agents that target microtubules have been reported to inhibit STAT3 phosphorylation in breast cancer cells: vinorelbine and paclitaxel106. Vinorelbine is a microtubule destabilizer and a semi-synthetic analogue of the vinca alkaloids isolated from the leaves of Catharanthus roseus. Paclitaxel is a microtubule stabilizer and was originally isolated from the bark of the yew tree. Both agents were reported to inhibit STAT3 phosphorylation in breast cancer cells; paclitaxel was shown to disrupt STAT3 interaction with tubulin106. Although these findings are interesting, the mechanistic basis for the inhibition of STAT3 phosphorylation by these two agents is unclear.

Drug-sensitive and drug-resistant multiple myeloma cells treated with 2.5 μM celastrol, a triterpene derived from the Chinese medicinal plant Tripterygium wilfordii, showed decreased expression of STAT3-regulated genes, diminished cell proliferation and enhanced sensitivity to chemotherapy107. Celastrol also inhibited growth of human HCC tumour xenografts in mice108. Although this agent blocked constitutive and inducible STAT3 activation and STAT3 nuclear translocation, the precise mechanism of action remains unknown.

Avicin D, a triterpenoid saponin that is present in the cactus plant Acacia victoriae, also decreased pSTAT3 activity and BCL-2 expression and induced apoptosis in both cutaneous T-cell lymphoma lines and CD4+ T cells isolated from patients with Sézary syndrome109. However, very little is known about how avicin D inhibits STAT3 phosphorylation.

Studies further show that withaferin A, a triterpenoid derived from Withania somnifera, inhibits constitutive and IL-6-induced STAT3 activation in human breast cancer cell lines at low micromolar concentration110. Withaferin A also decreased STAT3 and JAK2 protein levels and JAK2 activity in renal carcinoma Caki cells111. However, not much is understood of how withaferin suppresses the JAK–STAT3 signalling pathway.

The pentacyclic triterpene betulinic acid, which is isolated from the bark of the plant Ziziphus mauritiana, moderately inhibited both constitutive and inducible STAT3 activation, nuclear translocation and DNA-binding activity in multiple myeloma cells. It blocks JAK1, JAK2 and SRC activities and blocks the induction of the protein tyrosine phosphatase SHP1 (also known as PTPN6)112. The inhibition of STAT3 activity by betulinic acid also sensitized multiple myeloma cells to the apoptotic effects of bortezomib and thalidomide112.

Another pentacyclic triterpenoid, ursolic acid (3β-hydroxyurs-12-en-28-oic-acid), a natural dietary component found in many fruits, moderately suppressed both constitutive and inducible STAT3 activation in prostate cancer and other tumour cells by inhibiting SRC, JAK1 and JAK2 activities113,114. These effects led to decreased cyclin D1, BCL-2, survivin, MCL1 and VEGF expression113.

Oleanolic acid, found in Ganoderma lucidum and other plants, and its more potent derivative, CDDO-Me (active at 0.1 nM)115, inhibited the JAK–STAT3 pathway in multidrug-resistant ovarian cancer116 and in osteo-sarcoma cells117. CDDO-Me also induced antitumour responses in vivo in an aggressive oestrogen receptor-negative breast cancer model by modulating EGFR and STAT3 activities118. Details of the mechanism of inhibition of the EGFR–STAT3 or JAK–STAT3 signalling by CDDO-Me are limited.

Cucurbitacin agents isolated from Cucurbitaceae, Cruciferae (Brassicaceae) and other plant families have been identified as modulators of the JAK–STAT3 pathway. Cucurbitacin I (JSI-124) inhibited JAK–STAT3 signalling (IC50 value of 500 nM) and suppressed the growth in vivo of human and mouse tumours harbouring aberrantly active STAT3 (REF. 119). Treatment with cucurbitacin I blocked cell proliferation and enhanced the radiation sensitivity of CD133+ cancer stem cells isolated from patients with NSCLC120. Cucurbitacin I also suppressed growth of medulloblastoma121 and growth of thyroid cancer cells in vitro and in vivo in xenograft models122. Low micro-molar concentrations of cucurbitacin B in combination with cisplatin induced strong apoptotic effects in laryngeal squamous carcinoma cells by inhibiting the STAT3 pathway123. Reports also show that cucurbitacin E blocked VEGFR2-mediated JAK2–STAT3 activation in human umbilical vein endothelial cells and suppressed angiogenesis in human prostate tumour xenograft models124. This compound also induced viability loss and apoptosis of pancreatic125 and bladder cancer cells in vitro126 partly through inhibition of STAT3 phosphorylation.

Diosgenin, a steroidal saponin present in a number of plants, inhibited both constitutive and inducible STAT3 signalling in HCC cells, decreased cell proliferation, and induced chemosensitization by mechanisms that are unclear, but are likely to involve the induction of the protein tyrosine phosphatase SHPTP2, suppression of tyrosine kinases and/or blockade of STAT3 nuclear translocation127.

Studies in multiple myeloma cells show that emodin, which is derived from the root and rhizome of Rheum palmatum, moderately blocked JAK2 and STAT3 activation and inhibited cell viability128. Thymoquinone, the active ingredient from the volatile oil of black seed (Nigella sativa), blocked SRC and JAK2 activation, inhibited IL-6-induced and constitutive STAT3 activity, downregulated the expression of STAT3 target genes, and suppressed the viability of multiple myeloma cells with IC50 values of 8.5–10 μM129,130.

Furthermore, reports show that honokiol, isolated from the bark of Magnolia officinalis, moderately or weakly inhibited STAT3 activation in HNSCC, HCC and gastric cancer cells by undefined mechanisms, but probably involve the reduction of EGFR levels and the elevation of SHP1 expression131133.

Similarly, evodiamine, an alkaloid isolated from Evodia rutaecarpa, inhibited STAT3 signalling in HCC by blocking JAK activity and inducing SHP1 phosphatase134, and it induced antitumour responses in vivo in a HCC xenograft model134.

Carbazole, the active compound of coal tar and its N-alkyl derivatives, decreased IL-6-stimulated STAT3 activation and DNA-binding activity in embryonic kidney or human monocytic leukaemia cells through undefined mechanisms, without affecting phosphorylation of STAT3 (REFS 135,136).

A clinically used drug, sanguarine, a benzophenantridine alkaloid extracted primarily from the bloodroot plant, was recently shown to inhibit constitutive STAT3 activation through mechanisms that are currently unclear, but might contribute to its suppressive action on cell proliferation, migration and invasion of prostate tumour cells137.

A member of the vitamin E superfamily, γ-tocotrienol and acetyl-11-keto-β-boswellic acid, the active compound isolated from the Indian Boswellia serrate plant, both reportedly inhibited constitutive or inducible STAT3 signalling in HCC and multiple myeloma cells through the blockade of tyrosine kinases and the induction of SHP1 phosphatase activity138,139. These effects led to a decreased expression in cyclin D1, BCL-2, MCL1 and VEGF, inhibition of cell proliferation, and induction of apoptosis138140. Furthermore, 3,3′-diindolylmethane, an indole compound found in cruciferous vegetables, inhibited the JAK–STAT pathway through a yet undefined mechanism, which was partly responsible for the antitumour effects and the enhanced cisplatin sensitivity in an ovarian cancer model141. Brevilin A, a novel natural product isolated from Litsea glutinosa, inhibited the JAK JH1 (Janus homology 1) domain and attenuated STAT3 activation in prostate and breast cancer cells142.

Inhibitors of STAT1 and STAT4

Few inhibitors of the STAT1 or STAT4 signalling pathway have been reported in the literature (TABLE 1). Most STAT1 and STAT4 inhibitors are natural products except for pravastatin, a synthetic small-molecule inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, which reportedly modulated IFN γ activity and IFNγ-mediated STAT1-dependent events143,144. In studies of apolipoprotein E-deficient mice fed on a cholesterol-rich diet, pravastatin treatment suppressed IFN γ levels in both serum and atherosclerotic lesions, which was associated with decreased STAT1 activation, reduced IFN regulating factor 1 (IRF1) expression, and induced activity of SOCS1 in aorta tissue144. Although the mechanistic details are limited, these findings suggest a potential application of pravastatin in diseases in which STAT1 signalling is implicated144.

Studies also identified a series of phosphopeptide mimetics of STAT1, including ISS-840, which directly inhibited STAT1 signalling with moderate potency (IC50 value of 31 μM) through disruption of dimerization58. Furthermore, studies showed that an alkaloid isolated from Berberis vulgaris, berbamine, could modulate STAT1 signalling. In the EAE multiple sclerosis model, treatment with berbamine decreased the severity of the disease partly by suppressing IFN γ production and activity145.

Given the key role of STAT1 in the development of TH1 cells, which are implicated in the onset of EAE, compared with TH2 cells that promote recovery from the disease, the inhibition of IFNγ-dependent STAT1 activation could be used as a means to normalize the TH1–TH2 balance and promote recovery from the EAE phenotype145,146.

STAT4 protein levels also decreased in response to berbamine treatment, with little change in tyrosine (Y693) phosphorylation145. Studies also showed that the oral administration of water extract of cinnamon bark decreased IFNγ expression and inhibited STAT4 activation in activated murine T cells147.

The potential to modulate STAT1 or STAT4 and therefore modulation of T-cell functions suggests that berbamine and water extract of cinnamon bark could be used as therapeutics in inflammatory disorders147. Details of the mechanisms by which these two agents modulate IFN γ function and STAT1 or STAT4 signalling and their overall effects on the immune system, however, are unclear.

It is also reported that treatment with resveratrol (40–60 μM) inhibited both JAK1 phosphorylation and STAT1 activation in human epidermoid carcinoma cells148; the mechanism of action of resveratrol in this scenario is currently undefined.

Inhibitors of STAT5

Few STAT5 inhibitors have been reported. Pimozide, a diphenylbutylpiperidine derivative that is approved by the US Food and Drug Administration (FDA) as a neuroleptic drug, was shown to inhibit STAT5 activation with moderate activity41. In CML cells harbouring constitutively active STAT5, treatment with 5 μM pimozide suppressed pSTAT5 activity and induced apoptosis41. The mechanism of inhibition of STAT5 phosphorylation, however, is unclear but is suggested to be independent of dopamine D2 receptor activity41, and proceeds through the modulation of the negative regulators of STAT5 signalling45. Owing to its perceived inhibitory effect on STAT5 activity, it has been suggested that pimozide could be combined with TKIs, such as imatinib or dasatinib, as an approach to reduce the incidence of drug resistance in patients with CML41.

A non-peptidic chromone-based nicotinyl hydrazine, discovered through a screen of chemical libraries, was shown to weakly inhibit STAT5 activity (IC50 of 47 ± 17 μM)149. This agent selectively inhibited the phosphorylation of STAT5 in lymphoma cell lines by an undefined mechanism149. Inhibition of STAT5 activity is also reported for indirubin derivatives, including E804, which blocked pTyr-STAT5 and STAT5 DNA-binding activity in BCR–ABL-positive CML cells at 5 μM150. This effect was associated with the downregulation of MCL1 and BCL-XL expression 150. The mechanism for the inhibition of STAT5 signalling is likely to be suppression of tyrosine kinase activities.

Inhibitors of STAT6

Reports of inhibitors of STAT6 signalling are minimal (TABLE 1). The membrane-permeable version of the peptide STAT6BP, which was derived from the STAT6-binding region of IL-4Rα, inhibited IL-4-dependent STAT6 phosphorylation and transcriptional activity151. A recent report also showed that a chimeric STAT6-inhibitory peptide, STAT6-IP, blocked the TH2 cytokines IL-4 and IL-13, and chemokine production. The intranasal delivery of STAT6-IP inhibited the ovalbumin-induced lung inflammation, mucus production and eosinophil accumulation in the lungs152, while its topical application in an in vivo model of chronic ragweed-induced asthma suppressed the production of TH2 cytokines 153.

Treatment of the human B lymphoma cell line DND39 with TMC-264, an agent identified from the fermentation broth of Phoma spp., inhibited IL-4-dependent tyrosine phosphorylation and activation of STAT6 (REF. 154). At low micromolar concentrations (1.6 μM), TMC-264 selectively inhibited the phosphorylation of STAT6 over that of STAT5 or STAT1, and it also blocked STAT6 DNA-binding activity in a dose-dependent manner154. Its derivative, AS1517499, showed a significantly improved potency against STAT6 activity (IC50 of 21 nM) and inhibited IL-13-induced STAT6 phosphorylation51.

Other notable inhibitors of STAT6 signalling include YM-341619, a derivative of 4-aminopyrimidine-5-carboxamide (IC50 value of 0.70 nM)155, and AS1810722, a fused bicyclic pyrimidine derivative (IC50 value of 2.4 nM)156. Both AS1810722 and YM-341619 are potent, orally active STAT6 inhibitors that could be developed for the treatment of STAT6-dependent allergic diseases, such as asthma.

Leflunomide, an isoxazol derivative, N-(4-trifluoro-methylphenyl)-5-methylisoxazol-4-carboxamide, was reported to inhibit IL-13-induced STAT6 phosphorylation in human bronchial smooth muscle cells, but with a relatively weak potency (250 μM)50. The mechanisms are currently unknown and may include the inhibition of protein tyrosine kinases51.

Studies also indicate that the calcium-activated chloride channel blocker, niflumic acid, at 100 μM, inhibited JAK2 and STAT6 activation in the transformed human airway epithelial cell line BEAS-2B, and suppressed the pulmonary effects of IL-13 (REF. 157). While these findings suggest that niflumic acid could have potential in controlling IL-13-induced asthma, details of the mechanisms of action remain to be defined157.

Furthermore, berbamine reportedly inhibited STAT6 activation, as well as STAT1 and STAT4, implicating STAT6 in its pharmacological effect on TH2 cell development145. Finally, (R)-76 and its synthetic derivative, (R)-84, were identified through a chemical library screen and shown to bind and block STAT6 tyrosine phosphorylation, without competing at the phosphopeptide binding site of the SH2 domain158. These two small molecules also inhibited eotaxin-3 secretion in the bronchial epithelial cell line BEAS-2B158.

TKIs as modulators of STAT proteins

Tyrosine kinases have long been attractive targets for therapeutic development owing to their importance in promoting many cellular processes and hence, their involvement in human diseases. Occasions in which the abnormal activation of tyrosine kinases has been implicated in human diseases include the gain-of-function dominant mutation JAK2V617F identified in patients with myeloproliferative neoplasia159,160 and the BCR–ABL mutation that drives CML43. It is important to recognize that STAT activities could be involved in tyrosine kinase-associated diseases. Thus, the modulation of STAT function could be part of the underlying mechanisms for the therapeutic responses to TKIs, as long as dysregulated STAT signalling is evident downstream of the tyrosine kinase (FIG. 3b; TABLE 1). Several TKIs have been developed and approved for the treatment of various cancers, and these are predominantly small molecules that directly target the kinases or antibody-based therapeutics that compete for binding to the respective growth factor receptors on the cell surface. A few of these TKIs are presented here as examples.

Imatinib and dasatinib are small molecules that inhibit BCR–ABL, SRC and other tyrosine kinases and are approved for the treatment of CML161, gastrointestinal stromal tumour162, and other leukaemias including Philadelphia chromosome-positive acute lymphoblastic leukaemia, and myelodysplastic and myeloproliferative disorders163. Both imatinib and dasatinib inhibit STAT5 signalling164,165.

Similarly, gefitinib and erlotinib are small-molecule EGFR inhibitors approved for treating patients with advanced NSCLC with EGFR overexpression166 or for treating patients with metastatic NSCLC and pancreatic cancer. Both agents inhibit STAT signalling. Erlotinib moderately suppresses STAT3 expression levels and the formation of malignant lesions in a 4-nitroquinoline-1-oxide-induced mouse model of oral cancer 167. Furthermore, γ-tocotrienol, in combination with erlotinib or gefitinib, reportedly inhibited STAT5 signalling in mouse mammary tumour cells168.

The modulation of STAT3 signalling may be part of the underlying mechanisms for the survival benefits induced by the multiple kinase inhibitor sorafenib in patients with late-stage hepatocellular carcinoma (HCC) in Phase III clinical trials169,170. This conclusion is supported by the observation that HCC cells overexpressing mutant EGFR variant III (EGFRvIII) had a diminished response to sorafenib, whereas the combination of sorafenib and the EGFRvIII-specific monoclonal antibody CH12 enhanced the antitumour effect. The effect of sorafenib occurred partly by inhibiting multiple factors including STAT3, extracellular signal-regulated kinase (ERK), and phosphoinositide 3-kinase–AKT pathways171. Furthermore, sorafenib inhibited proliferation and induced apoptosis in glioblastoma cell lines and in primary cultures of cells from patients with grade IV glioblastoma, which were associated with the suppression of STAT3 signalling and the downregulation in expression of cyclin D, cyclin E and MCL1 proteins172. Moreover, treatment with sorafenib inhibited both JAK2 and STAT3 phosphorylation and the growth of human neuroblastoma tumours in a mouse xenograft model173. Studies of sorafenib analogues, including SC-1 and SC-49, further confirmed the inhibitory effects on STAT3 phosphorylation in HCC and liver adenocarcinoma in vitro and in tumour models in vivo174,175. Notably, the inhibitory action of the analogues against STAT3 phosphorylation was observed to be independent of their effects on kinases, and instead mediated through the induction of SHP1 phosphatase174,175. Specifically, SC-1 and SC-1 analogues that are structurally designed to preclude the binding to and the inhibition of kinases, were able to block STAT3 phosphorylation through the induction of SHP1 (REFS 174176).

Tetracyclic pyridone 2-tert-butyl-9-fluoro-3,6-dihy-dro-7H-benz[h]-imidaz[4,5-f]isoquinoline-7-one (P6), a reversible ATP inhibitor, decreased JAK2 phosphorylation and selectively inhibited the viability of murine and human myeloma cell lines harbouring activated STAT3 and JAKs177. Nifuroxazide, an oral nitrofuran antibiotic commonly used as an anti-diarrhoeal agent, was identified from the proprietary Prestwick chemical library, which was screened for agents that inhibit the activation of JAK2, TYK2 and STAT3 and decrease cell viability in multiple myeloma cells178.

Antibody-based tyrosine kinase-inhibiting therapeutics include cetuximab, which binds to the extracellular EGFR and is approved for certain types of HNSCC or colorectal cancers179. Cetuximab inhibited EGFR phosphorylation and the downstream signalling events, including STAT3 activation, in gastric and NSCLC cells180. Trastuzumab, which binds to EGFR2 (also known as HER2) and is approved to treat HER2-positive cancers, such as breast, gastric and gastroesophageal junction cancers181, inhibited the growth of gastric and breast cancer cell lines, decreased HER2 and STAT3 phosphorylation, and induced a synergistic response in combination with cisplatin182.

Several JAK inhibitors are also in clinical trials against various diseases, some of which are discussed here. Notable ones include the JAK inhibitor tofacitinib, which showed significant efficacy in rheumatoid arthritis and is FDA-approved for this indication183. Through the inhibition of JAKs, tofacitinib abrogated anti-CD3-induced IFNγ, IL-4 and IL-17 production in CD4+ T cells isolated from the peripheral blood of healthy volunteers183. Tofacitinib was also shown to inhibit STAT1, STAT3, STAT4, STAT5 and STAT6 activation in cultured anti-CD3-stimulated T cells, which suggests that targeting the cytokine signalling pathway with TKIs may be a consideration for immunosuppression183. Furthermore, the FDA-approved drug ruxolitinib, a potent inhibitor of JAK1 (IC50 value of 3.3 nM) and JAK2 (IC50 value 2.8 nM), blocked STAT3 and STAT5 activation in a human erythroleukemia cell line (HEL) expressing JAK2V617F (REF. 184). In a NSCLC model, this agent inhibited STAT3 activity without affecting cell proliferation and viability, and inhibited growth in soft agar and xenografts in vivo185. Adult patients with T-cell leukaemia are currently being recruited into a Phase II clinical trial (Clinical trials.gov identifier: NCT01712659) to examine the safety and effectiveness of this agent.

Additionally, the pyrazolyl pyrimidine AZD1480, which potently inhibits JAK1 (IC50 value of 1.3 nM) and JAK2 (IC50 value of 0.4 nM)186, differentially suppressed STAT1, STAT3, STAT5 and STAT6 activation at 0.1–5 μM concentration in prostate, ovarian, glioma, and breast cancer cells in vitro187,188. AZD1480 also inhibited prostate and ovarian tumour growth in vivo187, and, at the highest concentration (5 μM), inhibited the proliferation in vitro of Hodgkin’s lymphoma cells harbouring activated JAK by mechanisms that include effects on other targets186. Other studies showed that AZD1480 suppressed STAT3 activation in human and murine kidney carcinoma cell lines189 and in myeloid-derived suppressor cells in a murine renal carcinoma model, supporting the notion that targeting STAT3 could be used to control the tumour microenvironment189.

The cationic amphiphilic compound atiprimod (SK&F 106615) decreased JAK2 and pJAK2 levels in K562 cells190, inhibited STAT3 and STAT5 phosphorylation, and induced antitumour cell effects in multiple myeloma cell models191. In a model of essential thrombocythaemia harbouring an active JAK2 mutation, atiprimod induced potent antiproliferative and pro-apoptotic effects, which were associated with decreased STAT3 and STAT5 phosphorylation160. Moreover, the gold complex auranofin, which is currently undergoing Phase II clinical trials in chronic lymphocytic leukaemia (CLL) and ovarian and lung cancers (Clinical trials.gov identifiers: NCT01419691, NCT01747798 and NCT01737502, respectively), is reported to inhibit IL-6-induced phosphorylation of JAK1 and STAT3, leading to decreased MCL1 expression, caspase 3 activation, and the induction of apoptosis in multiple myeloma cells192,193. Although it inhibited JAK1 activity in in vitro kinase assays, the exact mechanism of auranofin-induced modulation of JAK–STAT3 signalling remains unclear192.

Notably, treatment with TKIs does not always lead to a suppressive response on STAT signalling. This may be partly due to the presence of compensatory mechanisms, whereby non-targeted tyrosine kinases can induce activation of STAT proteins. This is the case for lung cancer cells treated with the SRC family kinase inhibitor dasatinib, which showed no observable effect on STAT3 activation194, possibly due to the induction by JAKs and other tyrosine kinases195. Overall, STAT signalling modulation could be incorporated into studies that evaluate the underlying events for the responses to TKIs.

Conclusion and clinical perspectives

Although there is strong evidence supporting the role of STAT proteins in human diseases to justify the discovery of novel therapeutics against this family, there are no STAT-inhibiting modalities in clinical application, with the exception of TKIs. Even with the TKIs, their clinical applications are not specifically directed towards providing insights into the potential therapeutic benefits of targeting STAT proteins. It is only upon the availability of suitable small-molecule STAT inhibitors or other effective modalities as drugs that the clinical benefits of inhibiting STAT signalling can be realized.

With regard to STAT3, the public literature is full of reports of agents that modulate this pathway, although many of them have suboptimum potency and pharmacokinetic parameters, and/or poorly defined mechanisms. More recently reported small-molecule inhibitors of STAT3, including S3I-201.1066 and BP-1-102, have shown good in vitro activities and in vivo efficacies in preclinical models67,68, with the expectation that clinical candidates of direct STAT3 inhibitors could be forthcoming. Furthermore, it is particularly noteworthy that many of the reported inhibitors of STAT3 signalling have been identified from natural product sources, a pattern that parallels the trend that many of the existing drugs in clinical application trace certain aspects of their origin to natural products. Indeed, the large chemical diversity presented by natural products favours the chances of discovering active leads targeting the JAK–STAT signalling pathway. Yet, there are only isolated cases of research activity that currently tap into natural products as potential sources for discovering JAK–STAT inhibitors and these studies largely focus on targeting STAT3 signalling. These research efforts will need to be intensified to take full advantage of the potential that natural products offer in representing a promising source of drug leads.

So far, the only reported clinical trial of direct modulators of STAT signalling is the recent Phase 0 study of STAT3 ODN decoys for safety and pharmacodynamic monitoring28, the results of which are encouraging and indicate minimal toxicity in patients with HNSCC196. There is also a Phase I clinical trial that is presently recruiting patients with advanced cancers which are linked to STAT3, to evaluate the safety and clinical activity of the ISIS-STAT3Rx antisense oligonucleotide as a STAT3 inhibitor (ClinicalTrials.gov identifier: NCT01563302). Data from this trial should shed more light on the clinical and therapeutic significance of the inhibition of STAT3 function in cancer patients.

In the absence of clinical trials of direct small-molecule STAT inhibitors, the potential to safely modulate aberrant STAT signalling in human cancers can be evaluated based on the therapeutic use of TKIs. There are many TKIs that are in clinical application against human cancers, a few of which have already been discussed above. Other cases include an ongoing Phase II study in patients with CLL of dasatinib based on the strong in vitro cytotoxic effects observed following the pretreatment of primary CLL cells with this agent (ClinicalTrials. gov identifier: NCT01441882). Young patients with newly diagnosed glioma are also being recruited into a Phase I/II trial to determine the dose-limiting toxicity of erlotinib administered together with radiation therapy (ClinicalTrials.gov identifier: NCT00124657). The direct effect of the above-mentioned drugs on the activity of STAT3 or other STAT proteins is not one of the intended goals of these trials. However, the antitumour responses are likely to be a cumulative effect on their respective targets and against STAT (or STAT3) signalling, assuming the latter is aberrant as a consequence of the hyperactive tyrosine kinase that represents the target for the drug under evaluation.

Furthermore, a few natural products are presently undergoing clinical trials or have been approved for clinical applications that also modulate STAT signalling among others. The effect of curcumin on pancreatic cancer is being evaluated in a Phase II trial (ClinicalTrials.gov identifier: NCT00094445). The tolerability and pharmacodynamic properties of resveratrol are also being studied in patients with colorectal cancer (ClinicalTrials.gov identifier: NCT00433576). The effectiveness of 3,3′-diindolylmethane in the treatment of breast cancer is being studied in a Phase II/III study (ClinicalTrials.gov identifier: NCT01391689).

An ongoing pilot clinical study is also evaluating the tumour sensitivity of patients with melanoma to recombinant IFNα2b by determining the cellular levels and the activation status of STAT1 (ClinicalTrials.gov identifier: NCT01460875). There is also a Phase II study on the pharmacodynamic effects of the TKI AZD0530 on SRC and downstream STAT3 and STAT5 activation in patients with metastatic HNSCC (ClinicalTrials.gov identifier: NCT00513435), while a Phase I clinical trial will determine the maximum-tolerated dose and the pharmacokinetic profile of the multi-kinase inhibitor AT9283 in young patients with relapsed or refractory acute leukaemia, together with evaluating phosphorylated STAT5 as a means for monitoring the kinase inhibitory effects ex vivo and in vivo (ClinicalTrials. gov identifier: NCT01431664). Furthermore, an observational clinical study is ongoing to assess the activation status of STAT1, STAT3, STAT5A, STAT5B and STAT6 in leukocytes isolated from patients with rheumatoid arthritis treated with tocilizumab, a humanized IL-6R-specific monoclonal antibody (ClinicalTrials.gov identifier: NCT01633346).

The clinical benefits of exploiting dysregulated STAT signalling in human diseases remain to be harnessed, and the expectation is that clinical candidates of direct STAT-inhibitory modalities are forthcoming in the near future.

Key points.

  • Considerable evidence supports the crucial roles of the signal transducer and activator of transcription (STAT) family of proteins in human diseases, particularly in immune and inflammatory disorders, infection and cancer

  • Increasing emphasis is being placed on developing direct STAT inhibitors for clinical application, mainly through the discovery of small molecules, oligonucleotides and natural product derivatives.

  • A large part of the ongoing STATs drug discovery research for therapeutics is focused on targeting STAT3, of which the efforts to develop small-molecule STAT3 inhibitors is extensive. While research into oligodeoxynucleotide (ODN) decoys and antisense oligonucleotides (ASOs) as STAT-inhibitory approaches is not as widespread, these efforts appear to be advancing as a STAT3 ODN has progressed to clinical trials (Phase 0).

  • Tyrosine kinase inhibitors (TKIs) as therapeutic modalities are widely explored, and this approach is highly established. TKI agents may be therapeutic considerations in STAT-associated diseases in so far as a causal link could be established between the target tyrosine kinase and dysregulated STAT signalling that is prevalent in the disease.

  • The precedence of natural product-based therapeutics for many diseases and the number of reports on natural product inhibitors of STAT3 signalling together highlight the potential of this resource as an important source of leads for developing STAT inhibitors.

  • There is a clinical trial to evaluate ISIS-STAT3Rx ASO against advanced cancers. Other clinical trials are focusing on therapeutic modalities that can affect STAT function and STAT-associated diseases, including the evaluation of curcumin for pancreatic cancer, resveratrol for colorectal cancer and 3,3′-diindolylmethane for breast cancer, and TKIs against many cancer types.

Acknowledgments

The authors thank all their colleagues and members of their laboratory for the stimulating discussions related to this work. The authors also thank A. Chelsky for the art work for figure 3. This work was supported by grants from the National Cancer Institute (CA128865 and CA161931) and from the University of Hawaii to J.T.

Biographies

Gabriella Miklossy

Gabriella Miklossy obtained her Ph.D. degree in biomedical sciences at the University of Debrecen, Hungary. She was interested in designing dominant negative HIV-1 protease inhibitors and testing their activity using a novel high-throughput microtitre-plate fluorescent assay. She pursued her postdoctoral fellowship at the State University of New York, Upstate Medical University, Syracuse, New York, USA, where she studied the biochemistry of transaldolase and HRES1/RAB4 in the pathophysiology of systemic lupus erythematosus. She subsequently joined the laboratory of James Turkson at the University of Hawaii Cancer Center, Honolulu, USA, to pursue research on discovering natural product-based novel anticancer drugs targeting STAT3 signalling.

Tyvette S. Hilliard

Tyvette Hilliard recieved her Ph.D. degree in medicinal chemistry from the University of Illinois at Chicago, USA. During her pre-doctoral studies, she assisted in the development of a three-dimensional (3D) alginate organ culture system to study ovarian cancer. She investigated the role of the gonadotropins on normal ovarian surface epithelium (OSE) proliferation using the 3D system. This novel system allowed her to identify several oncogenic pathways activated in the OSE by the gonadotropins. She is presently a postdoctoral fellow in the Natural Products and Experimental Therapeutics Program in the laboratory of James Turkson pursuing the evaluation of marine natural products for novel therapeutics for cancers harbouring aberrant STAT3 signalling.

James Turkson

James Turkson obtained his Ph.D. degree in pharmacology from the University of Alberta, Edmonton, Alberta, Canada, where he studied the therapeutic significance of targeting protein kinase C to platelet and neutrophil functions and to nucleoside transport mechanisms in leukaemia. He pursued his postdoctoral fellowship training in molecular oncology with Richard Jove at the Moffitt Cancer Center (MCC), University of South Florida, Tampa, USA, where he studied the mechanisms of STAT3-mediated oncogenesis. He transitioned to join the faculty at MCC and pursued drug discovery research in collaboration with Richard Jove, Said Sebti and Andrew Hamilton focusing on targeting STAT3 for the discovery of small-molecule inhibitors as novel anti-cancer drugs. This research led to the identification of the first direct inhibitor of a STAT protein, which was a small peptide that disrupted STAT3 dimerization, signalling and functions. He then moved to the Burnett School of Biomedical Sciences at the University of Central Florida in Orlando, USA, and maintained a rigorous drug discovery research programme targeting STAT3. He subsequently relocated to the University of Hawaii Cancer Center, University of Hawaii, Honolulu, USA, where he is currently Professor and the Program Director of the Natural Products and Experimental Therapeutics Program. His current research interests include targeting STAT3 and other cancer-relevant signalling molecules for anticancer drug discovery, natural products, and drug resistance in ovarian and pancreatic cancers.

Footnotes

Competing interests statement

The authors declare no competing financial interests.

FURTHER INFORMATION

Clinicaltrials.gov: http://clinicaltrials.gov

ALL LINKS ARE ACTIVE IN THE ONLINE PDF

References

  • 1.Bromberg J, Darnell JE., Jr The role of STATs in transcriptional control and their impact on cellular function. Oncogene. 2000;19:2468–2473. doi: 10.1038/sj.onc.1203476. [DOI] [PubMed] [Google Scholar]
  • 2.Darnell JE., Jr STATs and gene regulation. Science. 1997;277:1630–1635. doi: 10.1126/science.277.5332.1630. [DOI] [PubMed] [Google Scholar]
  • 3.Stark GR, Darnell JE., Jr The JAK–STAT pathway at twenty. Immunity. 2012;36:503–514. doi: 10.1016/j.immuni.2012.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Adamkova L, Souckova K, Kovarik J. Transcription protein STAT1: biology and relation to cancer. Folia Biol. 2007;53:1–6. [PubMed] [Google Scholar]
  • 5.Szabo SJ, et al. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21:713–758. doi: 10.1146/annurev.immunol.21.120601.140942. [DOI] [PubMed] [Google Scholar]
  • 6.Meraz MA, et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK–STAT signaling pathway. Cell. 1996;84:431–442. doi: 10.1016/s0092-8674(00)81288-x. [DOI] [PubMed] [Google Scholar]
  • 7.Agrawal S, et al. Signal transducer and activator of transcription 1 is required for optimal foam cell formation and atherosclerotic lesion development. Circulation. 2007;115:2939–2947. doi: 10.1161/CIRCULATIONAHA.107.696922. [DOI] [PubMed] [Google Scholar]
  • 8.Liu L, et al. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J Exp Med. 2011;208:1635–1648. doi: 10.1084/jem.20110958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.van de Veerdonk FL, et al. STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. N Engl J Med. 2011;365:54–61. doi: 10.1056/NEJMoa1100102. [DOI] [PubMed] [Google Scholar]
  • 10.Gamero AM, et al. STAT2 contributes to promotion of colorectal and skin carcinogenesis. Cancer Prev Res. 2010;3:495–504. doi: 10.1158/1940-6207.CAPR-09-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Takeda K, et al. Targeted disruption of the mouse Stat3 gene leads to early embryonic lethality. Proc Natl Acad Sci USA. 1997;94:3801–3804. doi: 10.1073/pnas.94.8.3801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Akira S. Functional roles of STAT family proteins: lessons from knockout mice. Stem Cells. 1999;17:138–146. doi: 10.1002/stem.170138. A comprehensive review of the knowledge gained from STAT-knockout mice. [DOI] [PubMed] [Google Scholar]
  • 13.Takeda K, et al. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J Immunol. 1998;161:4652–4660. [PubMed] [Google Scholar]
  • 14.Sano S, et al. Keratinocyte-specific ablation of Stat3 exhibits impaired skin remodeling, but does not affect skin morphogenesis. EMBO J. 1999;18:4657–4668. doi: 10.1093/emboj/18.17.4657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Macias E, Rao D, Digiovanni J. Role of STAT3 in skin carcinogenesis: insights gained from relevant mouse models. J Skin Cancer. 2013;2013:684050. doi: 10.1155/2013/684050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bowman T, et al. STATs in oncogenesis. Oncogene. 2000;19:2474–2488. doi: 10.1038/sj.onc.1203527. [DOI] [PubMed] [Google Scholar]
  • 17.Darnell JE. Validating Stat3 in cancer therapy. Nature Med. 2005;11:595–596. doi: 10.1038/nm0605-595. [DOI] [PubMed] [Google Scholar]
  • 18.Jing N, Tweardy DJ. Targeting Stat3 in cancer therapy. Anticancer Drugs. 2005;16:601–607. doi: 10.1097/00001813-200507000-00002. [DOI] [PubMed] [Google Scholar]
  • 19.Yu H, Jove R. The STATs of cancer — new molecular targets come of age. Nature Rev Cancer. 2004;4:97–105. doi: 10.1038/nrc1275. [DOI] [PubMed] [Google Scholar]
  • 20.Buettner R, Mora LB, Jove R. Activated STAT signaling in human tumors provides novel molecular targets for therapeutic intervention. Clin Cancer Res. 2002;8:945–954. [PubMed] [Google Scholar]
  • 21.Turkson J, Jove R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene. 2000;19:6613–6626. doi: 10.1038/sj.onc.1204086. [DOI] [PubMed] [Google Scholar]
  • 22.Yue P, Turkson J. Targeting STAT3 in cancer: how successful are we? Expert Opin Investig Drugs. 2009;18:45–56. doi: 10.1517/13543780802565791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Turkson J. STAT proteins as novel targets for cancer drug discovery. Expert Opin Ther Targets. 2004;8:409–422. doi: 10.1517/14728222.8.5.409. [DOI] [PubMed] [Google Scholar]
  • 24.Kortylewski M, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nature Med. 2005;11:1314–1321. doi: 10.1038/nm1325. [DOI] [PubMed] [Google Scholar]
  • 25.Wang T, et al. Regulation of the innate and adaptive immune responses by Stat-3 signaling in tumor cells. Nature Med. 2004;10:48–54. doi: 10.1038/nm976. This paper proves that STAT3 signalling negatively influences the antitumour activity of tumour-infiltrating immune cells. [DOI] [PubMed] [Google Scholar]
  • 26.Siddiquee K, et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc Natl Acad Sci USA. 2007;104:7391–7396. doi: 10.1073/pnas.0609757104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Siddiquee KA, et al. An oxazole-based small-molecule Stat3 inhibitor modulates Stat3 stability and processing and induces antitumor cell effects. ACS Chem Biol. 2007;2:787–798. doi: 10.1021/cb7001973. [DOI] [PubMed] [Google Scholar]
  • 28.Sen M, et al. Lack of toxicity of a STAT3 decoy oligonucleotide. Cancer Chemother Pharmacol. 2009;63:983–995. doi: 10.1007/s00280-008-0823-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Niu G, et al. Gene therapy with dominant-negative Stat3 suppresses growth of the murine melanoma B16 tumor in vivo. Cancer Res. 1999;59:5059–5063. This study demonstrates for the first time that STAT3 is a valid anticancer therapeutic target, as a dominant-negative STAT3 variant significantly inhibits tumour growth in a melanoma xenograft mouse model. [PubMed] [Google Scholar]
  • 30.Turkson J, et al. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J Biol Chem. 2001;276:45443–45455. doi: 10.1074/jbc.M107527200. This study describes the first inhibitor of a STAT protein, which is a designed peptide that can successfully suppress STAT3 signalling. [DOI] [PubMed] [Google Scholar]
  • 31.Holland SM, et al. STAT3 mutations in the hyper-IgE syndrome. N Engl J Med. 2007;357:1608–1619. doi: 10.1056/NEJMoa073687. [DOI] [PubMed] [Google Scholar]
  • 32.Koskela HL, et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N Engl J Med. 2012;366:1905–1913. doi: 10.1056/NEJMoa1114885. This study identifies mutations within the STAT3 SH2 domain in patients with large granular lymphocytic leukaemia, causing STAT3 hyperactivation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mogensen TH, Jakobsen MA, Larsen CS. Identification of a novel STAT3 mutation in a patient with hyper-IgE syndrome. Scand J Infect Dis. 2013;45:235–238. doi: 10.3109/00365548.2012.715750. [DOI] [PubMed] [Google Scholar]
  • 34.Lovett-Racke AE, Yang Y, Racke MK. Th1 versus Th17: are T cell cytokines relevant in multiple sclerosis? Biochim Biophys Acta. 2011;1812:246–251. doi: 10.1016/j.bbadis.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chitnis T, et al. Effect of targeted disruption of STAT4 and STAT6 on the induction of experimental autoimmune encephalomyelitis. J Clin Invest. 2001;108:739–747. doi: 10.1172/JCI12563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Menke J, et al. Targeting transcription factor Stat4 uncovers a role for interleukin-18 in the pathogenesis of severe lupus nephritis in mice. Kidney Int. 2011;79:452–463. doi: 10.1038/ki.2010.438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lin JX, Leonard WJ. The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene. 2000;19:2566–2576. doi: 10.1038/sj.onc.1203523. [DOI] [PubMed] [Google Scholar]
  • 38.Buitenhuis M, et al. Signal transducer and activator of transcription 5a (STAT5a) is required for eosinophil differentiation of human cord blood-derived CD34+ cells. Blood. 2003;101:134–142. doi: 10.1182/blood-2002-03-0740. [DOI] [PubMed] [Google Scholar]
  • 39.Udy GB, et al. Requirement of STAT5b for sexual dimorphism of body growth rates and liver gene expression. Proc Natl Acad Sci USA. 1997;94:7239–7244. doi: 10.1073/pnas.94.14.7239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Imada K, et al. Stat5b is essential for natural killer cell-mediated proliferation and cytolytic activity. J Exp Med. 1998;188:2067–2074. doi: 10.1084/jem.188.11.2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Nelson EA, et al. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood. 2011;117:3421–3429. doi: 10.1182/blood-2009-11-255232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Cotarla I, et al. Stat5a is tyrosine phosphorylated and nuclear localized in a high proportion of human breast cancers. Int J Cancer. 2004;108:665–671. doi: 10.1002/ijc.11619. [DOI] [PubMed] [Google Scholar]
  • 43.Shuai K, et al. Constitutive activation of STAT5 by the BCR–ABL oncogene in chronic myelogenous leukemia. Oncogene. 1996;13:247–254. [PubMed] [Google Scholar]
  • 44.Benekli M, Baumann H, Wetzler M. Targeting signal transducer and activator of transcription signaling pathway in leukemias. J Clin Oncol. 2009;27:4422–4432. doi: 10.1200/JCO.2008.21.3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Nelson EA, et al. A chemical biology approach to developing STAT inhibitors: molecular strategies for accelerating clinical translation. Oncotarget. 2011;2:518–524. doi: 10.18632/oncotarget.296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hou J, et al. An interleukin-4-induced transcription factor: IL-4 Stat. Science. 1994;265:1701–1706. doi: 10.1126/science.8085155. [DOI] [PubMed] [Google Scholar]
  • 47.Sehra S, et al. IL-4 regulates skin homeostasis and the predisposition toward allergic skin inflammation. J Immunol. 2010;184:3186–3190. doi: 10.4049/jimmunol.0901860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chapoval SP, et al. STAT6 expression in multiple cell types mediates the cooperative development of allergic airway disease. J Immunol. 2011;186:2571–2583. doi: 10.4049/jimmunol.1002567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kuperman DA, Schleimer RP. Interleukin-4, interleukin-13, signal transducer and activator of transcription factor 6, and allergic asthma. Curr Mol Med. 2008;8:384–392. doi: 10.2174/156652408785161032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chiba Y, et al. Interleukin-13 augments bronchial smooth muscle contractility with an up-regulation of RhoA protein. Am J Respir Cell Mol Biol. 2009;40:159–167. doi: 10.1165/rcmb.2008-0162OC. [DOI] [PubMed] [Google Scholar]
  • 51.Chiba Y, et al. A novel STAT6 inhibitor AS1517499 ameliorates antigen-induced bronchial hypercontractility in mice. Am J Respir Cell Mol Biol. 2009;41:516–524. doi: 10.1165/rcmb.2008-0163OC. [DOI] [PubMed] [Google Scholar]
  • 52.Khaled WT, et al. The IL-4/IL-13/Stat6 signaling pathway promotes luminal mammary epithelial cell development. Development. 2007;134:2739–2750. doi: 10.1242/dev.003194. [DOI] [PubMed] [Google Scholar]
  • 53.O’Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med. 2013;368:161–170. doi: 10.1056/NEJMra1202117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Quintas-Cardama A, Verstovsek S. Molecular pathways: Jak/STAT pathway: mutations, inhibitors, and resistance. Clin Cancer Res. 2013;19:1933–1940. doi: 10.1158/1078-0432.CCR-12-0284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Turkson J, et al. Novel peptidomimetic inhibitors of signal transducer and activator of transcription 3 dimerization and biological activity. Mol Cancer Ther. 2004;3:261–269. [PubMed] [Google Scholar]
  • 56.Coleman DR, et al. Investigation of the binding determinants of phosphopeptides targeted to the SRC homology 2 domain of the signal transducer and activator of transcription 3. Development of a high-affinity peptide inhibitor. J Med Chem. 2005;48:6661–6670. doi: 10.1021/jm050513m. [DOI] [PubMed] [Google Scholar]
  • 57.Ren Z, et al. Identification of a high-affinity phosphopeptide inhibitor of Stat3. Bioorg Med Chem Lett. 2003;13:633–636. doi: 10.1016/s0960-894x(02)01050-8. [DOI] [PubMed] [Google Scholar]
  • 58.Gunning PT, et al. Isoform selective inhibition of STAT1 or STAT3 homo-dimerization via peptidomimetic probes: structural recognition of STAT SH2 domains. Bioorg Med Chem Lett. 2007;17:1875–1878. doi: 10.1016/j.bmcl.2007.01.077. [DOI] [PubMed] [Google Scholar]
  • 59.McMurray JS. Structural basis for the binding of high affinity phosphopeptides to Stat3. Biopolymers. 2008;90:69–79. doi: 10.1002/bip.20901. [DOI] [PubMed] [Google Scholar]
  • 60.Zhao W, Jaganathan S, Turkson J. A cell-permeable Stat3 SH2 domain mimetic inhibits Stat3 activation and induces antitumor cell effects in vitro. J Biol Chem. 2010;285:35855–35865. doi: 10.1074/jbc.M110.154088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chen J, et al. Structure-based design of conformationally constrained, cell-permeable STAT3 inhibitors. ACS Med Chem Lett. 2010;1:85–89. doi: 10.1021/ml100010j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mandal PK, et al. Potent and selective phosphopeptide mimetic prodrugs targeted to the Src homology 2 (SH2) domain of signal transducer and activator of transcription 3. J Med Chem. 2011;54:3549–3563. doi: 10.1021/jm2000882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Auzenne EJ, et al. A phosphopeptide mimetic prodrug targeting the SH2 domain of Stat3 inhibits tumor growth and angiogenesis. J Exp Ther Oncol. 2012;10:155–162. [PMC free article] [PubMed] [Google Scholar]
  • 64.Song H, et al. A low-molecular-weight compound discovered through virtual database screening inhibits Stat3 function in breast cancer cells. Proc Natl Acad Sci USA. 2005;102:4700–4705. doi: 10.1073/pnas.0409894102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Fuh B, et al. LLL-3 inhibits STAT3 activity, suppresses glioblastoma cell growth and prolongs survival in a mouse glioblastoma model. Br J Cancer. 2009;100:106–112. doi: 10.1038/sj.bjc.6604793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hao W, et al. Discovery of the catechol structural moiety as a Stat3 SH2 domain inhibitor by virtual screening. Bioorg Med Chem Lett. 2008;18:4988–4992. doi: 10.1016/j.bmcl.2008.08.032. [DOI] [PubMed] [Google Scholar]
  • 67.Zhang X, et al. A novel small-molecule disrupts Stat3 SH2 domain-phosphotyrosine interactions and Stat3-dependent tumor processes. Biochem Pharmacol. 2010;79:1398–1409. doi: 10.1016/j.bcp.2010.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zhang X, et al. Orally bioavailable small-molecule inhibitor of transcription factor Stat3 regresses human breast and lung cancer xenografts. Proc Natl Acad Sci USA. 2012;109:9623–9628. doi: 10.1073/pnas.1121606109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhang X, et al. A novel inhibitor of STAT3 homodimerization selectively suppresses STAT3 activity and malignant transformation. Cancer Res. 2013;73:1922–1933. doi: 10.1158/0008-5472.CAN-12-3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Xu X, et al. Chemical probes that competitively and selectively inhibit Stat3 activation. PLoS ONE. 2009;4:e4783. doi: 10.1371/journal.pone.0004783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dave B, et al. Selective small molecule Stat3 inhibitor reduces breast cancer tumor-initiating cells and improves recurrence free survival in a human-xenograft model. PLoS ONE. 2012;7:e30207. doi: 10.1371/journal.pone.0030207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schust J, et al. Stattic: a small-molecule inhibitor of STAT3 activation and dimerization. Chem Biol. 2006;13:1235–1242. doi: 10.1016/j.chembiol.2006.09.018. [DOI] [PubMed] [Google Scholar]
  • 73.Ashizawa T, et al. Antitumor activity of a novel small molecule STAT3 inhibitor against a human lymphoma cell line with high STAT3 activation. Int J Oncol. 2011;38:1245–1252. doi: 10.3892/ijo.2011.957. [DOI] [PubMed] [Google Scholar]
  • 74.Matsuno K, et al. Identification of a new series of STAT3 inhibitors by virtual screening. ACS Med Chem Lett. 2010;1:371–3755. doi: 10.1021/ml1000273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen H, et al. Fragment-based drug design and identification of HJC0123, a novel orally bioavailable STAT3 inhibitor for cancer therapy. Eur J Med Chem. 2013;62C:498–507. doi: 10.1016/j.ejmech.2013.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhang M, et al. Identification and characterization of small molecule inhibitors of signal transducer and activator of transcription 3 (STAT3) signaling pathway by virtual screening. Bioorg Med Chem Lett. 2013;23:2225–2229. doi: 10.1016/j.bmcl.2013.01.056. [DOI] [PubMed] [Google Scholar]
  • 77.Kim MJ, et al. OPB-31121, a novel small molecular inhibitor, disrupts the JAK2/STAT3 pathway and exhibits an antitumor activity in gastric cancer cells. Cancer Lett. 2013;335:145–152. doi: 10.1016/j.canlet.2013.02.010. [DOI] [PubMed] [Google Scholar]
  • 78.Yang CL, et al. Curcumin blocks small cell lung cancer cells migration, invasion, angiogenesis, cell cycle and neoplasia through Janus kinase–STAT3 signaling pathway. PLoS ONE. 2012;7:e37960. doi: 10.1371/journal.pone.0037960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tu SP, et al. Curcumin induces the differentiation of myeloid-derived suppressor cells and inhibits their interaction with cancer cells and related tumor growth. Cancer Prev Res. 2012;5:205–215. doi: 10.1158/1940-6207.CAPR-11-0247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Fossey SL, et al. The novel curcumin analog FLLL32 decreases STAT3 DNA binding activity and expression, and induces apoptosis in osteosarcoma cell lines. BMC Cancer. 2011;11:112. doi: 10.1186/1471-2407-11-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Selvendiran K, et al. HO-3867, a curcumin analog, sensitizes cisplatin-resistant ovarian carcinoma, leading to therapeutic synergy through STAT3 inhibition. Cancer Biol Ther. 2011;12:837–845. doi: 10.4161/cbt.12.9.17713. [DOI] [PubMed] [Google Scholar]
  • 82.Tierney BJ, et al. HO-3867, a STAT3 inhibitor induces apoptosis by inactivation of STAT3 activity in BRCA1-mutated ovarian cancer cells. Cancer Biol Ther. 2012;13:766–775. doi: 10.4161/cbt.20559. [DOI] [PubMed] [Google Scholar]
  • 83.Lin L, et al. Novel STAT3 phosphorylation inhibitors exhibit potent growth-suppressive activity in pancreatic and breast cancer cells. Cancer Res. 2010;70:2445–2454. doi: 10.1158/0008-5472.CAN-09-2468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Onimoe GI, et al. Small molecules, LLL12 and FLLL32, inhibit STAT3 and exhibit potent growth suppressive activity in osteosarcoma cells and tumor growth in mice. Invest New Drugs. 2012;30:916–926. doi: 10.1007/s10637-011-9645-1. [DOI] [PubMed] [Google Scholar]
  • 85.Bid HK, et al. Anti-angiogenic activity of a small molecule STAT3 inhibitor LLL12. PLoS ONE. 2012;7:e35513. doi: 10.1371/journal.pone.0035513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lin L, et al. A novel small molecule inhibits STAT3 phosphorylation and DNA binding activity and exhibits potent growth suppressive activity in human cancer cells. Mol Cancer. 2010;9:217. doi: 10.1186/1476-4598-9-217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bill MA, et al. Structurally modified curcumin analogs inhibit STAT3 phosphorylation and promote apoptosis of human renal cell carcinoma and melanoma cell lines. PLoS ONE. 2012;7:e40724. doi: 10.1371/journal.pone.0040724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Nam S, et al. Novel synthetic derivatives of the natural product berbamine inhibit Jak2/Stat3 signaling and induce apoptosis of human melanoma cells. Mol Oncol. 2012;6:484–493. doi: 10.1016/j.molonc.2012.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhang X, et al. Indirubin inhibits tumor growth by antitumor angiogenesis via blocking VEGFR2-mediated JAK/STAT3 signaling in endothelial cell. Int J Cancer. 2011;129:2502–2511. doi: 10.1002/ijc.25909. [DOI] [PubMed] [Google Scholar]
  • 90.Nam S, et al. Indirubin derivatives inhibit Stat3 signaling and induce apoptosis in human cancer cells. Proc Natl Acad Sci USA. 2005;102:5998–6003. doi: 10.1073/pnas.0409467102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liu L, et al. A novel 7-bromoindirubin with potent anticancer activity suppresses survival of human melanoma cells associated with inhibition of STAT3 and Akt signaling. Cancer Biol Ther. 2012;13:13. doi: 10.4161/cbt.21781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Nam S, et al. Dual inhibition of Janus and Src family kinases by novel indirubin derivative blocks constitutively-activated Stat3 signaling associated with apoptosis of human pancreatic cancer cells. Mol Oncol. 2013;7:369–378. doi: 10.1016/j.molonc.2012.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shakibaei M, Harikumar KB, Aggarwal BB. Resveratrol addiction: to die or not to die. Mol Nutr Food Res. 2009;53:115–128. doi: 10.1002/mnfr.200800148. [DOI] [PubMed] [Google Scholar]
  • 94.Li T, et al. Evaluation of anti-leukemia effect of resveratrol by modulating STAT3 signaling. Int Immunopharmacol. 2010;10:18–25. doi: 10.1016/j.intimp.2009.09.009. [DOI] [PubMed] [Google Scholar]
  • 95.Kim JE, et al. LYR71, a derivative of trimeric resveratrol, inhibits tumorigenesis by blocking STAT3-mediated matrix metalloproteinase 9 expression. Exp Mol Med. 2008;40:514–522. doi: 10.3858/emm.2008.40.5.514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yang YP, et al. Resveratrol suppresses tumorigenicity and enhances radiosensitivity in primary glioblastoma tumor initiating cells by inhibiting the STAT3 axis. J Cell Physiol. 2012;227:976–993. doi: 10.1002/jcp.22806. [DOI] [PubMed] [Google Scholar]
  • 97.Gupta SC, et al. Chemosensitization of tumors by resveratrol. Ann NY Acad Sci. 2011;1215:150–160. doi: 10.1111/j.1749-6632.2010.05852.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Piotrowska H, Kucinska M, Murias M. Biological activity of piceatannol: leaving the shadow of resveratrol. Mutat Res. 2012;750:60–82. doi: 10.1016/j.mrrev.2011.11.001. [DOI] [PubMed] [Google Scholar]
  • 99.Santandreu FM, et al. Resveratrol potentiates the cytotoxic oxidative stress induced by chemotherapy in human colon cancer cells. Cell Physiol Biochem. 2011;28:219–228. doi: 10.1159/000331733. [DOI] [PubMed] [Google Scholar]
  • 100.Lin CL, et al. Protective effect of caffeic acid on paclitaxel induced anti-proliferation and apoptosis of lung cancer cells involves NF-κB pathway. Int J Mol Sci. 2012;13:6236–6245. doi: 10.3390/ijms13056236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Jung JE, et al. Caffeic acid and its synthetic derivative CADPE suppress tumor angiogenesis by blocking STAT3-mediated VEGF expression in human renal carcinoma cells. Carcinogenesis. 2007;28:1780–1787. doi: 10.1093/carcin/bgm130. [DOI] [PubMed] [Google Scholar]
  • 102.Kong LY, et al. Inhibition of p-STAT3 enhances IFN-α efficacy against metastatic melanoma in a murine model. Clin Cancer Res. 2010;16:2550–2561. doi: 10.1158/1078-0432.CCR-10-0279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Sai K, et al. Induction of cell-cycle arrest and apoptosis in glioblastoma stem-like cells by WP1193, a novel small molecule inhibitor of the JAK2/STAT3 pathway. J Neurooncol. 2012;107:487–501. doi: 10.1007/s11060-011-0786-z. [DOI] [PubMed] [Google Scholar]
  • 104.Lee HK, et al. Capsaicin inhibits the IL-6/STAT3 pathway by depleting intracellular gp130 pools through endoplasmic reticulum stress. Biochem Biophys Res Commun. 2009;382:445–450. doi: 10.1016/j.bbrc.2009.03.046. [DOI] [PubMed] [Google Scholar]
  • 105.Shin DS, et al. Cryptotanshinone inhibits constitutive signal transducer and activator of transcription 3 function through blocking the dimerization in DU145 prostate cancer cells. Cancer Res. 2009;69:193–202. doi: 10.1158/0008-5472.CAN-08-2575. [DOI] [PubMed] [Google Scholar]
  • 106.Walker SR, et al. Microtubule-targeted chemotherapeutic agents inhibit signal transducer and activator of transcription 3 (STAT3) signaling. Mol Pharmacol. 2010;78:903–908. doi: 10.1124/mol.110.066316. [DOI] [PubMed] [Google Scholar]
  • 107.Kannaiyan R, et al. Celastrol inhibits proliferation and induces chemosensitization through down-regulation of NF-κB and STAT3 regulated gene products in multiple myeloma cells. Br J Pharmacol. 2011;164:1506–1521. doi: 10.1111/j.1476-5381.2011.01449.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rajendran P, et al. Celastrol suppresses growth and induces apoptosis of human hepatocellular carcinoma through the modulation of STAT3/JAK2 signaling cascade in vitro and in vivo. Cancer Prev Res. 2012;5:631–643. doi: 10.1158/1940-6207.CAPR-11-0420. [DOI] [PubMed] [Google Scholar]
  • 109.Zhang C, et al. Avicin D selectively induces apoptosis and downregulates p-STAT-3, BCL-2, and survivin in cutaneous T-cell lymphoma cells. J Invest Dermatol. 2008;128:2728–2735. doi: 10.1038/jid.2008.138. [DOI] [PubMed] [Google Scholar]
  • 110.Lee J, Hahm ER, Singh SV. Withaferin A inhibits activation of signal transducer and activator of transcription 3 in human breast cancer cells. Carcinogenesis. 2010;31:1991–1998. doi: 10.1093/carcin/bgq175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Um HJ, et al. Withaferin A inhibits JAK/STAT3 signaling and induces apoptosis of human renal carcinoma Caki cells. Biochem Biophys Res Commun. 2012;427:24–29. doi: 10.1016/j.bbrc.2012.08.133. [DOI] [PubMed] [Google Scholar]
  • 112.Pandey MK, Sung B, Aggarwal BB. Betulinic acid suppresses STAT3 activation pathway through induction of protein tyrosine phosphatase SHP-1 in human multiple myeloma cells. Int J Cancer. 2010;127:282–292. doi: 10.1002/ijc.25059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Pathak AK, et al. Ursolic acid inhibits STAT3 activation pathway leading to suppression of proliferation and chemosensitization of human multiple myeloma cells. Mol Cancer Res. 2007;5:943–955. doi: 10.1158/1541-7786.MCR-06-0348. [DOI] [PubMed] [Google Scholar]
  • 114.Shanmugam MK, et al. Ursolic acid inhibits multiple cell survival pathways leading to suppression of growth of prostate cancer xenograft in nude mice. J Mol Med. 2011;89:713–727. doi: 10.1007/s00109-011-0746-2. [DOI] [PubMed] [Google Scholar]
  • 115.Honda T, et al. Synthetic oleanane and ursane triterpenoids with modified rings A and C: a series of highly active inhibitors of nitric oxide production in mouse macrophages. J Med Chem. 2000;43:4233–4246. doi: 10.1021/jm0002230. [DOI] [PubMed] [Google Scholar]
  • 116.Duan Z, et al. CDDO-Me, a synthetic triterpenoid, inhibits expression of IL-6 and Stat3 phosphorylation in multi-drug resistant ovarian cancer cells. Cancer Chemother Pharmacol. 2009;63:681–689. doi: 10.1007/s00280-008-0785-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Ryu K, et al. Oleanane triterpenoid CDDO-Me induces apoptosis in multidrug resistant osteosarcoma cells through inhibition of Stat3 pathway. BMC Cancer. 2010;10:187. doi: 10.1186/1471-2407-10-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tran K, et al. The synthetic triterpenoid CDDO-methyl ester delays estrogen receptor-negative mammary carcinogenesis in polyoma middle T mice. Cancer Prev Res. 2012;5:726–734. doi: 10.1158/1940-6207.CAPR-11-0404. [DOI] [PubMed] [Google Scholar]
  • 119.Blaskovich MA, et al. Discovery of JSI-124 (cucurbitacin I), a selective Janus kinase/signal transducer and activator of transcription 3 signaling pathway inhibitor with potent antitumor activity against human and murine cancer cells in mice. Cancer Res. 2003;63:1270–1279. [PubMed] [Google Scholar]
  • 120.Hsu HS, et al. Cucurbitacin I inhibits tumorigenic ability and enhances radiochemosensitivity in nonsmall cell lung cancer-derived CD133-positive cells. Cancer. 2011;117:2970–2985. doi: 10.1002/cncr.25869. [DOI] [PubMed] [Google Scholar]
  • 121.Chang CJ, et al. Inhibition of phosphorylated STAT3 by cucurbitacin I enhances chemoradiosensitivity in medulloblastoma-derived cancer stem cells. Childs Nerv Syst. 2012;28:363–373. doi: 10.1007/s00381-011-1672-x. [DOI] [PubMed] [Google Scholar]
  • 122.Tseng LM, et al. Targeting signal transducer and activator of transcription 3 pathway by cucurbitacin I diminishes self-renewing and radiochemoresistant abilities in thyroid cancer-derived CD133+ cells. J Pharmacol Exp Ther. 2012;341:410–423. doi: 10.1124/jpet.111.188730. [DOI] [PubMed] [Google Scholar]
  • 123.Liu T, et al. Cucurbitacin B, a small molecule inhibitor of the Stat3 signaling pathway, enhances the chemosensitivity of laryngeal squamous cell carcinoma cells to cisplatin. Eur J Pharmacol. 2010;641:15–22. doi: 10.1016/j.ejphar.2010.04.062. [DOI] [PubMed] [Google Scholar]
  • 124.Dong Y, et al. Cucurbitacin E, a tetracyclic triterpenes compound from Chinese medicine, inhibits tumor angiogenesis through VEGFR2-mediated Jak2–STAT3 signaling pathway. Carcinogenesis. 2010;31:2097–2104. doi: 10.1093/carcin/bgq167. [DOI] [PubMed] [Google Scholar]
  • 125.Sun C, et al. Inhibitory effect of cucurbitacin E on pancreatic cancer cells growth via STAT3 signaling. J Cancer Res Clin Oncol. 2010;136:603–610. doi: 10.1007/s00432-009-0698-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Huang WW, et al. Cucurbitacin E induces G(2)/M phase arrest through STAT3/p53/p21 signaling and provokes apoptosis via Fas/CD95 and mitochondria-dependent pathways in human bladder cancer T24 cells. Evid Based Complement Alternat Med. 2012;2012:952762. doi: 10.1155/2012/952762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Li F, et al. Diosgenin, a steroidal saponin, inhibits STAT3 signaling pathway leading to suppression of proliferation and chemosensitization of human hepatocellular carcinoma cells. Cancer Lett. 2010;292:197–207. doi: 10.1016/j.canlet.2009.12.003. [DOI] [PubMed] [Google Scholar]
  • 128.Muto A, et al. Emodin has a cytotoxic activity against human multiple myeloma as a Janus-activated kinase 2 inhibitor. Mol Cancer Ther. 2007;6:987–994. doi: 10.1158/1535-7163.MCT-06-0605. [DOI] [PubMed] [Google Scholar]
  • 129.Badr G, Mohany M, Abu-Tarboush F. Thymoquinone decreases F-actin polymerization and the proliferation of human multiple myeloma cells by suppressing STAT3 phosphorylation and Bcl2/Bcl-XL expression. Lipids Health Dis. 2011;10:236. doi: 10.1186/1476-511X-10-236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Li F, Rajendran P, Sethi G. Thymoquinone inhibits proliferation, induces apoptosis and chemosensitizes human multiple myeloma cells through suppression of signal transducer and activator of transcription 3 activation pathway. Br J Pharmacol. 2010;161:541–554. doi: 10.1111/j.1476-5381.2010.00874.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Leeman-Neill RJ, et al. Honokiol inhibits epidermal growth factor receptor signaling and enhances the antitumor effects of epidermal growth factor receptor inhibitors. Clin Cancer Res. 2010;16:2571–2579. doi: 10.1158/1078-0432.CCR-10-0333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Rajendran P, et al. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J Cell Physiol. 2012;227:2184–2195. doi: 10.1002/jcp.22954. [DOI] [PubMed] [Google Scholar]
  • 133.Liu SH, et al. Calpain/SHP-1 interaction by honokiol dampening peritoneal dissemination of gastric cancer in nu/nu mice. PLoS ONE. 2012;7:e43711. doi: 10.1371/journal.pone.0043711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Yang J, et al. Evodiamine inhibits STAT3 signaling by inducing phosphatase shatterproof 1 in hepatocellular carcinoma cells. Cancer Lett. 2013;325:243–251. doi: 10.1016/j.canlet.2012.09.019. [DOI] [PubMed] [Google Scholar]
  • 135.Arbiser JL, et al. Carbazole is a naturally occurring inhibitor of angiogenesis and inflammation isolated from antipsoriatic coal tar. J Invest Dermatol. 2006;126:1396–1402. doi: 10.1038/sj.jid.5700276. [DOI] [PubMed] [Google Scholar]
  • 136.Saturnino C, et al. Synthesis and biological evaluation of new N-alkylcarbazole derivatives as STAT3 inhibitors: preliminary study. Eur J Med Chem. 2012;60C:112–119. doi: 10.1016/j.ejmech.2012.11.004. [DOI] [PubMed] [Google Scholar]
  • 137.Sun M, et al. Inhibition of Stat3 activation by sanguinarine suppresses prostate cancer cell growth and invasion. Prostate. 2012;72:82–89. doi: 10.1002/pros.21409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kannappan R, Yadav VR, Aggarwal BB. γ-Tocotrienol but not γ-tocopherol blocks STAT3 cell signaling pathway through induction of protein-tyrosine phosphatase SHP-1 and sensitizes tumor cells to chemotherapeutic agents. J Biol Chem. 2010;285:33520–33528. doi: 10.1074/jbc.M110.158378. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 139.Rajendran P, et al. γ-Tocotrienol is a novel inhibitor of constitutive and inducible STAT3 signaling pathway in human hepatocellular carcinoma: potential role as an antiproliferative, pro-apoptotic and chemosensitizing agent. Br J Pharmacol. 2011;163:283–298. doi: 10.1111/j.1476-5381.2010.01187.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kunnumakkara AB, et al. Boswellic acid blocks signal transducers and activators of transcription 3 signaling, proliferation, and survival of multiple myeloma via the protein tyrosine phosphatase SHP-1. Mol Cancer Res. 2009;7:118–128. doi: 10.1158/1541-7786.MCR-08-0154. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 141.Kandala PK, Srivastava SK. Regulation of Janus-activated kinase-2 (JAK2) by diindolylmethane in ovarian cancer in vitro and in vivo. Drug Discov Ther. 2012;6:94–101. [PubMed] [Google Scholar]
  • 142.Chen X, et al. Brevilin A, a novel natural product, inhibits Janus kinase activity and blocks STAT3 signaling in cancer cells. PLoS ONE. 2013;8:e63697. doi: 10.1371/journal.pone.0063697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Leon ML, Zuckerman SH. Gamma interferon: a central mediator in atherosclerosis. Inflamm Res. 2005;54:395–411. doi: 10.1007/s00011-005-1377-2. [DOI] [PubMed] [Google Scholar]
  • 144.Zhou XX, Gao PJ, Sun BG. Pravastatin attenuates interferon-γ action via modulation of STAT1 to prevent aortic atherosclerosis in apolipoprotein E-knockout mice. Clin Exp Pharmacol Physiol. 2009;36:373–379. doi: 10.1111/j.1440-1681.2008.05067.x. [DOI] [PubMed] [Google Scholar]
  • 145.Ren Y, et al. Novel immunomodulatory properties of berbamine through selective down-regulation of STAT4 and action of IFN-γ in experimental autoimmune encephalomyelitis. J Immunol. 2008;181:1491–1498. doi: 10.4049/jimmunol.181.2.1491. [DOI] [PubMed] [Google Scholar]
  • 146.Nagelkerken L. Role of Th1 and Th2 cells in autoimmune demyelinating disease. Braz J Med Biol Res. 1998;31:55–60. doi: 10.1590/s0100-879x1998000100007. [DOI] [PubMed] [Google Scholar]
  • 147.Lee BJ, et al. Immunomodulatory effect of water extract of cinnamon on anti-CD3-induced cytokine responses and p38, JNK, ERK1/2, and STAT4 activation. Immunopharmacol Immunotoxicol. 2011;33:714–722. doi: 10.3109/08923973.2011.564185. [DOI] [PubMed] [Google Scholar]
  • 148.Madan E, et al. Regulation of apoptosis by resveratrol through JAK/STAT and mitochondria mediated pathway in human epidermoid carcinoma A431 cells. Biochem Biophys Res Commun. 2008;377:1232–1237. doi: 10.1016/j.bbrc.2008.10.158. [DOI] [PubMed] [Google Scholar]
  • 149.Muller J, et al. Discovery of chromone-based inhibitors of the transcription factor STAT5. Chembiochem. 2008;9:723–727. doi: 10.1002/cbic.200700701. [DOI] [PubMed] [Google Scholar]
  • 150.Nam S, et al. Indirubin derivatives induce apoptosis of chronic myelogenous leukemia cells involving inhibition of Stat5 signaling. Mol Oncol. 2012;6:276–283. doi: 10.1016/j.molonc.2012.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Stolzenberger S, Haake M, Duschl A. Specific inhibition of interleukin-4-dependent Stat6 activation by an intracellularly delivered peptide. Eur J Biochem. 2001;268:4809–4814. doi: 10.1046/j.1432-1327.2001.02417.x. [DOI] [PubMed] [Google Scholar]
  • 152.McCusker CT, et al. Inhibition of experimental allergic airways disease by local application of a cell-penetrating dominant-negative STAT-6 peptide. J Immunol. 2007;179:2556–2564. doi: 10.4049/jimmunol.179.4.2556. [DOI] [PubMed] [Google Scholar]
  • 153.Wang Y, et al. Effective treatment of experimental ragweed-induced asthma with STAT-6-IP, a topically delivered cell-penetrating peptide. Clin Exp Allergy. 2011;41:1622–1630. doi: 10.1111/j.1365-2222.2011.03853.x. [DOI] [PubMed] [Google Scholar]
  • 154.Sakurai M, et al. TMC-264, a novel inhibitor of STAT6 activation produced by Phoma sp TC 1674. J Antibiot. 2003;56:513–519. doi: 10.7164/antibiotics.56.513. [DOI] [PubMed] [Google Scholar]
  • 155.Nagashima S, et al. Identification of 4-benzylamino-2-[(4-morpholin-4-ylphenyl)amino]pyrimidine-5-carboxamide derivatives as potent and orally bioavailable STAT6 inhibitors. Bioorg Med Chem. 2008;16:6509–6521. doi: 10.1016/j.bmc.2008.05.031. [DOI] [PubMed] [Google Scholar]
  • 156.Nagashima S, et al. Novel 7H-pyrrolo[2,3-d]pyrimidine derivatives as potent and orally active STAT6 inhibitors. Bioorg Med Chem. 2009;17:6926–6936. doi: 10.1016/j.bmc.2009.08.021. [DOI] [PubMed] [Google Scholar]
  • 157.Nakano T, et al. Niflumic acid suppresses interleukin-13-induced asthma phenotypes. Am J Respir Crit Care Med. 2006;173:1216–1221. doi: 10.1164/rccm.200410-1420OC. [DOI] [PubMed] [Google Scholar]
  • 158.Zhou L, et al. STAT6 phosphorylation inhibitors block eotaxin-3 secretion in bronchial epithelial cells. Bioorg Med Chem. 2012;20:750–758. doi: 10.1016/j.bmc.2011.12.006. [DOI] [PubMed] [Google Scholar]
  • 159.Manshouri T, et al. Bone marrow stroma-secreted cytokines protect JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor. Cancer Res. 2011;71:3831–3840. doi: 10.1158/0008-5472.CAN-10-4002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Quintas-Cardama A, et al. Preclinical characterization of atiprimod, a novel JAK2 AND JAK3 inhibitor. Invest New Drugs. 2011;29:818–826. doi: 10.1007/s10637-010-9429-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Kantarjian H, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010;362:2260–2270. doi: 10.1056/NEJMoa1002315. [DOI] [PubMed] [Google Scholar]
  • 162.Duensing A, et al. Mechanisms of oncogenic KIT signal transduction in primary gastrointestinal stromal tumors (GISTs) Oncogene. 2004;23:3999–4006. doi: 10.1038/sj.onc.1207525. [DOI] [PubMed] [Google Scholar]
  • 163.Ravandi F, et al. First report of phase 2 study of dasatinib with hyper-CVAD for the frontline treatment of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia. Blood. 2010;116:2070–2077. doi: 10.1182/blood-2009-12-261586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Nam S, et al. Dasatinib (BMS-354825) inhibits Stat5 signaling associated with apoptosis in chronic myelogenous leukemia cells. Mol Cancer Ther. 2007;6:1400–1405. doi: 10.1158/1535-7163.MCT-06-0446. [DOI] [PubMed] [Google Scholar]
  • 165.Quentmeier H, et al. BCR–ABL1-independent PI3Kinase activation causing imatinib-resistance. J Hematol Oncol. 2011;4:6. doi: 10.1186/1756-8722-4-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Pallis AG, Syrigos KN. Epidermal growth factor receptor tyrosine kinase inhibitors in the treatment of NSCLC. Lung Cancer. 2013;80:120–130. doi: 10.1016/j.lungcan.2012.12.025. [DOI] [PubMed] [Google Scholar]
  • 167.Leeman-Neill RJ, et al. Inhibition of EGFR–STAT3 signaling with erlotinib prevents carcinogenesis in a chemically-induced mouse model of oral squamous cell carcinoma. Cancer Prev Res (Phila) 2011;4:230–237. doi: 10.1158/1940-6207.CAPR-10-0249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Bachawal SV, Wali VB, Sylvester PW. Combined γ-tocotrienol and erlotinib/gefitinib treatment suppresses Stat and Akt signaling in murine mammary tumor cells. Anticancer Res. 2010;30:429–437. [PubMed] [Google Scholar]
  • 169.Cheng AL, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:25–34. doi: 10.1016/S1470-2045(08)70285-7. [DOI] [PubMed] [Google Scholar]
  • 170.Llovet JM, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
  • 171.Yang Y, et al. The monoclonal antibody CH12 enhances the sorafenib-mediated growth inhibition of hepatocellular carcinoma xenografts expressing epidermal growth factor receptor variant III. Neoplasia. 2012;14:509–518. doi: 10.1593/neo.12328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Yang F, et al. Sorafenib induces growth arrest and apoptosis of human glioblastoma cells through the dephosphorylation of signal transducers and activators of transcription 3. Mol Cancer Ther. 2010;9:953–962. doi: 10.1158/1535-7163.MCT-09-0947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Yang F, et al. Sorafenib inhibits endogenous and IL-6/ S1P induced JAK2–STAT3 signaling in human neuroblastoma, associated with growth suppression and apoptosis. Cancer Biol Ther. 2012;13:534–541. doi: 10.4161/cbt.19603. [DOI] [PubMed] [Google Scholar]
  • 174.Tai WT, et al. Signal transducer and activator of transcription 3 is a major kinase-independent target of sorafenib in hepatocellular carcinoma. J Hepatol. 2011;55:1041–1048. doi: 10.1016/j.jhep.2011.01.047. [DOI] [PubMed] [Google Scholar]
  • 175.Chen KF, et al. Sorafenib and its derivative SC-49 sensitize hepatocellular carcinoma cells to CS-1008, a humanized anti-TNFRSF10B (DR5) antibody. Br J Pharmacol. 2013;168:658–672. doi: 10.1111/j.1476-5381.2012.02212.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Chen KF, et al. Blockade of STAT3 activation by sorafenib derivatives through enhancing SHP-1 phosphatase activity. Eur J Med Chem. 2012;55:220–227. doi: 10.1016/j.ejmech.2012.07.023. [DOI] [PubMed] [Google Scholar]
  • 177.Pedranzini L, et al. Pyridone 6, a pan-Janus-activated kinase inhibitor, induces growth inhibition of multiple myeloma cells. Cancer Res. 2006;66:9714–9721. doi: 10.1158/0008-5472.CAN-05-4280. [DOI] [PubMed] [Google Scholar]
  • 178.Nelson EA, et al. Nifuroxazide inhibits survival of multiple myeloma cells by directly inhibiting STAT3. Blood. 2008;112:5095–5102. doi: 10.1182/blood-2007-12-129718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Muhammad S, et al. The role of EGFR monoclonal antibodies (MoABs) cetuximab/panitumab, and BRAF inhibitors in BRAF mutated colorectal cancer. J Gastrointest Oncol. 2013;4:72–81. doi: 10.3978/j.issn.2078-6891.2012.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Patel D, et al. Anti-epidermal growth factor receptor monoclonal antibody cetuximab inhibits EGFR/HER-2 heterodimerization and activation. Int J Oncol. 2009;34:25–32. [PubMed] [Google Scholar]
  • 181.Gravalos C, et al. Phase II study of trastuzumab and cisplatin as first-line therapy in patients with HER2-positive advanced gastric or gastroesophageal junction cancer. Clin Transl Oncol. 2011;13:179–184. doi: 10.1007/s12094-011-0637-6. [DOI] [PubMed] [Google Scholar]
  • 182.Kim SY, et al. Trastuzumab inhibits the growth of human gastric cancer cell lines with HER2 amplification synergistically with cisplatin. Int J Oncol. 2008;32:89–95. [PubMed] [Google Scholar]
  • 183.Migita K, et al. Inhibitory effects of the JAK inhibitor CP690,550 on human CD4+ T lymphocyte cytokine production. BMC Immunol. 2011;12:51. doi: 10.1186/1471-2172-12-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Quintas-Cardama A, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109–3117. doi: 10.1182/blood-2009-04-214957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Looyenga BD, et al. STAT3 is activated by JAK2 independent of key oncogenic driver mutations in non-small cell lung carcinoma. PLoS ONE. 2012;7:e30820. doi: 10.1371/journal.pone.0030820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Derenzini E, et al. The JAK inhibitor AZD1480 regulates proliferation and immunity in Hodgkin lymphoma. Blood Cancer J. 2011;1:e46. doi: 10.1038/bcj.2011.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Hedvat M, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell. 2009;16:487–497. doi: 10.1016/j.ccr.2009.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.McFarland BC, et al. Therapeutic potential of AZD1480 for the treatment of human glioblastoma. Mol Cancer Ther. 2011;10:2384–2393. doi: 10.1158/1535-7163.MCT-11-0480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Xin H, et al. Antiangiogenic and antimetastatic activity of JAK inhibitor AZD1480. Cancer Res. 2011;71:6601–6610. doi: 10.1158/0008-5472.CAN-11-1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Faderl S, et al. Atiprimod blocks phosphorylation of JAK–STAT and inhibits proliferation of acute myeloid leukemia (AML) cells. Leuk Res. 2007;31:91–95. doi: 10.1016/j.leukres.2006.05.027. [DOI] [PubMed] [Google Scholar]
  • 191.Amit-Vazina M, et al. Atiprimod blocks STAT3 phosphorylation and induces apoptosis in multiple myeloma cells. Br J Cancer. 2005;93:70–80. doi: 10.1038/sj.bjc.6602637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Kim NH, et al. Auranofin blocks interleukin-6 signaling by inhibiting phosphorylation of JAK1 and STAT3. Immunology. 2007;122:607–614. doi: 10.1111/j.1365-2567.2007.02679.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Nakaya A, et al. The gold compound auranofin induces apoptosis of human multiple myeloma cells through both down-regulation of STAT3 and inhibition of NF-κB activity. Leuk Res. 2011;35:243–249. doi: 10.1016/j.leukres.2010.05.011. [DOI] [PubMed] [Google Scholar]
  • 194.Song L, et al. Dasatinib (BMS-354825) selectively induces apoptosis in lung cancer cells dependent on epidermal growth factor receptor signaling for survival. Cancer Res. 2006;66:5542–5548. doi: 10.1158/0008-5472.CAN-05-4620. [DOI] [PubMed] [Google Scholar]
  • 195.Song L, et al. JAK1 activates STAT3 activity in non-small-cell lung cancer cells and IL-6 neutralizing antibodies can suppress JAK1-STAT3 signaling. Mol Cancer Ther. 2011;10:481–494. doi: 10.1158/1535-7163.MCT-10-0502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Sen M, et al. First-in-human trial of a STAT3 decoy oligonucleotide in head and neck tumors: implications for cancer therapy. Cancer Discov. 2012;2:694–705. doi: 10.1158/2159-8290.CD-12-0191. This is the first-in-human study of a STAT3-selective inhibitor, which is a transcription factor decoy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Souissi I, et al. A STAT3-inhibitory hairpin decoy oligodeoxynucleotide discriminates between STAT1 and STAT3 and induces death in a human colon carcinoma cell line. Mol Cancer. 2012;11:12. doi: 10.1186/1476-4598-11-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Huckel M, et al. Attenuation of murine antigen-induced arthritis by treatment with a decoy oligodeoxynucleotide inhibiting signal transducer and activator of transcription-1 (STAT-1) Arthritis Res Ther. 2006;8:R17. doi: 10.1186/ar1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Stadlbauer TH, et al. AP-1 and STAT-1 decoy oligodeoxynucleotides attenuate transplant vasculopathy in rat cardiac allografts. Cardiovasc Res. 2008;79:698–705. doi: 10.1093/cvr/cvn135. [DOI] [PubMed] [Google Scholar]
  • 200.Wang X, et al. Targeted blockage of signal transducer and activator of transcription 5 signaling pathway with decoy oligodeoxynucleotides suppresses leukemic K562 cell growth. DNA Cell Biol. 2011;30:71–78. doi: 10.1089/dna.2010.1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Shen J, Li R, Li G. Inhibitory effects of decoy-ODN targeting activated STAT3 on human glioma growth in vivo. In Vivo. 2009;23:237–243. [PubMed] [Google Scholar]
  • 202.Sen M, et al. Targeting Stat3 abrogates EGFR inhibitor resistance in cancer. Clin Cancer Res. 2012;18:4986–4996. doi: 10.1158/1078-0432.CCR-12-0792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Lui VW, et al. Antiproliferative mechanisms of a transcription factor decoy targeting signal transducer and activator of transcription (STAT) 3: the role of STAT1. Mol Pharmacol. 2007;71:1435–1443. doi: 10.1124/mol.106.032284. [DOI] [PubMed] [Google Scholar]
  • 204.Niu G, et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene. 2002;21:2000–2008. doi: 10.1038/sj.onc.1205260. [DOI] [PubMed] [Google Scholar]
  • 205.Barton BE, et al. Novel single-stranded oligonucleotides that inhibit signal transducer and activator of transcription 3 induce apoptosis in vitro and in vivo in prostate cancer cell lines. Mol Cancer Ther. 2004;3:1183–1191. [PubMed] [Google Scholar]
  • 206.Li WC, et al. Inhibition of growth and metastasis of human hepatocellular carcinoma by antisense oligonucleotide targeting signal transducer and activator of transcription 3. Clin Cancer Res. 2006;12:7140–7148. doi: 10.1158/1078-0432.CCR-06-0484. [DOI] [PubMed] [Google Scholar]
  • 207.Burel SA, et al. Preclinical evaluation of the toxicological effects of a novel constrained ethyl modified antisense compound targeting signal transducer and activator of transcription 3 in mice and cynomolgus monkeys. Nucleic Acid Ther. 2013;23:213–227. doi: 10.1089/nat.2013.0422. [DOI] [PubMed] [Google Scholar]

RESOURCES