Abstract
A number of solid tumors are treated with radiation therapy (RT) as a curative modality. At the same time, for certain types of cancers the applicable doses of RT are not high enough to result in a successful eradication of cancer cells. This is often caused by limited pharmacological tools and strategies to selectively sensitize tumors to RT while simultaneously sparing normal tissues from RT. We present an outline of a novel strategy for RT sensitization of solid tumors utilizing Jak inhibitors. Here, recently published pre-clinical data are reviewed which demonstrate the promising role of Jak inhibition in sensitization of tumors to RT. A wide number of currently approved Jak inhibitors for non-malignant conditions are summarized including Jak inhibitors currently in clinical development. Finally, intersection between Jak/Stat and the levels of serum cytokines are presented and discussed as they relate to susceptibility to RT.
Keywords: DNA repair, Jak, Stat, Solid tumors
1. Introduction
Radiation therapy (RT) is a critical and central component of treatment of numerous solid tumor types. For certain solid tumors, RT is the single curative modality. Radiation-induced DNA damage causes death of cancer cells by multiple mechanisms1–5. Indeed, DNA damage imparted by RT exposure has been a robustly studied for many decades. There is a wealth of information on the mechanisms by which a cancer cell embarks on repair of DNA after radiation-induced damage including mitotic cell death, apoptotic cell death, and in some circumstances ablative necrosis2–5. While there are multiple types of DNA damage induced by RT, double strand DNA breaks (DSDBs) constitute the most lethal events associated with radiation exposure1,3,6–8. This makes targeting those pathways a highly appealing strategy in order to optimize radiation induced cell damage for treatment of solid tumors4,8,9.
2. The Key Mechanisms of Repair of Double-Strand DNA Breaks in Solid Tumors
Two major categories of DSDB damage repair include homologous recombination (HR) repair and non-homologous end joining repair (NHEJ) (Fig. 1) 1,5,6,10,11. In brief, NHEJ DNA repair mechanism, unlike HR DNA repair, does not require a long homologous sequence to guide the repair. NHEJ is a straightforward process of re-ligation of the broken DNA ends12 and therefore does not require a template12. The important repair protein responsible for NHEJ is DNA-dependent protein kinase (DNA-PK), a serine-threonine protein kinase consisting of three subunits (DNA-PKcs, Ku70 and Ku80)1,13–15. The first protein to bind the DSB is Ku heterodimer and is capable of interacting with a nuclease (Artemis-DNA-PKcs), the polymerases (u and λ) and a ligase (XLF-XRCC4-DNA ligase IV)12,16. In contrast, HR DNA repair is a relatively error free process that uses a DNA template of the same base sequence around the break site to perform accurate repair of the DNA break17–19. The double strand DNA breaks are initially recognized by the MRN (MRE11, RAD50, NbS1) protein complex, which together with phosphorylated CtIP (C-terminal binding protein interacting protein) and BRCA1 (Breast Cancer Susceptibility 1), generate the necessary 3’ single-stranded DNA (ssDNA) overhangs for HR repair1,11,20–23. BRCA1 promotes this resection by dephosphorylating the inhibitory 53BP1 protein11,20–24. Following end-resection, the MRN complex recruits and activates ATM (ataxia telangiectasia mutated) kinase, which phosphorylates histone H2AX on regions around DNA DSDBs25. Importantly, loading of RAD51 recombinase onto exposed ssDNA repair sites is carried out by BRCA1 and requires assistance of BRCA223,26. RAD51 is a requisite for HR DNA repair as it catalyzes DNA strand invasion and exchange of the DNA strands23,27. RAD51 paralogues including RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3 have only a supporting role in this process by assisting RAD51 in the initiation and execution of HR DNA repair28. It is important to note that while BRCA1/2 assist RAD51 in the HR DNA repair, RAD51 is indispensable for HR DNA repair 11,20–22 in its key role for strand invasion and exchange11,20–22,28. BRCA1/2-deficient cancer cells have defective HR DNA repair (termed BRCA-ness), and consequently increased sensitivity to RT, platinum chemotherapy and PARP-inhibitors29–31.
Increased levels of RAD51 are commonly seen in solid tumors, and higher levels are associated with improved tumor repair capabilities. Numerous studies have suggested that higher levels of RAD51 can lead to worse clinical outcomes, which often are evident specifically in tumors that are commonly treated with RT. The correlation with RAD51 expression and clinical outcomes has been shown in esophageal cancer32, breast cancer33, head and neck cancer34, prostate cancer35, soft tissue sarcoma36, and pancreatic cancer37, to name a few examples. In addition, cancer cells tend to have a higher ratio of cells in S and G2 phases of the cell cycle, which causes them to favor HR DNA repair over NHEJ repair3,6,7,38.
Given the central role of RAD51 in the HR process, together with the fact that DSDBs represent a primary form of DNA damage secondary to RT exposure, RAD51 is potentially a critical protein mediating resistance of tumors to treatment with RT. Solid tumor patients with germ-line BRCA1/2 mutations, which incapacitate BRCA1/2 functions and HR DNA repair (termed BRCA-ness), are known to be more responsive to RT, platinum-based chemotherapeutics and PARP-inhibitors 29–31,39. PARP1, through the addition of poly-ADP ribose (PAR) moieties to sites of single-strand (ss) DNA breaks, is critical for recognition and recruitment of DNA repair machinery for a variety of different DNA repair processes40. If ssDNA breaks go unrepaired, DSDBs form during the cell cycle. In cells with intact repair pathways, HR DNA repair will function to repair the newly formed DSB of DNA. In the case of solid tumors with BRCA1/2 mutations, where BRCA-mediated HR repair is already deficient, the use of PARP inhibitors alone can promote the accumulation of DSDBs leading to selective death of tumor cells – a concept referred to as “synthetic lethality”41–46. Given that RAD51 is overexpressed in a number of solid tumors treated with RT, targeting RAD51 represents an appealing strategy to preferentially sensitize cancer cells to treatment with RT and induce transient BRCA-ness to solid tumors47,48.
3. Jak-Stat Signaling Pathway
Signal Transducer and Activator of Transcription (STAT) proteins are critical mediators of the cellular actions of a number of peptide hormones, growth factors and cytokines (Fig. 2)49. The canonical pathway for activating of STAT involves tyrosine phosphorylation of STAT by a JAK tyrosine kinase. Activation of STAT is a two-step process where, first, the STAT monomer docks transiently to a phospho-tyrosyl moiety of the tyrosine kinase-receptor complex, which results in the phosphorylation of a specific tyrosine residue in the C-terminus of STAT protein by a tyrosine kinase50. In the next step, the SH2-domain of a phosphorylated STAT monomer binds the phosphorylated tyrosine residue of the partner STAT to form a transcriptionally active parallel dimer, which translocates to the nucleus to regulate transcription50. In other words, STAT dimerization is also mediated by the SH2-domain of STAT molecule. This leads to nuclear translocation of dimerized STAT which binds to DNA to directly regulate transcription (Fig. 3)51. STAT5 has been shown to regulate growth and differentiation of cells in several solid tumors, including hormone-regulated cancers such as prostate cancer and breast cancer52. In addition, STAT5 has been shown to be expressed in approximately 50% of pancreatic adenocarcinomas53. In prostate tumors, STAT5 is associated with more biologically aggressive cancer54 and development of epithelial-to-mesenchymal transition32,55. Numerous independent studies have provided proof-of-concept that Stat5 drives prostate cancer growth52,54–77. Stat5 sustains prostate cancer cell viability in vitro and induces prostate tumor growth in mice. Consequently, blockade of Stat5 signaling induces death of prostate cancer cells and blocks growth of prostate tumors55,57,58,60–62,64,67,68,78. Overexpression of active Stat5, in turn, induces growth of PC cells in culture and tumor growth in mice65. Recent work demonstrates a novel concept that the second-generation anti-androgen Enzalutamide induces sustained Jak2-Stat5 phosphorylation in prostate cancer, which promotes prostate cancer growth during treatment of prostate cancer with Enzalutamide 78. Enzalutamide-induced Jak2 phosphorylation was shown to occur through a process involving Jak2-regulating protein SHP2 which leads to Stat5 phosphorylation and a formation of a hyperactivated feed-forward loop78. An appealing aspect of the STAT5 pathway is that in 30–40% of advanced prostate cancers, cytological analysis of the chromosome 17 locus encompassing STAT5A and STAT5B genes has revealed STAT5A/B gene amplification, resulting in increased STAT5 protein levels65, especially when compared to normal prostate epithelium54. Notably, high STAT5 expression in clinical prostate cancers, at the time of the initial treatment, predicted development of castrate-resistant disease in three independent cohorts totaling 1,035 patients74,75,77. A recent study shows that combined positive status for both STAT5 protein expression and gene amplification in prostate cancer at the time of radical prostatectomy is a powerful predictor of prostate cancer recurrence in a multivariate analysis even when comparing to the variables of the CAPRA-S nomogram77. The finding of STAT5 as a strong predictor of clinical progression of prostate cancer to lethal castrate-resistant state74,75,77 demonstrates the significance of STAT5 in prostate cancer growth and progression of prostate cancer in patients, corroborating the results obtained using preclinical prostate cancer model systems52,55–73.
In regards of radiation response of solid tumors, RAD51 is central in the repair of RT induced DNA damage via HR and is upregulated in most solid tumors. This has recently lead to a highly compelling finding that STAT5 inhibition sensitizes prostate cancer cells to radiation-induced cell death and increased exposure to RT, without a similar effect on neighboring normal tissues76. The molecular mechanisms underlying STAT5-regulation of radiation-induced prostate cancer cell death involves up-regulation of RAD51 levels by JAK2-STAT5 signaling pathway76. Specifically, the regulatory regions of the RAD51 gene have been shown to contain STAT5 response elements, and STAT5 directly induces the levels of RAD51 mRNA and protein in prostate cancer79. Such a finding supports a persuasive concept to be clinically translated into early phase clinical trial as a novel method of radiation sensitization of prostate cancer42,80.
4. STAT3 and DNA repair in solid tumors
Activation of STAT3 has been demonstrated in several different human cancer cell lines along with actual tumor specimen. Tumor examples in which activation of STAT3 has been demonstrated include pancreatic adenocarcinoma, prostate cancer, head and neck cancer, lymphoma, and breast cancer81. Aberrant STAT3 activation in tumor cells has been shown to be associated with increased cell survival, increased cell growth, along with potentially invasion and the development of metastatic disease81.
The role of STAT3 in DNA repair is supported by the following findings: STAT3 deficient mouse fibroblast cells showed reduced activity of the ATM-Chk1 and ATR-Chk2 pathways, both important in sensing DNA damage82. Cells lacking STAT3 also failed to induce mRNA expression of MDC1, a regulator of the ATM-Chk1 pathway and facilitator of the DNA damage response82. In conjunction with reduced active ATM, STAT3-deficient cells exhibited reduced phosphorylation of H2AX82. Reduced activity of H2AX slows down the rate of accumulation of DNA repair factors around the site of strand breakage, thereby hampering the DNA repair. In DU145 human prostate cancer cells, enhanced expression of BRCA1 resulted in the constitutive activation of JAK-STAT3 signalling pathway, providing critical survival signal for tumor formation83.
DSDBs are the most critical type of DNA damage induced by radio/chemotherapy and activate ATM-ATR-DNA PKcs signalling to promote DNA repair84. Lung, bone, and prostate cancer cells demonstrate that DSDBs upregulate PD-L1 expression in an ATM/ATR/Chk1-dependent manner and lead to the activation of STAT3 signalling together with STAT1/IRF1 for DSB-dependent PD-L1 upregulation84. The major role of PD-1/PD-L1 is to regulate autoimmune response in the peripheral tissue to maintain a balanced immune response in the body.
Recent studies also revealed that Cetuximab, monoclonal antibody designed to inhibit EGFR-ligand interaction led to the activation of STAT3 in several tumor cell lines as well as primary glioblastoma cells85. Cetuximab mediated STAT3 activation has been shown to promote elevated levels of the regulatory subunit Eme1 of the heterodimeric endonuclease Mus81/Eme1 complex, that is involved in DNA repair pathways that remove UV light–induced DNA lesions and cross-links between DNA strands85. Cetuximab treatment increased the phosphorylation of the checkpoint kinases Chk1 and Chk2 and its downstream effectors p53 and Histone H2AX85. In summary, these findings support the concept that cancer cells which overexpress STAT3 may be resistant to chemotherapeutics through their increased efficiency of DNA repair and greater resistance to genotoxic stress.
5. Cytokines and JAK-STAT pathway in solid tumors
The JAK and subsequent STAT activation pathway is known to be triggered by a variety of cytokines, particularly IL-10 and IL-6. These cytokine levels are highly relevant to consider in the context of several solid tumors, as both have been associated with higher rates of metastatic disease, together with potentially resistance to treatment with RT. Engagement of IL-10 receptor leads to JAK1 activation, which induces phosphorylation of STAT1, STAT3, and STAT586. Such an association may imply that IL-10 is associated with poor response to RT and associate with the development of distant metastatic disease in patients with high risk prostate cancer87. In addition, IL-6 has been shown to activate JAK family members JAK1, JAK2, and TYK2, which leads to the activation STAT388. Similarly serum levels of IL-6 have been associated with poor response to RT, which could be potentially mechanistically explained by the IL-6 activation of STAT3 and JAK1/2 signaling89. In addition, it has recently been shown that IL-6 leads to prostate cancer resistance to RT through upregulation of DNA repair90.
6. Oncological Relevance
The ability to preferentially sensitize certain tumors to RT without simultaneous sensitization of the surrounding normal tissues is highly attractive approach for the treatment of several malignancies. This is because RT is associated with a potential for toxicities to the neighboring organs at risk. In addition, RT can not be given at sufficient doses to induce effective killing of cancer cells in a number of malignancies and therefore requires the use of surgery or concurrent chemotherapy. Furthermore, many tumors progress despite treatment with RT. Oftentimes these progression events happen directly within the RT-treated volume of the tumor tissue. Combining RT with novel mechanisms of radiation sensitization would enable higher efficacy of RT while maintaining acceptable toxicity levels. This would both improve the therapeutic ratio and limit the potential for side-effects of radiation for multiple malignancies. These include certain types of prostate cancer, such as high risk, node positive prostate cancer which both have a poor prognosis with the current standard of care treatment strategies using RT91. It is important to note that effective RT sensitization strategies for this malignancy are currently absent.
Inoperable pancreatic cancer presents another highly appealing malignancy for improved attempted RT sensitization as current treatment strategies with concurrent chemotherapy and RT are ineffective and novel RT sensitization strategies are lacking92. Some tumors that currently have poor response rates to RT, such as glioblastomas, have known associations with STAT transcription factors, including STAT3 which has been shown to be elevated in glioblastoma tissue93. These tumors also have high in field failure rates when treated with RT and also require the use of surgery94. In addition, patients with colon cancer positive for JAK-1 and STAT have been shown to have shorter survival95. This introduces a potentially attractive option for inhibition of JAK proteins and subsequent treatment with RT for colon cancer. The malignancies addressed in this section are just a fraction of the potential cancers that could benefit from increased sensitization to RT via exploitation of the JAK-STAT pathway.
There exists a number of inhibitors of JAK tyrosine kinases, several of which are FDA approved for treatment of non-malignant conditions or under clinical investigation. Table 1 provides a list of compounds that are either currently approved, or in current clinical development.
Table 1.
Compound Name | Target | Clinical Indications | Citations |
---|---|---|---|
Approved: | |||
Baricitinib (trade name Olumiant) | JAK1/JAK2 | Rheumatoid Arthritis | 28 |
Ruxolitinib (trade names Jakafi/Jakavi) | JAK1/JAK2 | Psoriasis, Myelofibrosis, Rheumatoid Arthritis, Polycythemia Vera | 96 |
Tofacitinib (trade names Xeljanz/Jakvinus | JAK3 | Psoriasis and Rheumatoid Arthritis | 97 |
Peficitinib (ASP015 K, JNJ-54781532; trade name Smyraf) | JAK3 | Rheumatoid Arthritis | 98 |
Fedratinib (SAR302503; trade name Inrebic) | JAK2 | Myelofibrosis | 99 |
Upadacitinib (trade name Rinvoq) | JAK1 | Rheumatoid Arthritis | 100 |
In Trials: | |||
Filgotinib (G-146034, GLPG-0634) | JAK1 (Phase III) | Rheumatoid Arthritis, Chron’s disease | 101 |
Gandotinib (LY-2784544) | JAK2 (Phase II) | Myeloproliferative neoplasm | 102 |
Lestaurtinib (CEP-701) | JAK2 (Phase II-III) | Acute Myeloid leukemia | 103 |
Pacritinib (SB1518) | JAK2 (Phase III) | Relapse lymphoma, advanced myeloid malignancies, myelofibrosis | 104 |
Momelotinib(GS-0387, CYT-387) | JAK1 and JAK2 (Phase I/II) | Myelofibrosis | 105 |
7. Conclusions
Relatively little research has been conducted to examine the combination of JAK or STAT inhibition with RT to sensitize solid tumors to treatment with RT. This is despite compelling pre-clinical and clinical evidence that the synergy between these treatment modalities could potentially be very strong. Clinical trials are needed to evaluate the combination of RT and JAK-STAT inhibition in a variety of solid tumors.
Acknowledgements
This work was financially supported by NCI/NIH Exploratory/Developmental Research Grant (1R21CA178755–01), Advancing a Healthier Wisconsin (#5520368) and Wisconsin Cancer Showhouse Grant (#15437) to M.T. Nevalainen. The project described was supported by the National Center for Advancing Translational Sciences, National Institutes of Health, Award Number KL2TR001438 to William A. Hall. The content is solely the responsibility of the author(s) and does not necessarily represent the official views of the NIH.
Footnotes
Declaration of Competing Interest
William A. Hall receives departmental research and travel support from Elekta AB Stockholm.
References
- [1].Ranjha L, Howard SM, Cejka P, 2018. Main steps in DNA double-strand break repair: an introduction to homologous recombination and related processes. Chromosoma 127, 187–214. [DOI] [PubMed] [Google Scholar]
- [2].Her J, Bunting SF, 2018. How cells ensure correct repair of DNA double-strand breaks. J Biol Chem 293, 10502–10511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Roos WP, Thomas AD, Kaina B, 2016. DNA damage and the balance between survival and death in cancer biology. Nat Rev Cancer 16, 20–33. [DOI] [PubMed] [Google Scholar]
- [4].Hosoya N, Miyagawa K, 2014. Targeting DNA damage response in cancer therapy. Cancer Sci 105, 370–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Ceccaldi R, Rondinelli B, D’Andrea AD, 2016. Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol 26, 52–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Krenning L, van den Berg J, Medema RH, 2019. Life or Death after a Break: What Determines the Choice? Mol Cell 76, 346–358. [DOI] [PubMed] [Google Scholar]
- [7].Santivasi WL, Xia F, 2014. Ionizing radiation-induced DNA damage, response, and repair. Antioxid Redox Signal 21, 251–259. [DOI] [PubMed] [Google Scholar]
- [8].Bernier J, Hall EJ, Giaccia A, 2004. Radiation oncology: a century of achievements. Nat Rev Cancer 4, 737–747. [DOI] [PubMed] [Google Scholar]
- [9].Pilie PG, Tang C, Mills GB, Yap TA, 2019. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol 16, 81–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].West SC, 2003. Molecular views of recombination proteins and their control. Nat Rev Mol Cell Biol 4, 435–445. [DOI] [PubMed] [Google Scholar]
- [11].Jasin M, Rothstein R, 2013. Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol 5, a012740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Lieber MR, 2008. The mechanism of human nonhomologous DNA end joining. J Biol Chem 283, 1–5. [DOI] [PubMed] [Google Scholar]
- [13].Nemoz C, Ropars V, Frit P, Gontier A, Drevet P, Yu J, Guerois R, Pitois A, Comte A, Delteil C, Barboule N, Legrand P, Baconnais S, Yin Y, Tadi S, Barbet-Massin E, Berger I, Le Cam E, Modesti M, Rothenberg E, Calsou P, Charbonnier JB, 2018. XLF and APLF bind Ku80 at two remote sites to ensure DNA repair by non-homologous end joining. Nat Struct Mol Biol 25, 971–980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Pannunzio NR, Li S, Watanabe G, Lieber MR, 2014. Non-homologous end joining often uses microhomology: implications for alternative end joining. DNA Repair (Amst) 17, 74–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Pannunzio NR, Watanabe G, Lieber MR, 2018. Nonhomologous DNA end-joining for repair of DNA double-strand breaks. J Biol Chem 293, 10512–10523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Hnizda A, Blundell TL, 2019. Multicomponent assemblies in DNA-double-strand break repair by NHEJ. Curr Opin Struct Biol 55, 154–160. [DOI] [PubMed] [Google Scholar]
- [17].Aparicio T, Baer R, Gautier J, 2014. DNA double-strand break repair pathway choice and cancer. DNA Repair (Amst) 19, 169–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Groschel S, Hubschmann D, Raimondi F, Horak P, Warsow G, Frohlich M, Klink B, Gieldon L, Hutter B, Kleinheinz K, Bonekamp D, Marschal O, Chudasama P, Mika J, Groth M, Uhrig S, Kramer S, Heining C, Heilig CE, Richter D, Reisinger E, Pfutze K, Eils R, Wolf S, von Kalle C, Brandts C, Scholl C, Weichert W, Richter S, Bauer S, Penzel R, Schrock E, Stenzinger A, Schlenk RF, Brors B, Russell RB, Glimm H, Schlesner M, Frohling S, 2019. Defective homologous recombination DNA repair as therapeutic target in advanced chordoma. Nat Commun 10, 1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Liu H, Zhang H, Wu X, Ma D, Wu J, Wang L, Jiang Y, Fei Y, Zhu C, Tan R, Jungblut P, Pei G, Dorhoi A, Yan Q, Zhang F, Zheng R, Liu S, Liang H, Liu Z, Yang H, Chen J, Wang P, Tang T, Peng W, Hu Z, Xu Z, Huang X, Wang J, Li H, Zhou Y, Liu F, Yan D, Kaufmann SHE, Chen C, Mao Z, Ge B, 2018. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature 563, 131–136. [DOI] [PubMed] [Google Scholar]
- [20].Mateo J, Boysen G, Barbieri CE, Bryant HE, Castro E, Nelson PS, Olmos D, Pritchard CC, Rubin MA, de Bono JS, 2017. DNA Repair in Prostate Cancer: Biology and Clinical Implications. Eur Urol 71, 417–425. [DOI] [PubMed] [Google Scholar]
- [21].Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA, 2008. DNA repair pathways as targets for cancer therapy. Nat Rev Cancer 8, 193–204. [DOI] [PubMed] [Google Scholar]
- [22].McLornan DP, List A, Mufti GJ, 2014. Applying synthetic lethality for the selective targeting of cancer. N Engl J Med 371, 1725–1735. [DOI] [PubMed] [Google Scholar]
- [23].Prakash R, Zhang Y, Feng W, Jasin M, 2015. Homologous recombination and human health: the roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb Perspect Biol 7, a016600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Kolinjivadi AM, Sannino V, de Antoni A, Techer H, Baldi G, Costanzo V, 2017. Moonlighting at replication forks - a new life for homologous recombination proteins BRCA1, BRCA2 and RAD51. FEBS Lett 591, 1083–1100. [DOI] [PubMed] [Google Scholar]
- [25].Marechal A, Zou L, 2013. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Lord CJ, Ashworth A, 2016. BRCAness revisited. Nat Rev Cancer 16, 110–120. [DOI] [PubMed] [Google Scholar]
- [27].Moynahan ME, Pierce AJ, Jasin M, 2001. BRCA2 is required for homology-directed repair of chromosomal breaks. Mol Cell 7, 263–272. [DOI] [PubMed] [Google Scholar]
- [28].Takata M, Sasaki MS, Tachiiri S, Fukushima T, Sonoda E, Schild D, Thompson LH, Takeda S, 2001. Chromosome instability and defective recombinational repair in knockout mutants of the five Rad51 paralogs. Mol Cell Biol 21, 2858–2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, Kyle S, Meuth M, Curtin NJ, Helleday T, 2005. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917. [DOI] [PubMed] [Google Scholar]
- [30].Farmer H, McCabe N, Lord CJ, Tutt AN, Johnson DA, Richardson TB, Santarosa M, Dillon KJ, Hickson I, Knights C, Martin NM, Jackson SP, Smith GC, Ashworth A, 2005. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921. [DOI] [PubMed] [Google Scholar]
- [31].Tutt AN, Lord CJ, McCabe N, Farmer H, Turner N, Martin NM, Jackson SP, Smith GC, Ashworth A, 2005. Exploiting the DNA repair defect in BRCA mutant cells in the design of new therapeutic strategies for cancer. Cold Spring Harb Symp Quant Biol 70, 139–148. [DOI] [PubMed] [Google Scholar]
- [32].Chiu WC, Fang PT, Lee YC, Wang YY, Su YH, Hu SC, Chen YK, Tsui YT, Kao YH, Huang MY, Yuan SF, 2020. DNA Repair Protein Rad51 Induces Tumor Growth and Metastasis in Esophageal Squamous Cell Carcinoma via a p38/Akt-Dependent Pathway. Ann Surg Oncol 27, 2090–2101. [DOI] [PubMed] [Google Scholar]
- [33].Barbano R, Copetti M, Perrone G, Pazienza V, Muscarella LA, Balsamo T, Storlazzi CT, Ripoli M, Rinaldi M, Valori VM, Latiano TP, Maiello E, Stanziale P, Carella M, Mangia A, Pellegrini F, Bisceglia M, Muda AO, Altomare V, Murgo R, Fazio VM, Parrella P, 2011. High RAD51 mRNA expression characterize estrogen receptor-positive/progesteron receptor-negative breast cancer and is associated with patient’s outcome. Int J Cancer 129, 536–545. [DOI] [PubMed] [Google Scholar]
- [34].Connell PP, Jayathilaka K, Haraf DJ, Weichselbaum RR, Vokes EE, Lingen MW, 2006. Pilot study examining tumor expression of RAD51 and clinical outcomes in human head cancers. Int J Oncol 28, 1113–1119. [PubMed] [Google Scholar]
- [35].Mitra A, Jameson C, Barbachano Y, Sanchez L, Kote-Jarai Z, Peock S, Sodha N, Bancroft E, Fletcher A, Cooper C, Easton D, Committee IS, Impact, Collaborators E, Eeles R, Foster CS, 2009. Overexpression of RAD51 occurs in aggressive prostatic cancer. Histopathology 55, 696–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Hannay JA, Liu J, Zhu QS, Bolshakov SV, Li L, Pisters PW, Lazar AJ, Yu D, Pollock RE, Lev D, 2007. Rad51 overexpression contributes to chemoresistance in human soft tissue sarcoma cells: a role for p53/activator protein 2 transcriptional regulation. Mol Cancer Ther 6, 1650–1660. [DOI] [PubMed] [Google Scholar]
- [37].Maacke H, Jost K, Opitz S, Miska S, Yuan Y, Hasselbach L, Luttges J, Kalthoff H, Sturzbecher HW, 2000. DNA repair and recombination factor Rad51 is over-expressed in human pancreatic adenocarcinoma. Oncogene 19, 2791–2795. [DOI] [PubMed] [Google Scholar]
- [38].Giaccia, EJHaAJ, 2006. Radiobiology for the Radiologist, Sixth Edition ed. [Google Scholar]
- [39].Abbotts R, Topper MJ, Biondi C, Fontaine D, Goswami R, Stojanovic L, Choi EY, McLaughlin L, Kogan AA, Xia L, Lapidus R, Mahmood J, Baylin SB, Rassool FV, 2019. DNA methyltransferase inhibitors induce a BRCAness phenotype that sensitizes NSCLC to PARP inhibitor and ionizing radiation. Proc Natl Acad Sci U S A 116, 22609–22618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Ray Chaudhuri A, Nussenzweig A, 2017. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol 18, 610–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].O’Neil NJ, Bailey ML, Hieter P, 2017. Synthetic lethality and cancer. Nat Rev Genet 18, 613–623. [DOI] [PubMed] [Google Scholar]
- [42].Ashworth A, Lord CJ, 2018. Synthetic lethal therapies for cancer: what’s next after PARP inhibitors? Nat Rev Clin Oncol 15, 564–576. [DOI] [PubMed] [Google Scholar]
- [43].Lord CJ, Ashworth A, 2017. PARP inhibitors: Synthetic lethality in the clinic. Science 355, 1152–1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Bajrami I, Kigozi A, Van Weverwijk A, Brough R, Frankum J, Lord CJ, Ashworth A, 2012. Synthetic lethality of PARP and NAMPT inhibition in triple-negative breast cancer cells. EMBO Mol Med 4, 1087–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Lord CJ, Tutt AN, Ashworth A, 2015. Synthetic lethality and cancer therapy: lessons learned from the development of PARP inhibitors. Annu Rev Med 66, 455–470. [DOI] [PubMed] [Google Scholar]
- [46].Rehman FL, Lord CJ, Ashworth A, 2010. Synthetic lethal approaches to breast cancer therapy. Nat Rev Clin Oncol 7, 718–724. [DOI] [PubMed] [Google Scholar]
- [47].Huang A, Garraway LA, Ashworth A, Weber B, 2020. Synthetic lethality as an engine for cancer drug target discovery. Nat Rev Drug Discov 19, 23–38. [DOI] [PubMed] [Google Scholar]
- [48].Wera AC, Lobbens A, Stoyanov M, Lucas S, Michiels C, 2019. Radiation-induced synthetic lethality: combination of poly(ADP-ribose) polymerase and RAD51 inhibitors to sensitize cells to proton irradiation. Cell Cycle 18, 1770–1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Levy DE, Darnell JE Jr, 2002. Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol 3, 651–662. [DOI] [PubMed] [Google Scholar]
- [50].Darnell JE Jr., Kerr IM, Stark GR, 1994. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264, 1415–1421. [DOI] [PubMed] [Google Scholar]
- [51].Li WX, 2008. Canonical and non-canonical JAK-STAT signaling. Trends Cell Biol 18, 545–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Tan SH, Nevalainen MT, 2008. Signal transducer and activator of transcription 5A/B in prostate and breast cancers. Endocr Relat Cancer 15, 367–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Moser C, Ruemmele P, Gehmert S, Schenk H, Kreutz MP, Mycielska ME, Hackl C, Kroemer A, Schnitzbauer AA, Stoeltzing O, Schlitt HJ, Geissler EK, Lang SA, 2012. STAT5b as molecular target in pancreatic cancer–inhibition of tumor growth, angiogenesis, and metastases. Neoplasia 14, 915–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Li H, Ahonen TJ, Alanen K, Xie J, LeBaron MJ, Pretlow TG, Ealley EL, Zhang Y, Nurmi M, Singh B, Martikainen PM, Nevalainen MT, 2004. Activation of signal transducer and activator of transcription 5 in human prostate cancer is associated with high histological grade. Cancer Res 64, 4774–4782. [DOI] [PubMed] [Google Scholar]
- [55].Talati PG, Gu L, Ellsworth EM, Girondo MA, Trerotola M, Hoang DT, Leiby B, Dagvadorj A, McCue PA, Lallas CD, Trabulsi EJ, Gomella L, Aplin AE, Languino L, Fatatis A, Rui H, Nevalainen MT, 2015. Jak2-Stat5a/b Signaling Induces Epithelial-to-Mesenchymal Transition and Stem-Like Cell Properties in Prostate Cancer. Am J Pathol 185, 2505–2522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Ahonen TJ, Xie J, LeBaron MJ, Zhu J, Nurmi M, Alanen K, Rui H, Nevalainen MT, 2003. Inhibition of transcription factor Stat5 induces cell death of human prostate cancer cells. J Biol Chem 278, 27287–27292. [DOI] [PubMed] [Google Scholar]
- [57].Dagvadorj A, Kirken RA, Leiby B, Karras J, Nevalainen MT, 2008. Transcription factor signal transducer and activator of transcription 5 promotes growth of human prostate cancer cells in vivo. Clin Cancer Res 14, 1317–1324. [DOI] [PubMed] [Google Scholar]
- [58].Gu L, Liao Z, Hoang DT, Dagvadorj A, Gupta S, Blackmon S, Ellsworth E, Talati P, Leiby B, Zinda M, Lallas CD, Trabulsi EJ, McCue P, Gomella L, Huszar D, Nevalainen MT, 2013. Pharmacologic Inhibition of Jak2-Stat5 Signaling By Jak2 Inhibitor AZD1480 Potently Suppresses Growth of Both Primary and Castrate-Resistant Prostate Cancer. Clin Cancer Res 19, 5658–5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Hoang DT, Gu L, Liao Z, Shen F, Talati PG, Koptyra M, Tan SH, Ellsworth E, Gupta S, Montie H, Dagvadorj A, Savolainen S, Leiby B, Mirtti T, Merry DE, Nevalainen MT, 2015. Inhibition of Stat5a/b Enhances Proteasomal Degradation of Androgen Receptor Liganded by Antiandrogens in Prostate Cancer. Mol Cancer Ther 14, 713–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Gu LZX, Visakorpi T, Alanen K, Mirtti T, Edmonston T, Nevalainen MT, 2010. Activating mutation (V617F) in the tyrosine kinase Jak2 is absent in local-confined and castration-resistant prostate cancer. Cellular Oncology 33, 55–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Gu L, Vogiatzi P, Puhr M, Dagvadorj A, Lutz J, Ryder A, Addya S, Fortina P, Cooper C, Leiby B, Dasgupta A, Hyslop T, Bubendorf L, Alanen K, Mirtti T, Nevalainen MT, 2010. Stat5 promotes metastatic behavior of human prostate cancer cells in vitro and in vivo. Endocr Relat Cancer 17, 481–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Hoang DT, Iczkowski KA, Kilari D, See W, Nevalainen MT, 2017. Androgen receptor-dependent and -independent mechanisms driving prostate cancer progression: Opportunities for therapeutic targeting from multiple angles. Oncotarget 8, 3724–3745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Gu L, Dagvadorj A, Lutz J, Leiby B, Bonuccelli G, Lisanti MP, Addya S, Fortina P, Dasgupta A, Hyslop T, Bubendorf L, Nevalainen MT, 2010. Transcription factor Stat3 stimulates metastatic behavior of human prostate cancer cells in vivo, whereas Stat5b has a preferential role in the promotion of prostate cancer cell viability and tumor growth. Am J Pathol 176, 1959–1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Liao Z, Gu L, Vergalli J, Mariani SA, De Dominici M, Lokareddy RK, Dagvadorj A, Purushottamachar P, McCue PA, Trabulsi E, Lallas CD, Gupta S, Ellsworth E, Blackmon S, Ertel A, Fortina P, Leiby B, Xia G, Rui H, Hoang DT, Gomella LG, Cingolani G, Njar V, Pattabiraman N, Calabretta B, Nevalainen MT, 2015. Structure-Based Screen Identifies a Potent Small Molecule Inhibitor of Stat5a/b with Therapeutic Potential for Prostate Cancer and Chronic Myeloid Leukemia. Mol Cancer Ther 14, 1777–1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Haddad BR, Gu L, Mirtti T, Dagvadorj A, Vogiatzi P, Hoang DT, Bajaj R, Leiby B, Ellsworth E, Blackmon S, Ruiz C, Curtis M, Fortina P, Ertel A, Liu C, Rui H, Visakorpi T, Bubendorf L, Lallas CD, Trabulsi EJ, McCue P, Gomella L, Nevalainen MT, 2013. STAT5A/B gene locus undergoes amplification during human prostate cancer progression. Am J Pathol 182, 2264–2275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Tan SH, Dagvadorj A, Shen F, Gu L, Liao Z, Abdulghani J, Zhang Y, Gelmann EP, Zellweger T, Culig Z, Visakorpi T, Bubendorf L, Kirken RA, Karras J, Nevalainen MT, 2008. Transcription factor Stat5 synergizes with androgen receptor in prostate cancer cells. Cancer Res 68, 236–248. [DOI] [PubMed] [Google Scholar]
- [67].Kazansky AV, Spencer DM, Greenberg NM, 2003. Activation of signal transducer and activator of transcription 5 is required for progression of autochthonous prostate cancer: evidence from the transgenic adenocarcinoma of the mouse prostate system. Cancer Res 63, 8757–8762. [PubMed] [Google Scholar]
- [68].Thomas C, Zoubeidi A, Kuruma H, Fazli L, Lamoureux F, Beraldi E, Monia BP, MacLeod AR, Thuroff JW, Gleave ME, 2011. Transcription factor Stat5 knockdown enhances androgen receptor degradation and delays castration-resistant prostate cancer progression in vivo. Mol Cancer Ther 10, 347–359. [DOI] [PubMed] [Google Scholar]
- [69].Sackmann-Sala L, Chiche A, Mosquera-Garrote N, Boutillon F, Cordier C, Pourmir I, Pascual-Mathey L, Kessal K, Pigat N, Camparo P, Goffin V, 2014. Prolactin-induced prostate tumorigenesis links sustained Stat5 signaling with the amplification of basal/stem cells and emergence of putative luminal progenitors. Am J Pathol 184, 3105–3119. [DOI] [PubMed] [Google Scholar]
- [70].Reddy AHC, Liu H, DeLisi C, Nevalainen MT, Szalma S, Bhanot G, 2010. Robust network analysis reveals alteration of the Stat5a network as a hallmark of prostate cancer. Genome Informatics 24, 139–153. [PMC free article] [PubMed] [Google Scholar]
- [71].Song C, Kim Y, Min GE, Ahn H, 2014. Dihydrotestosterone enhances castration-resistant prostate cancer cell proliferation through STAT5 activation via glucocorticoid receptor pathway. Prostate 74, 1240–1248. [DOI] [PubMed] [Google Scholar]
- [72].Roe K, Bratland A, Vlatkovic L, Ragnum HB, Saelen MG, Olsen DR, Marignol L, Ree AH, 2013. Hypoxic tumor kinase signaling mediated by STAT5A in development of castration-resistant prostate cancer. PLoS One 8, e63723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [73].Feldman L, Wang Y, Rhim JS, Bhattacharya N, Loda M, Sytkowski AJ, 2006. Erythropoietin stimulates growth and STAT5 phosphorylation in human prostate epithelial and prostate cancer cells. Prostate 66, 135–145. [DOI] [PubMed] [Google Scholar]
- [74].Li H, Zhang Y, Glass A, Zellweger T, Gehan E, Bubendorf L, Gelmann EP, Nevalainen MT, 2005. Activation of signal transducer and activator of transcription-5 in prostate cancer predicts early recurrence. Clin Cancer Res 11, 5863–5868. [DOI] [PubMed] [Google Scholar]
- [75].Mirtti T, Leiby BE, Abdulghani J, Aaltonen E, Pavela M, Mamtani A, Alanen K, Egevad L, Granfors T, Josefsson A, Stattin P, Bergh A, Nevalainen MT, 2013. Nuclear Stat5a/b predicts early recurrence and prostate cancer-specific death in patients treated by radical prostatectomy. Hum Pathol 44, 310–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Maranto C, Udhane V, Hoang DT, Gu L, Alexeev V, Malas K, Cardenas K, Brody JR, Rodeck U, Bergom C, Iczkowski KA, Jacobsohn K, See W, Schmitt SM, Nevalainen MT, 2018. STAT5A/B Blockade Sensitizes Prostate Cancer to Radiation through Inhibition of RAD51 and DNA Repair. Clin Cancer Res 24, 1917–1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Haddad BR, Erickson A, Udhane V, LaViolette PS, Rone JD, Kallajoki MA, See WA, Rannikko A, Mirtti T, Nevalainen MT, 2019. Positive STAT5 Protein and Locus Amplification Status Predicts Recurrence after Radical Prostatectomy to Assist Clinical Precision Management of Prostate Cancer. Cancer Epidemiol Biomarkers Prev 28, 1642–1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Udhane V, Maranto C, Hoang DT, Gu L, Erickson A, Devi S, Talati PG, Banerjee A, Iczkowski KA, Jacobsohn K, See WA, Mirtti T, Kilari D, Nevalainen MT, 2020. Enzalutamide-Induced Feed-Forward Signaling Loop Promotes Therapy-Resistant Prostate Cancer Growth Providing an Exploitable Molecular Target for Jak2 Inhibitors. Mol Cancer Ther 19, 231–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Hasselbach L, Haase S, Fischer D, Kolberg HC, Sturzbecher HW, 2005. Characterisation of the promoter region of the human DNA-repair gene Rad51. Eur J Gynaecol Oncol 26, 589–598. [PubMed] [Google Scholar]
- [80].Thoma C, 2018. Prostate cancer: Radiosensitized by STAT5 blockade. Nat Rev Urol 15, 203. [DOI] [PubMed] [Google Scholar]
- [81].Kamran MZ, Patil P, Gude RP, 2013. Role of STAT3 in cancer metastasis and translational advances. BioMed research international 2013, 421821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Barry SP, Townsend PA, Knight RA, Scarabelli TM, Latchman DS, Stephanou A, 2010. STAT3 modulates the DNA damage response pathway. Int J Exp Pathol 91, 506–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Gao B, Shen X, Kunos G, Meng Q, Goldberg ID, Rosen EM, Fan S, 2001. Constitutive activation of JAK-STAT3 signaling by BRCA1 in human prostate cancer cells. FEBS Lett 488, 179–184. [DOI] [PubMed] [Google Scholar]
- [84].Sato H, Niimi A, Yasuhara T, Permata TBM, Hagiwara Y, Isono M, Nuryadi E, Sekine R, Oike T, Kakoti S, Yoshimoto Y, Held KD, Suzuki Y, Kono K, Miyagawa K, Nakano T, Shibata A, 2017. DNA double-strand break repair pathway regulates PD-L1 expression in cancer cells. Nat Commun 8, 1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Weinandy A, Piroth MD, Goswami A, Nolte K, Sellhaus B, Gerardo-Nava J, Eble M, Weinandy S, Cornelissen C, Clusmann H, Luscher B, Weis J, 2014. Cetuximab induces eme1-mediated DNA repair: a novel mechanism for cetuximab resistance. Neoplasia 16, 207–220, 20 e1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Wehinger J, Gouilleux F, Groner B, Finke J, Mertelsmann R, Weber-Nordt RM, 1996. IL-10 induces DNA binding activity of three STAT proteins (Stat1, Stat3, and Stat5) and their distinct combinatorial assembly in the promoters of selected genes. FEBS Lett 394, 365–370. [DOI] [PubMed] [Google Scholar]
- [87].Shao N, Xu B, Mi YY, Hua LX, 2011. IL-10 polymorphisms and prostate cancer risk: a meta-analysis. Prostate Cancer Prostatic Dis 14, 129–135. [DOI] [PubMed] [Google Scholar]
- [88].Cokic VP, Mitrovic-Ajtic O, Beleslin-Cokic BB, Markovic D, Buac M, Diklic M, Kraguljac-Kurtovic N, Damjanovic S, Milenkovic P, Gotic M, Raj PK, 2015. Proinflammatory Cytokine IL-6 and JAK-STAT Signaling Pathway in Myeloproliferative Neoplasms. Mediators Inflamm 2015, 453020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Culig Z, Puhr M, 2012. Interleukin-6: a multifunctional targetable cytokine in human prostate cancer. Mol Cell Endocrinol 360, 52–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Chen X, Chen F, Ren Y, Weng G, Xu L, Xue X, Keng PC, Lee SO, Chen Y, 2019. IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2. J Cancer Res Clin Oncol 145, 1471–1484. [DOI] [PubMed] [Google Scholar]
- [91].Rusthoven CG, Carlson JA, Waxweiler TV, Raben D, Dewitt PE, Crawford ED, Maroni PD, Kavanagh BD, 2014. The impact of definitive local therapy for lymph node-positive prostate cancer: a population-based study. Int J Radiat Oncol Biol Phys 88, 1064–1073. [DOI] [PubMed] [Google Scholar]
- [92].Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, Borbath I, Bouche O, Shannon J, Andre T, Mineur L, Chibaudel B, Bonnetain F, Louvet C, Group LAPT, 2016. Effect of Chemoradiotherapy vs Chemotherapy on Survival in Patients With Locally Advanced Pancreatic Cancer Controlled After 4 Months of Gemcitabine With or Without Erlotinib: The LAP07 Randomized Clinical Trial. JAMA 315, 1844–1853. [DOI] [PubMed] [Google Scholar]
- [93].Brantley EC, Nabors LB, Gillespie GY, Choi YH, Palmer CA, Harrison K, Roarty K, Benveniste EN, 2008. Loss of protein inhibitors of activated STAT-3 expression in glioblastoma multiforme tumors: implications for STAT-3 activation and gene expression. Clin Cancer Res 14, 4694–4704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, European Organisation for R, Treatment of Cancer Brain T, Radiotherapy G, National Cancer Institute of Canada Clinical Trials G, 2005. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352, 987–996. [DOI] [PubMed] [Google Scholar]
- [95].Tang S, Yuan X, Song J, Chen Y, Tan X, Li Q, 2019. Association analyses of the JAK/STAT signaling pathway with the progression and prognosis of colon cancer. Oncol Lett 17, 159–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Verstovsek S, Vannucchi AM, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Kirito K, Besses C, Hino M, Moiraghi B, Miller CB, Cazzola M, Rosti V, Blau I, Mesa R, Jones MM, Zhen H, Li J, Francillard N, Habr D, Kiladjian JJ, 2016. Ruxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trial. Haematologica 101, 821–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Bird P, Hall S, Nash P, Connell CA, Kwok K, Witcombe D, Thirunavukkarasu K, 2019. Treatment outcomes in patients with seropositive versus seronegative rheumatoid arthritis in Phase III randomised clinical trials of tofacitinib. RMD Open 5, e000742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Markham A, Keam SJ, 2019. Peficitinib: First Global Approval. Drugs 79, 887–891. [DOI] [PubMed] [Google Scholar]
- [99].Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, Masszi T, Mishchenko E, Jourdan E, Vannucchi AM, Drummond MW, Jurgutis M, Kuliczkowski K, Gheorghita E, Passamonti F, Neumann F, Patki A, Gao G, Tefferi A, 2015. Safety and Efficacy of Fedratinib in Patients With Primary or Secondary Myelofibrosis: A Randomized Clinical Trial. JAMA Oncol 1, 643–651. [DOI] [PubMed] [Google Scholar]
- [100].Burmester GR, Kremer JM, Van den Bosch F, Kivitz A, Bessette L, Li Y, Zhou Y, Othman AA, Pangan AL, Camp HS, 2018. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 391, 2503–2512. [DOI] [PubMed] [Google Scholar]
- [101].Westhovens R, Taylor PC, Alten R, Pavlova D, Enriquez-Sosa F, Mazur M, Greenwald M, Van der Aa A, Vanhoutte F, Tasset C, Harrison P, 2017. Filgotinib (GLPG0634/GS-6034), an oral JAK1 selective inhibitor, is effective in combination with methotrexate (MTX) in patients with active rheumatoid arthritis and insufficient response to MTX: results from a randomised, dose-finding study (DARWIN 1). Ann Rheum Dis 76, 998–1008. [DOI] [PubMed] [Google Scholar]
- [102].Berdeja J, Palandri F, Baer MR, Quick D, Kiladjian JJ, Martinelli G, Verma A, Hamid O, Walgren R, Pitou C, Li PL, Gerds AT, 2018. Phase 2 study of gandotinib (LY2784544) in patients with myeloproliferative neoplasms. Leuk Res 71, 82–88. [DOI] [PubMed] [Google Scholar]
- [103].Knapper S, Russell N, Gilkes A, Hills RK, Gale RE, Cavenagh JD, Jones G, Kjeldsen L, Grunwald MR, Thomas I, Konig H, Levis MJ, Burnett AK, 2017. A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML. Blood 129, 1143–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S, 2018. Pacritinib vs Best Available Therapy, Including Ruxolitinib, in Patients With Myelofibrosis: A Randomized Clinical Trial. JAMA Oncol 4, 652–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Mesa RA, Kiladjian JJ, Catalano JV, Devos T, Egyed M, Hellmann A, McLornan D, Shimoda K, Winton EF, Deng W, Dubowy RL, Maltzman JD, Cervantes F, Gotlib J, 2017. SIMPLIFY-1: A Phase III Randomized Trial of Momelotinib Versus Ruxolitinib in Janus Kinase Inhibitor-Naive Patients With Myelofibrosis. J Clin Oncol 35, 3844–3850. [DOI] [PMC free article] [PubMed] [Google Scholar]