Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Therapeutic targeting of STAT3 pathways in pancreatic adenocarcinoma: A systematic review of clinical and preclinical literature

  • Sarah Peisl,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Surgery, HFR Fribourg, Fribourg, Switzerland

  • Claudia Mellenthin,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Project administration, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliation Department of Surgery, HFR Fribourg, Fribourg, Switzerland

  • Lucie Vignot,

    Roles Conceptualization, Validation, Writing – review & editing

    Affiliation Department of Oncology, HFR Fribourg, Fribourg, Switzerland

  • Carmen Gonelle-Gispert,

    Roles Conceptualization, Validation, Writing – review & editing

    Affiliation Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland

  • Leo Bühler,

    Roles Conceptualization, Supervision, Validation, Writing – review & editing

    Affiliation Department of Surgery, HFR Fribourg, Fribourg, Switzerland

  • Bernhard Egger

    Roles Conceptualization, Supervision, Validation, Writing – review & editing

    bernhard-egger@bluewin.ch

    Affiliations Department of Surgery, HFR Fribourg, Fribourg, Switzerland, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland

Abstract

Background/Objectives

Pancreatic ductal adenocarcinoma is a highly lethal disease with increasing incidence. Due to high resistance, chemo/radiotherapy has limited success in pancreatic cancer and only marginally prolongs patient survival. Therefore, novel biomarkers and therapeutic targets are needed. In the present review, we performed a comprehensive summary of therapeutic approaches targeting the GP130/JAK/STAT3 pathway.

Methods

We systematically reviewed the PubMed and Embase databases for preclinical and clinical studies, from inception to October 4, 2020, on drugs targeting the GP130/JAK/STAT3 pathway. Bias assessments and qualitative analyses were performed.

Results

Twenty-five preclinical and nine clinical trials were included in the review. All preclinical studies reported a favorable outcome in terms of pancreatic ductal adenocarcinoma progression. Futhermore, drugs targeting the GP130/JAK/STAT3 pathway were shown to be efficient chemosensitizers. However, high publication bias was assumed. In the clinical setting, bazedoxifene and itacitinib improved patient outcomes.

Conclusion

Preclinical studies strongly suggest significant efficacy of drugs targeting GP130/JAK/STAT3 in the treatment of pancreatic ductal adenocarcinoma and that these molecules are effective chemosensitizers. Though only a few trials have shown the efficacy in a clinical setting, the STAT3 pathway remains a promising drug target for future treatment of pancreatic ductal adenocarcinoma and may help overcome chemotherapy resistance.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease with increasing incidence. In most cases, pancreatic cancer presents at an advanced stage, with only 20% of all cases undergoing surgical resection. In terms of prognostic outcomes for patients, pancreatic adenocarcinoma ranks last, with an overall 5-year survival rate of 2–9% [1, 2]. Even though the management of pancreatic adenocarcinoma is evolving with the introduction of novel surgical techniques and medical therapies, only minor improvements in outcomes have been achieved. Due to high resistance, chemotherapy and radiotherapy have limited success in metastatic PDAC and only marginally prolong patient survival [3]. Current treatment options for metastatic PDAC are modified FOLFIRINOX/FOLFIRINOX or nab-paclitaxel and gemcitabine in patients with good performance status, and gemcitabine with or without a second agent for those with poor performance status [4]. Most recently, trials studying the update of immunotherapy in PDAC were negative except in a subgroup of adenocarcinoma with microsatellite instability [5].

Considering the lack of effective treatment, the identification of novel biomarkers and therapeutic targets is fundamental to developing new treatment strategies and improving clinical outcomes. Recent studies suggest that signaling pathways involving STAT3 play a key role in tumorigenesis, progression and drug resistance in several human malignancies such as leukemia, lymphomas as well as solid tumors such as hepatocelullar carcinoma, esophageal, lung, prostate, bladder and breast cancer [6, 7]. Animal models of PDAC have shown that STAT3 is an important regulator of stem cell self-renewal and cancer cell survival [8, 9]. Upregulation of STAT3 has been shown to promote the development of PDAC from pancreatic intraepithelial neoplasia [10, 11], as well as pro-metastatic niche formation in the liver [12]. Furthermore, STAT3 has been shown to mediate resistance to chemotherapy and to be associated with adverse outcomes following resection of PDAC with curative intent [1315].

As illustrated in Fig 1, IL-6-type cytokines (IL-6, IL-10, IL-11, Leukemia inhibitory factor (LIF), Cardiotrophin-1 (CT-1), Oncostatin-M (OSM), Ciliary neurotrophic factor (CNTF)), bind glycoprotein-130 (GP130) and activate janus kinase (JAK), which in turn phosphorylates STAT3, among other signaling mediators in PDAC tumor cells as well as cells of tumor microenvironment (TME) [16]. TME in PDAC is a complex system which consists, along with extensive stromal networks, of different cell components such as pancreatic stellate cells (PSCs), cancer associated fibroblasts (CAFs), tumor associated macrophages (TAMs), mast cells, regulatory T-cells and myeloid derived suppressor cells (MDSCs), synergizing to support tumor progression, immune evasion and metastatic spreading. Interactions between different cells within the TME are mediated through signaling molecules such as STAT3 activation via IL-6-type cytokines. For instance, PDAC tumor cells can stimulate immune cells to secrete IL-6-type cytokines, supporting the development of immunosuppressive TAMs and MDSCs as well as the activation of PSCs and CAFs, which in turn induce the secretion of inflammatory cytokines through positive feedback loops [11, 1722]. Thus, STAT3 activation drives immune cells towards immunosuppressive phenotype by inhibiting regulatory T-cells, which in turn sustains tumor immune evasion. Furthermore, the phosphorylation of STAT3 leads to enhanced transcription of downstream target genes, which promote angiogenesis, invasion, and epithelial-mesenchymal transition (EMT) [23].

thumbnail
Fig 1. Schematic presentation of IL-6/JAK/STAT3 pathway in pancreatic cancer cells and tumor microenvironment.

(PSC: pancreatic stellate cell, CAF: cancer associated fibroblast, TAM: tumor associated macrophages, MDSC: myeloid derived suppressor cells).

https://doi.org/10.1371/journal.pone.0252397.g001

Accordingly, pathways involving STAT3 appear to be promising drug targets for the treatment of PDAC. In particular, IL-6 has been shown to be a potentially efficient therapeutic approach for overcoming chemotherapy resistance. The purpose of this study was to provide a comprehensive summary of therapeutic approaches targeting the GP130/JAK/STAT3 pathway in pancreatic adenocarcinoma through a systematic qualitative review of the literature.

Methods

This systematic review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) [24]. Studies were identified by searching PubMed using the following search terms:

  1. (carcinoma, pancreatic ductal[MeSH Terms]) AND (interleukin-6[MeSH Terms])
  2. (carcinoma, pancreatic ductal[MeSH Terms]) AND (jak 2 protein tyrosine kinase[MeSH Terms])
  3. (carcinoma, pancreatic ductal[MeSH Terms]) AND (jak 1 protein tyrosine kinase[MeSH Terms])
  4. (carcinoma, pancreatic ductal[Mesh]) AND (stat3 transcription factor[MeSH Terms])
  5. (carcinoma, pancreatic ductal[MeSH Terms]) AND (gp130, cytokine receptor[MeSH Terms])

Embase was searched using the following search query:

  1. (‘pancreas adenocarcinoma’/exp OR ‘adenocarcinoma, pancreas’ OR ‘pancreatic adenocarcinoma’ OR ‘pancreatic ductal adenocarcinoma’) AND (‘stat3 protein’/exp OR ‘stat3 protein’ OR ‘stat3 transcription factor’ OR ‘protein stat3’ OR ‘signal transducer and activator of transcription 3’ OR ‘stat3’ OR ‘transcription factor stat3’)
  2. (‘pancreas adenocarcinoma’/exp AND ‘interleukin 6’/exp, filter for articles)
  3. (‘pancreas cancer’/exp AND ‘janus kinase 2’/exp, filter for articles)
  4. (‘janus kinase 1’/exp AND ‘pancreas cancer’/exp, filter for articles)
  5. (‘gp130’/exp AND ‘pancreas cancer’/exp, filter for articles)

The electronic search was supplemented by a manual search of the reference lists of relevant articles to identify any studies that may have been missed in the database searches. The original database search was performed on May 5, 2020. The electronic search was updated on October 4, 2020.

Inclusion criteria were defined as all trials studying the pharmacological targeting of the GP130-related cytokine/JAK/STAT3 pathway in pancreatic cancer, including studies on animal models or cell cultures. Only studies with an English abstract were included. Reviews, comments, and conference or meeting abstracts were excluded from the analysis. No restrictions on publication date or publication status were imposed.

After exclusion of duplicates, records identified from the literature search were screened for eligibility independently by the two main authors using the title and abstract in an unblinded manner. Disagreements between the reviewers were resolved by consensus. The full-text of articles meeting the inclusion criteria was assessed by the two main authors and reevaluated for the inclusion criteria. Disagreements were, again, resolved by consensus.

We extracted data using a previously prepared extraction form. The information from each included study on the study design, characteristics of analyzed subjects or trial participants, characteristics of the pharmacological agent studied, type of outcome measures, and outcomes was tabulated. On this basis, we performed a qualitative data synthesis.

We performed a quality assessment of clinical trials according to the ROB tool, which was adapted to match non-randomized clinical trials [25]. For preclinical studies, we used the SYRCLE’s risk of bias tool, which was adapted to match in vivo and in vitro studies [26]. Results were displayed in an analogous fashion as suggested by Higgins et al for systematic reviews of interventions [25]. Bias assessment was conducted for every study by two independent assessors and disagreements resolved by consensus.

Due to the nature of this study, approval from the local Ethics Committee was not required.

Results

Study selection

Our search identified 756 records through the database searches (Embase, Pubmed) and the manual search of the reference lists of relevant articles. Initial screening excluded 689 records, including 145 duplicates. The remaining 67 articles were assessed based on the full text, 29 of which were found to be ineligible due to absence of a tested pharmacological substance or the absence of GP130-related cytokine/JAK/STAT3 pathway targeting.

A summary of the study selection process is provided in Fig 2. Ultimately, 38 studies were included in the review, including 4 ongoing trials. All included studies were published in English and no unpublished data were included. No other studies were identified through the electronic search update on October 4, 2020.

Bias assessment

Table 1 shows the risk of bias assessment for the preclinical studies. Preclinical studies had strong limitations to rigorous bias assessment because few provided sufficient details regarding selection and performance bias. Study protocols were not published beforehand, so a comparison between intended interventions and published interventions was not possible. In animal trials, few studies explicitly stated a randomization process for treatment groups, and treatment results were often assessed manually with semi-quantitative methods. This lack of reporting makes it difficult to accurately determine the risk of bias of the preclinical studies. However, more details were available on the risk of attrition bias, reporting bias, and other bias.

The quality assessment of the included clinical trials is provided in Table 2. The overall quality of the studies was good, with only one study presenting high risk of selection bias.

Preclinical studies

As summarized in Table 3, 25 of the included studies were preclinical trials testing 20 substances targeting the GP130/JAK/STAT3-pathway. Twenty-four studies performed in vitro experiments using human pancreatic cancer cells [17, 2732, 3436, 3841, 4347, 49, 50, 60, 61]. In vivo experiments were performed in 17 studies using mouse xenograft tumor models (n = 14) [28, 31, 34, 35, 38, 4042, 44, 45, 4850, 60], chicken chorio-allantoic membrane xenograft tumor models (n = 1) [32], or KPC mice (n = 2) [33, 45]. All studies reported favorable outcomes in terms of pancreatic cancer cell viability, proliferation, migration, colony formation ability, apoptosis, or effects on downstream target genes, as well as tumor growth, tumor volume, or weight in in vivo models. Eight studies analyzed the combinational effect of the investigated drug with chemotherapy (i.e., gemcitabine, paclitaxel, 5-fluorouracil, and oxaliplatin) [31, 33, 38, 40, 41, 45, 49, 60]. In all studies, the inhibitory effect on pancreatic cancer cells by the investigated drug was enhanced by chemotherapy.

thumbnail
Table 3. Characteristics of the included preclinical studies.

https://doi.org/10.1371/journal.pone.0252397.t003

Clinical studies

As summarized in Table 4, nine of the studies were clinical trials including 880 individuals and assessing 5 drugs. One study performed a retrospective analysis of bazedoxifene, an inhibitor of the IL-6/IL-6R/GP130 complex, in patients with pancreatic (n = 5) or gastric adenocarcinoma (n = 2), showing biological tumor marker reduction in 80% and disease regression on PET-CT in 60% of cases [51]. Icatinib, a selective JAK1 inhibitor, was tested in combination with nab-paclitaxel and gemcitabine, showing a synergistic effect with an overall response rate of 24% with an acceptable safety profile in a phase 1b/2 study [52]. However, this study was terminated early due to negative phase 3 results for JAK1/2 inhibitor ruxolitinib [55]. Momelotinib, a JAK1/2 inhibitor, resulted in a partial response in 28% of patients with previously untreated metastatic PDAC (n = 25) in a phase 1 study. However, no significant difference was reported from treatment with paclitaxel and gemcitabine [53]. Ruxolitinib, a JAK1/2 inhibitor, has been investigated in phase 1b, 2, and 3 clinical trials in combination with capecitabine, gemcitabine, and paclitaxel, revealing no significant difference in overall survival or progression-free survival in patients with PDAC [5456]. Finally, phase 2 and 3 studies have been performed assessing tipifarnib, an inhibitor of STAT3 phosphorylation that showed no single-agent antitumor activity and no difference in overall survival in combination with gemcitabine [5759]. Four ongoing clinical trials were found, involving tocilizumab, an anti-IL6Rα antibody with favorable results in preclinical studies [31, 33], and napabucasin, a STAT3 inhibitor that is also under investigation in colorectal cancer [63].

Discussion

The present systematic review of 25 preclinical studies and 9 clinical trials revealed a good overall effect of the investigated drugs targeting the GP130/JAK/STAT3 pathway in the treatment of PDAC. Table 5 summarizes the outcome and the state of research for each assessed drug. Favorable outcomes have been reported for all 20 drugs investigated in a preclinical setting. Even though these substances appear promising in the treatment of PDAC, only five of these drugs have been investigated in clinical trials. Favorable outcomes and acceptable toxicity profiles have been found in studies investigating bazedoxifene and itacitinib [51, 52]. Notably, bazedoxifene is already approved for the treatment of osteoporosis [64], and itacitinib has been shown to have great potential in recent clinical trials studying the treatment of connective tissue diseases and graft-versus-host disease, among others [6567].

Even though the PDAC tumor micro-environment (TME) has been shown to be a promising target for improving PDAC treatment, none of the included studies in this systematic review examined the influence of the analyzed substances on stromal or immune cells.

The TME plays an important role in tumorigenesis and chemoresistance by close interaction with tumor cells. Furthermore, TME has been shown to be highly immunosuppressive, promoting immune evasion, hence sustaining tumor progression [1820]. Immunotherapy, so far, has not demonstrated substantial clinical improvement as single agent in the treatment of PDAC [5]. Therefore, strategies simultaneously targeting PDAC tumor cells as well as different immune checkpoints might be needed. The interactions of PDAC tumor cells and different cells within the TME such as CAFs, MDSCs, TAMs, are mediated through GP130/JAK/STAT3 pathway [11, 1720, 68]. STAT3 inhibition might thus have consequences in shaping TME towards anti-tumor phenotype by acting on both immune and tumor cells [1820]. In combination with chemotherapeutic agents and immunotherapy, it might significantly increase therapeutic efficacy in the treatment of PDAC.

Recent studies have shown the important role of the STAT3 pathway in tumorigensis, as well as the STAT3-mediated resistance to chemotherapy in in vivo models of PDAC [815]. The results from the preclinical trials presented in this review confirmed the importance of the GP130/JAK/STAT3 pathway in PDAC and its role as a possible drug target. Furthermore, several of the studies showed a synergy between the investigational drug and chemotherapy, such as gemcitabine, paclitaxel, 5-fluorouracil, and oxaliplatin [31, 33, 38, 40, 41, 45, 49, 5355, 57, 60]. However, to the best of our knowledge, drugs targeting GP130/JAK/STAT3 have never been studied as chemosensitizers in addition to the currently emerging FOLFIRINOX regimen [4]. Even though some promising outcomes have been shown in clinical trials [51, 52], several studies were terminated prematurely due to high progression rates and futility. This may reflect the difficulty showing a significant benefit in patients presenting with PDAC, as it is known to be a highly lethal disease that is often diagnosed at an advanced stage and has a poor prognosis with an overall 5-year survival rate of 2–9% [1, 2].

The discrepancy between preclinical and clinical data may also result from the fact that, in contrast to the preclinical studies, the clinical trials did not verify the activation of the STAT3 pathway in PDAC. The benefit of targeted GP130/JAK/STAT3 therapy may be increased by selecting patients with previously known STAT3 pathway activation in PDAC cells.

The present systematic review included all preclinical and clinical trials of drugs targeting the GP130/JAK/STAT3 pathway. Furthermore, we searched for ongoing, unpublished trials, leading to a thorough analysis of the current state of research. However, because all published preclinical studies reported a positive outcome, we suspect that several negative studies may not have been published and concluded relevant publication bias, leading to an overestimation of the effect of GP130/JAK/STAT3-targeting drugs in the treatment of PDAC in the preclinical setting. Furthermore, the substantial heterogeneity among the preclinical and clinical studies did not allow a quantitative analysis or measurement of the effect size.

Conclusion

Preclinical studies strongly suggest significant efficacy of drugs targeting GP130/JAK/STAT3 in the treatment of PDAC and that these molecules are effective chemosensitizers, possibly through simultaneous effect on tumor cells and TME. Though only a few trials have shown the efficacy in a clinical setting, the GP130/JAK/STAT3 pathway remains a promising drug target for the development of future treatments for PDAC and may help overcome chemotherapy resistance.

References

  1. 1. Yeo TP. Demographics, Epidemiology, and Inheritance of Pancreatic Ductal Adenocarcinoma. Semin Oncol. 2015 Feb;42(1):8–18. pmid:25726048
  2. 2. Baugh KA, Tran Cao HS, van Buren G, Silberfein EJ, Hsu C, Chai C, et al. Understaging of clinical stage I pancreatic cancer and the impact of multimodality therapy. Surgery. 2019 Feb;165(2):307–14. pmid:30243481
  3. 3. McGuigan A, Kelly P, Turkington RC, Jones C, Coleman HG, McCain RS. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol. 2018 Nov 21;24(43):4846–61. pmid:30487695
  4. 4. Singh RR, O’Reilly EM. New Treatment Strategies for Metastatic Pancreatic Ductal Adenocarcinoma. Drugs. 2020 May 1;80(7):647–69. pmid:32306207
  5. 5. Schizas D, Charalampakis N, Kole C, Economopoulou P, Koustas E, Gkotsis E, et al. Immunotherapy for pancreatic cancer: A 2020 update. Cancer Treat Rev. 2020 Jun;86:102016. pmid:32247999
  6. 6. Lee JH, Mohan CD, Deivasigamani A, Jung YY, Rangappa S, Basappa S, et al. Brusatol suppresses STAT3-driven metastasis by downregulating epithelial-mesenchymal transition in hepatocellular carcinoma. J Adv Res. 2020 Nov;26:83–94. pmid:33133685
  7. 7. Garg M, Shanmugam MK, Bhardwaj V, Goel A, Gupta R, Sharma A, et al. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med Res Rev. 2020 Dec 1; pmid:33289118
  8. 8. Corcoran RB, Contino G, Deshpande V, Tzatsos A, Conrad C, Benes CH, et al. STAT3 Plays a Critical Role in KRAS-Induced Pancreatic Tumorigenesis. Cancer Res. 2011 Jul 15;71(14):5020–9. pmid:21586612
  9. 9. Öhlund D, Handly-Santana A, Biffi G, Elyada E, Almeida AS, Ponz-Sarvise M, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med. 2017;214(3):579–96. pmid:28232471
  10. 10. Gruber R, Panayiotou R, Nye E, Spencer-Dene B, Stamp G, Behrens A. YAP1 and TAZ Control Pancreatic Cancer Initiation in Mice by Direct Up-regulation of JAK–STAT3 Signaling. Gastroenterology. 2016 Sep;151(3):526–39. pmid:27215660
  11. 11. Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Klöppel G, et al. Stat3/Socs3 Activation by IL-6 Transsignaling Promotes Progression of Pancreatic Intraepithelial Neoplasia and Development of Pancreatic Cancer. Cancer Cell. 2011 Apr;19(4):456–69. pmid:21481788
  12. 12. Lee JW, Stone ML, Porrett PM, Thomas SK, Komar CA, Li JH, et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature. 2019 Mar;567(7747):249–52. pmid:30842658
  13. 13. Tan F, Putoczki T, Stylli S, Luwor R. The Role of STAT3 Signaling in Mediating Tumor Resistance to Cancer Therapy. Curr Drug Targets. 2014 Dec 16;15(14):1341–53. pmid:25410411
  14. 14. Denley SM, Jamieson NB, McCall P, Oien KA, Morton JP, Carter CR, et al. Activation of the IL-6R/Jak/Stat Pathway is Associated with a Poor Outcome in Resected Pancreatic Ductal Adenocarcinoma. J Gastrointest Surg. 2013 May;17(5):887–98. pmid:23435739
  15. 15. Xing H-B, Tong M-T, Wang J, Hu H, Zhai C-Y, Huang C-X, et al. Suppression of IL-6 Gene by shRNA Augments Gemcitabine Chemosensitization in Pancreatic Adenocarcinoma Cells. BioMed Res Int. 2018;2018:1–10. pmid:29693005
  16. 16. Xu S, Neamati N. Gp130: A promising drug target for cancer therapy. Expert Opin Ther Targets. 2013;17(11):1303–28. pmid:24099136
  17. 17. Chen J, Wang S, Su J, Chu G, You H, Chen Z, et al. Interleukin-32α inactivates JAK2/STAT3 signaling and reverses interleukin-6-induced epithelial-mesenchymal transition, invasion, and metastasis in pancreatic cancer cells. OncoTargets Ther. 2016;9:4225–37.
  18. 18. van Duijneveldt G, Griffin MDW, Putoczki TL. Emerging roles for the IL-6 family of cytokines in pancreatic cancer. Clin Sci. 2020 Aug 28;134(16):2091–115. pmid:32808663
  19. 19. Weber R, Groth C, Lasser S, Arkhypov I, Petrova V, Altevogt P, et al. IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021 Jan;359:104254. pmid:33296753
  20. 20. Trovato R, Fiore A, Sartori S, Canè S, Giugno R, Cascione L, et al. Immunosuppression by monocytic myeloid-derived suppressor cells in patients with pancreatic ductal carcinoma is orchestrated by STAT3. J Immunother Cancer. 2019 Dec;7(1):255. pmid:31533831
  21. 21. Wolf J, Rose-John S, Garbers C. Interleukin-6 and its receptors: A highly regulated and dynamic system. Cytokine. 2014 Nov;70(1):11–20. pmid:24986424
  22. 22. Jin G, Hong W, Guo Y, Bai Y, Chen B. Molecular Mechanism of Pancreatic Stellate Cells Activation in Chronic Pancreatitis and Pancreatic Cancer. J Cancer. 2020;11(6):1505–15. pmid:32047557
  23. 23. Chang Q, Bournazou E, Sansone P, Berishaj M, Gao SP, Daly L, et al. The IL-6/JAK/Stat3 feed-forward loop drives tumorigenesis and metastasis. Neoplasia U S. 2013;15(7):848–62. pmid:23814496
  24. 24. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. Int J Surg. 2010;8(5):336–41. pmid:20171303
  25. 25. Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011 Oct 18;343:d5928–d5928. pmid:22008217
  26. 26. Hooijmans CR, Rovers MM, de Vries RB, Leenaars M, Ritskes-Hoitinga M, Langendam MW. SYRCLE’s risk of bias tool for animal studies. BMC Med Res Methodol. 2014 Mar 26;14(1):43. pmid:24667063
  27. 27. Chen X, Tian J, Su GH, Lin J. Blocking IL-6/GP130 Signaling Inhibits Cell Viability/ Proliferation, Glycolysis, and Colony Forming Activity in Human Pancreatic Cancer Cells. Curr Cancer Drug Targets. 2019;19(5):417–27. pmid:29714141
  28. 28. Edderkaoui M, Lugea A, Hui H, Eibl G, Lu QY, Moro A, et al. Ellagic acid and embelin affect key cellular components of pancreatic adenocarcinoma, cancer, and stellate cells. Nutr Cancer. 2013;65(8):1232–44. pmid:24127740
  29. 29. Fu S, Lin J. Blocking interleukin-6 and interleukin-8 signaling inhibits cell viability, colony-forming activity, and cell migration in human triple-negative breast cancer and pancreatic cancer cells. Anticancer Res. 2018;38(11):6271–9. pmid:30396947
  30. 30. Ge Y, Yang B, Chen Z, Cheng R. Cryptotanshinone suppresses the proliferation and induces the apoptosis of pancreatic cancer cells via the STAT3 signaling pathway. Mol Med Rep. 2015;12(5):7782–8. pmid:26459366
  31. 31. Goumas FA, Holmer R, Egberts JH, Gontarewicz A, Heneweer C, Geisen U, et al. Inhibition of IL-6 signaling significantly reduces primary tumor growth and recurrencies in orthotopic xenograft models of pancreatic cancer. Int J Cancer. 2015;137(5):1035–46. pmid:25604508
  32. 32. Lin L, Hutzen B, Zuo M, Ball S, Deangelis S, Foust E, et al. Novel STAT3 phosphorylation inhibitors exhibit potent growth-suppressive activity in pancreatic and breast cancer cells. Cancer Res. 2010;70(6):2445–54. pmid:20215512
  33. 33. Long KB, Tooker G, Tooker E, Luque SL, Lee JW, Pan X, et al. IL6 receptor blockade enhances chemotherapy efficacy in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2017;16(9):1898–908. pmid:28611107
  34. 34. Palagani V, Bozko P, El khatib M, Belahmer H, Giese N, Sipos B, et al. Combined inhibition of Notch and JAK/STAT is superior to monotherapies and impairs pancreatic cancer progression. Carcinogenesis. 2014;35(4):859–66. pmid:24293409
  35. 35. Sahu N, Chan E, Chu F, Pham T, Koeppen H, Forrest W, et al. Cotargeting of MEK and PDGFR/STAT3 pathways to treat pancreatic ductal adenocarcinoma. Vol. 16, Molecular Cancer Therapeutics. 2017. 1729 p. pmid:28619758
  36. 36. Sun C, Zhang M, Shan X, Zhou X, Yang J, Wang Y, et al. Inhibitory effect of cucurbitacin E on pancreatic cancer cells growth via STAT3 signaling. J Cancer Res Clin Oncol. 2010 Apr;136(4):603–10. pmid:19816711
  37. 37. Thoennissen NH, Iwanski GB, Doan NB, Okamoto R, Lin P, Abbassi S, et al. Cucurbitacin B induces apoptosis by inhibition of the JAK/STAT pathway and potentiates antiproliferative effects of gemcitabine on pancreatic cancer cells. Cancer Res. 2009;69(14):5876–84. pmid:19605406
  38. 38. Wu X, Cao Y, Xiao H, Li C, Lin J. Bazedoxifene as a novel GP130 inhibitor for pancreatic cancer therapy. Mol Cancer Ther. 2016;15(11):2609–19. pmid:27535971
  39. 39. Zhang X, Lu H, Hong W, Liu L, Wang S, Zhou M, et al. Tyrphostin B42 attenuates trichostatin A-mediated resistance in pancreatic cancer cells by antagonizing IL-6/JAK2/STAT3 signaling. Oncol Rep. 2018;39(4):1892–900. pmid:29393444
  40. 40. Nagaraju GP, Mezina A, Shaib WL, Landry J, El-Rayes BF. Targeting the Janus-activated kinase-2-STAT3 signalling pathway in pancreatic cancer using the HSP90 inhibitor ganetespib. Eur J Cancer. 2016;52:109–19. pmid:26682870
  41. 41. Nagaraju GP, Zakka KM, Landry JC, Shaib WL, Lesinski GB, El-Rayes BF. Inhibition of HSP90 overcomes resistance to chemotherapy and radiotherapy in pancreatic cancer. Int J Cancer. 2019 Sep 15;145(6):1529–37. pmid:30801702
  42. 42. Lu D, Qin Q, Lei R, Hu B, Qin S. Targeted blockade of interleukin 9 inhibits tumor growth in murine model of pancreatic cancer. Adv Clin Exp Med. 2019;28(10):1285–92. pmid:31647203
  43. 43. Liu A, Yan L, Pui-Kai L, Chenglong L, Lin J. LLL12 Inhibits Endogenous and Exogenous Interleukin-6- induced STAT3 Phosphorylation in Human Pancreatic Cancer Cells Aiguo. Anticancer Res. 2011;(1).
  44. 44. Huang HL, Chao MW, Chen CC, Cheng CC, Chen MC, Lin CF, et al. LTP-1, a novel antimitotic agent and Stat3 inhibitor, inhibits human pancreatic carcinomas in vitro and in vivo. Sci Rep. 2016;6(January):1–11. pmid:27278358
  45. 45. Luo D, D M.G., Wei R, LComb JL, Williams BR, Macken . Phospho-valproic acid (MDC-1112) reduces pancreatic cancer growth in patient-derived tumor xenografts and KPC mice: enhanced efficacy when combined with gemcitabine. 2019;40:1–30.
  46. 46. Kim C, Kim JH, Oh EY, Nam D, Lee SG, Lee J, et al. Blockage of STAT3 signaling pathway by morusin induces apoptosis and inhibits invasion in human pancreatic tumor cells. Pancreas. 2016;45(3):409–19. pmid:26646273
  47. 47. Venkatasubbarao K, Choudary A, Freeman JW. Farnesyl transferase inhibitor (R115777)-induced inhibition of STAT3 (Tyr705) phosphorylation in human pancreatic cancer cell lines require extracellular signal-regulated kinases. Cancer Res. 2005;65(7):2861–71. pmid:15805288
  48. 48. Lu C, Talukder A, Savage NM, Singh N, Liu K. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. OncoImmunology. 2017;6(3):1–15. pmid:28405527
  49. 49. Liu Y, Bi T, Liu L, Gao Q, Shen G, Qin L. S-Adenosylmethionine synergistically enhances the antitumor effect of gemcitabine against pancreatic cancer through JAK2/STAT3 pathway. Naunyn Schmiedebergs Arch Pharmacol. 2019;392(5):615–22. pmid:30683944
  50. 50. Song Y, Gao L, Tang Z, Li H, Sun B, Chu P, et al. Anticancer effect of SZC015 on pancreatic cancer via mitochondria-dependent apoptosis and the constitutive suppression of activated nuclear factor κB and STAT3 in vitro and in vivo. J Cell Physiol. 2018;234(1):777–88. pmid:30078206
  51. 51. Burkhardt C, Bühler L, Tihy M, Morel P, Forni M. Bazedoxifene as a novel strategy for treatment of pancreatic and gastric adenocarcinoma. Oncotarget. 2019;10(34):3198–202. pmid:31139333
  52. 52. Beatty GL, Shahda S, Beck T, Uppal N, Cohen SJ, Donehower R, et al. A Phase Ib/II Study of the JAK1 Inhibitor, Itacitinib, plus nab -Paclitaxel and Gemcitabine in Advanced Solid Tumors. The Oncologist. 2019;24(1):14–14. pmid:30115734
  53. 53. Ng K, Hendifar A, Starodub A, Chaves J, Yang Y, Koh B, et al. Phase 1 dose-escalation study of momelotinib, a Janus kinase 1/2 inhibitor, combined with gemcitabine and nab-paclitaxel in patients with previously untreated metastatic pancreatic ductal adenocarcinoma. Invest New Drugs. 2019;37(1):159–65. pmid:30105668
  54. 54. Bauer TM, Rpatel M, Forero-Torres A, George TJ, Assad A, Du Y, et al. Aphase Ib study of ruxolitinib + gemcitabine ± nab-paclitaxel in patients with advanced solid tumors. OncoTargets Ther. 2018;11:2399–407.
  55. 55. Hurwitz H, Van Cutsem E, Bendell J, Hidalgo M, Li CP, Salvo MG, et al. Ruxolitinib + capecitabine in advanced/metastatic pancreatic cancer after disease progression/intolerance to first-line therapy: JANUS 1 and 2 randomized phase III studies. Invest New Drugs. 2018;36(4):683–95. pmid:29508247
  56. 56. Hurwitz HI, Uppal N, Wagner SA, Bendell JC, Beck JT, Wade SM, et al. Randomized, double-blind, phase II study of ruxolitinib or placebo in combination with capecitabine in patients with metastatic pancreatic cancer for whom therapy with gemcitabine has failed. J Clin Oncol. 2015;33(34):4039–47. pmid:26351344
  57. 57. Eckhardt SG, De Porre P, Smith D, Maurel J, Steward WP, Bouche O, et al. Patient-Reported Outcomes as a Component of the Primary Endpoint in a Double-Blind, Placebo-Controlled Trial in Advanced Pancreatic Cancer. J Pain Symptom Manage. 2009;37(2):135–43. pmid:18723314
  58. 58. Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade JL, Giguere JK, et al. A phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: A Southwest oncology group (SWOG 9924) study. Invest New Drugs. 2005 Oct 1;23(5):485–7. pmid:16133800
  59. 59. Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and Pharmacodynamic Study of the Farnesyltransferase Inhibitor R115777 as Initial Therapy in Patients With Metastatic Pancreatic Adenocarcinoma. J Clin Oncol. 2003 Apr 1;21(7):1301–6. pmid:12663718
  60. 60. Thoennissen NH, Iwanski GB, Doan NB, Okamoto R, Lin P, Abbassi S, et al. Cucurbitacin B Induces Apoptosis by Inhibition of the JAK/STAT Pathway and Potentiates Antiproliferative Effects of Gemcitabine on Pancreatic Cancer Cells. Cancer Res. 2009 Jul 15;69(14):5876–84. pmid:19605406
  61. 61. Lu C, Talukder A, Savage NM, Singh N, Liu K. JAK-STAT-mediated chronic inflammation impairs cytotoxic T lymphocyte activation to decrease anti-PD-1 immunotherapy efficacy in pancreatic cancer. Oncoimmunology. 2017;6(3):e1291106. pmid:28405527
  62. 62. Sun C, Zhang M, Shan X, Zhou X, Yang J, Wang Y, et al. Inhibitory effect of cucurbitacin e on pancreatic cancer cells growth via STAT3 signaling. J Cancer Res Clin Oncol. 2010;136(4):603–10. pmid:19816711
  63. 63. Jonker DJ, Nott L, Yoshino T, Gill S, Shapiro J, Ohtsu A, et al. Napabucasin versus placebo in refractory advanced colorectal cancer: a randomised phase 3 trial. Lancet Gastroenterol Hepatol. 2018 Apr 1;3(4):263–70. pmid:29397354
  64. 64. Yavropoulou MP, Makras P, Anastasilakis AD. Bazedoxifene for the treatment of osteoporosis. Expert Opin Pharmacother. 2019 Jul 3;20(10):1201–10. pmid:31091133
  65. 65. You H, Xu D, Zhao J, Li J, Wang Q, Tian X, et al. JAK Inhibitors: Prospects in Connective Tissue Diseases. Clin Rev Allergy Immunol. 2020 Dec;59(3):334–51. pmid:32222877
  66. 66. Srinivas N, Barbour AM, Epstein N, Zhou G, Petusky S, Xun Z, et al. The Effect of Renal Impairment on the Pharmacokinetics and Safety of Itacitinib. J Clin Pharmacol. 2020 Aug;60(8):1022–9. pmid:32149388
  67. 67. Angelini J, Talotta R, Roncato R, Fornasier G, Barbiero G, Dal Cin L, et al. JAK-Inhibitors for the Treatment of Rheumatoid Arthritis: A Focus on the Present and an Outlook on the Future. Biomolecules. 2020 Jul 5;10(7):1002. pmid:32635659
  68. 68. von Ahrens D, Bhagat TD, Nagrath D, Maitra A, Verma A. The role of stromal cancer-associated fibroblasts in pancreatic cancer. J Hematol OncolJ Hematol Oncol. 2017 Dec;10(1):76. pmid:28351381