Skip to content
2000
Volume 25, Issue 1
  • ISSN: 1568-0096
  • E-ISSN: 1873-5576

Abstract

Triple-negative Breast Cancer (TNBC), the most aggressive breast cancer subtype, is characterized by the non-appearance of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Clinically, TNBC is marked by its low survival rate, poor therapeutic outcomes, high aggressiveness, and lack of targeted therapies. Over the past few decades, many clinical trials have been ongoing for targeted therapies in TNBC. Although some classes, such as Poly (ADP Ribose) Polymerase (PARP) inhibitors and immunotherapies, have shown positive therapeutic outcomes, however, clinical effects are not much satisfiable. Moreover, the development of drug resistance is the major pattern observed in many targeted monotherapies. The heterogeneity of TNBC might be the cause for limited clinical benefits. Hence,, there is a need for the potential identification of new therapeutic targets to address the above limitations. In this context, some novel targets that can address the above-mentioned concerns are emerging in the era of TNBC therapy, which include Hypoxia Inducible Factor (HIF-1α), Matrix Metalloproteinase 9 (MMP-9), Tumour Necrosis Factor-α (TNF-α), β-Adrenergic Receptor (β-AR), Voltage Gated Sodium Channels (VGSCs), and Cell Cycle Regulators. Currently, we summarize the ongoing clinical trials and discuss the novel therapeutic targets in the management of TNBC.

Loading

Article metrics loading...

/content/journals/ccdt/10.2174/0115680096280750240123054936
2024-02-21
2024-11-21
Loading full text...

Full text loading...

References

  1. ArslanC. DizdarO. AltundagK. Pharmacotherapy of triple-negative breast cancer.Expert Opin. Pharmacother.200910132081209310.1517/1465656090311730919640211
    [Google Scholar]
  2. DentR. TrudeauM. PritchardK.I. HannaW.M. KahnH.K. SawkaC.A. LickleyL.A. RawlinsonE. SunP. NarodS.A. Triple-negative breast cancer: Clinical features and patterns of recurrence.Clin. Cancer Res.200713154429443410.1158/1078‑0432.CCR‑06‑304517671126
    [Google Scholar]
  3. AksoyS. DizdarO. HarputluogluH. AltundagK. Demographic, clinical, and pathological characteristics of Turkish triple-negative breast cancer patients: Single center experience.Ann. Oncol.200718111904190610.1093/annonc/mdm48717993632
    [Google Scholar]
  4. Van CalsterB. Vanden BemptI. DrijkoningenM. PochetN. ChengJ. Van HuffelS. HendrickxW. DecockJ. HuangH-J. LeunenK. AmantF. BertelootP. ParidaensR. WildiersH. Van LimbergenE. WeltensC. TimmermanD. Van GorpT. SmeetsA. Van den BogaertW. VergoteI. ChristiaensM-R. NevenP. Axillary lymph node status of operable breast cancers by combined steroid receptor and HER-2 status: triple positive tumours are more likely lymph node positive.Breast Cancer Res. Treat.2009113118118710.1007/s10549‑008‑9914‑7
    [Google Scholar]
  5. PerouC.M. SørlieT. EisenM.B. van de RijnM. JeffreyS.S. ReesC.A. PollackJ.R. RossD.T. JohnsenH. AkslenL.A. FlugeØ. PergamenschikovA. WilliamsC. ZhuS.X. LønningP.E. Børresen-DaleA.L. BrownP.O. BotsteinD. Molecular portraits of human breast tumours.Nature2000406679774775210.1038/3502109310963602
    [Google Scholar]
  6. NielsenT.O. HsuF.D. JensenK. CheangM. KaracaG. HuZ. Hernandez-BoussardT. LivasyC. CowanD. DresslerL. AkslenL.A. RagazJ. GownA.M. GilksC.B. van de RijnM. PerouC.M. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma.Clin. Cancer Res.200410165367537410.1158/1078‑0432.CCR‑04‑022015328174
    [Google Scholar]
  7. CouchF. Genetic epidemiology of BRCA1.Cancer Biol. Ther.20043650951410.4161/cbt.3.6.84015254414
    [Google Scholar]
  8. ChenQ.J. WangY-A. SituH-L. LuD-M. RenL-P. LaiX-W. LiuP-X. LinY. Effects of runing recipeⅡ on expressions of p53 and ras oncogene proteins and cell cycle of the transplanted Ca761 breast cancer in mice.J. Chin. Integr. Med.20053322522810.3736/jcim2005031715885175
    [Google Scholar]
  9. HanahanD. CoussensL.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment.Cancer Cell201221330932210.1016/j.ccr.2012.02.02222439926
    [Google Scholar]
  10. NilenduP. SarodeS.C. JahagirdarD. TandonI. PatilS. SarodeG.S. PalJ.K. SharmaN.K. Mutual concessions and compromises between stromal cells and cancer cells: Driving tumor development and drug resistance.Cell Oncol.201841435336710.1007/s13402‑018‑0388‑230027403
    [Google Scholar]
  11. BadodekarN. SharmaA. PatilV. TelangG. SharmaR. PatilS. VyasN. SomasundaramI. Angiogenesis induction in breast cancer: A paracrine paradigm.Cell Biochem. Funct.202139786087310.1002/cbf.366334505714
    [Google Scholar]
  12. CarmelietP. JainR.K. Molecular mechanisms and clinical applications of angiogenesis.Nature2011473734729830710.1038/nature1014421593862
    [Google Scholar]
  13. HanahanD. FolkmanJ. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis.Cell199686335336410.1016/S0092‑8674(00)80108‑78756718
    [Google Scholar]
  14. MessehaS.S. ZarmouhN.O. AntonieL. SolimanK.F.A. Sanguinarine inhibition of TNF-α-induced CCL2, IKBKE/NF-κB/ERK1/2 signaling pathway, and cell migration in human triple-negative breast cancer cells.Int J Mol Sci20222315832910.3390/ijms23158329
    [Google Scholar]
  15. HaoQ. VadgamaJ.V. WangP. CCL2/CCR2 signaling in cancer pathogenesis.Cell Commun. Signal.20201818210.1186/s12964‑020‑00589‑832471499
    [Google Scholar]
  16. VaupelP. HöckelM. MayerA. Detection and characterization of tumor hypoxia using pO2 histography.Antioxid. Redox Signal.2007981221123610.1089/ars.2007.162817536958
    [Google Scholar]
  17. LiedtkeC. MazouniC. HessK.R. AndréF. TordaiA. MejiaJ.A. SymmansW.F. Gonzalez-AnguloA.M. HennessyB. GreenM. CristofanilliM. HortobagyiG.N. PusztaiL. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer.J. Clin. Oncol.20082681275128110.1200/JCO.2007.14.414718250347
    [Google Scholar]
  18. CareyL.A. DeesE.C. SawyerL. GattiL. MooreD.T. CollichioF. OllilaD.W. SartorC.I. GrahamM.L. PerouC.M. The triple negative paradox: Primary tumor chemosensitivity of breast cancer subtypes.Clin. Cancer Res.20071382329233410.1158/1078‑0432.CCR‑06‑110917438091
    [Google Scholar]
  19. DianaA. FranzeseE. CentonzeS. CarlinoF. Della CorteC.M. VentrigliaJ. PetrilloA. De VitaF. AlfanoR. CiardielloF. OrdituraM. Triple-negative breast cancers: Systematic review of the literature on molecular and clinical features with a focus on treatment with innovative drugs.Curr. Oncol. Rep.201820107610.1007/s11912‑018‑0726‑630128845
    [Google Scholar]
  20. XiangL. GilkesD.M. ChaturvediP. LuoW. HuH. TakanoN. LiangH. SemenzaG.L. Ganetespib blocks HIF-1 activity and inhibits tumor growth, vascularization, stem cell maintenance, invasion, and metastasis in orthotopic mouse models of triple-negative breast cancer.J. Mol. Med.201492151164
    [Google Scholar]
  21. ZoltánS. KochA.E.Z. Mechanisms of disease: Angiogenesis in inflammatory diseases.Nat Clin Pract Rheumatol311635643
    [Google Scholar]
  22. TangJ. LiZ. LuL. ChoC.H. β-Adrenergic system, a backstage manipulator regulating tumour progression and drug target in cancer therapy.Semin. Cancer Biol.201323653354210.1016/j.semcancer.2013.08.00924012659
    [Google Scholar]
  23. DriffortV. GilletL. BonE. Marionneau-LambotS. OullierT. JoulinV. CollinC. PagèsJ.C. JourdanM.L. ChevalierS. BougnouxP. Le GuennecJ.Y. BessonP. RogerS. Ranolazine inhibits NaV1.5-mediated breast cancer cell invasiveness and lung colonization.Mol. Cancer201413126410.1186/1476‑4598‑13‑26425496128
    [Google Scholar]
  24. FraserS.P. DissJ.K.J. ChioniA.M. MycielskaM.E. PanH. YamaciR.F. PaniF. SiwyZ. KrasowskaM. GrzywnaZ. BrackenburyW.J. TheodorouD. KoyutürkM. KayaH. BattalogluE. De BellaM.T. SladeM.J. TolhurstR. PalmieriC. JiangJ. LatchmanD.S. CoombesR.C. DjamgozM.B.A. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis.Clin. Cancer Res.200511155381538910.1158/1078‑0432.CCR‑05‑032716061851
    [Google Scholar]
  25. SpeyerC.L. NassarM.A. HachemA.H. BukhshM.A. JafryW.S. KhansaR.M. GorskiD.H. Riluzole mediates anti-tumor properties in breast cancer cells independent of metabotropic glutamate receptor-1.Breast Cancer Res. Treat.2016157221722810.1007/s10549‑016‑3816‑x27146584
    [Google Scholar]
  26. ZhuS. WuY. SongB. YiM. YanY. MeiQ. WuK. Recent advances in targeted strategies for triple-negative breast cancer.J. Hematol. Oncol.202316110010.1186/s13045‑023‑01497‑337641116
    [Google Scholar]
  27. EikesdalH.P. YndestadS. ElzawahryA. Llop-GuevaraA. GiljeB. BlixE.S. EspelidH. LundgrenS. GeislerJ. VagstadG. VenizelosA. MinsaasL. LeirvaagB. GudlaugssonE.G. VintermyrO.K. AaseH.S. AasT. BalmañaJ. SerraV. JanssenE.A.M. KnappskogS. LønningP.E. Olaparib monotherapy as primary treatment in unselected triple negative breast cancer.Ann. Oncol.202132224024910.1016/j.annonc.2020.11.00933242536
    [Google Scholar]
  28. RobsonM.E. TungN. ConteP. ImS.A. SenkusE. XuB. MasudaN. DelalogeS. LiW. ArmstrongA. WuW. GoesslC. RunswickS. DomchekS.M. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer.Ann. Oncol.201930455856610.1093/annonc/mdz01230689707
    [Google Scholar]
  29. TuttA.N.J. GarberJ.E. KaufmanB. VialeG. FumagalliD. RastogiP. GelberR.D. de AzambujaE. FieldingA. BalmañaJ. DomchekS.M. GelmonK.A. HollingsworthS.J. KordeL.A. LinderholmB. BandosH. SenkusE. SugaJ.M. ShaoZ. PippasA.W. NoweckiZ. HuzarskiT. GanzP.A. LucasP.C. BakerN. LoiblS. McConnellR. PiccartM. SchmutzlerR. StegerG.G. CostantinoJ.P. ArahmaniA. WolmarkN. McFaddenE. KarantzaV. LakhaniS.R. YothersG. CampbellC. GeyerC.E. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer.N Engl J Med2021384252394240510.1056/NEJMoa2105215
    [Google Scholar]
  30. LittonJ.K. ScogginsM.E. HessK.R. AdradaB.E. MurthyR.K. DamodaranS. DeSnyderS.M. BrewsterA.M. BarcenasC.H. ValeroV. WhitmanG.J. Schwartz-GomezJ. MittendorfE.A. ThompsonA.M. HelgasonT. IbrahimN. Piwnica-WormsH. MoulderS.L. ArunB.K. Neoadjuvant talazoparib for patients with operable breast cancer with a germline BRCA pathogenic variant.J. Clin. Oncol.202038538839410.1200/JCO.19.0130431461380
    [Google Scholar]
  31. BenieyM. HubertA. HaqueT. CotteA.K. BéchirN. ZhangX. Tran-ThanhD. HassanS. Sequential targeting of PARP with carboplatin inhibits primary tumour growth and distant metastasis in triple-negative breast cancer.Br. J. Cancer2023128101964197510.1038/s41416‑023‑02226‑w36941406
    [Google Scholar]
  32. BianchiniG. De AngelisC. LicataL. GianniL. Treatment landscape of triple-negative breast cancer — expanded options, evolving needs.Nat. Rev. Clin. Oncol.20221929111310.1038/s41571‑021‑00565‑234754128
    [Google Scholar]
  33. LuoJ. JinJ. YangF. SunZ. ZhangW. ShiY. XuJ. GuanX. The correlation between PARP1 and BRCA1 in AR positive triple-negative breast cancer.Int. J. Biol. Sci.201612121500151010.7150/ijbs.1617627994514
    [Google Scholar]
  34. LehmannB.D. AbramsonV.G. SandersM.E. MayerE.L. HaddadT.C. NandaR. Van PoznakC. StornioloA.M. NangiaJ.R. Gonzalez-EricssonP.I. SanchezV. JohnsonK.N. AbramsonR.G. ChenS.C. ShyrY. ArteagaC.L. WolffA.C. PietenpolJ.A. TBCRC 032 IB/II multicenter study: Molecular insights to AR antagonist and PI3K inhibitor efficacy in patients with AR+ metastatic triple-negative breast cancer.Clin. Cancer Res.20202692111212310.1158/1078‑0432.CCR‑19‑217031822498
    [Google Scholar]
  35. PascualJ. TurnerN.C. Targeting the PI3-kinase pathway in triple-negative breast cancer.Ann. Oncol.20193071051106010.1093/annonc/mdz13331050709
    [Google Scholar]
  36. NicholsonK.M. QuinnD.M. KellettG.L. WarrJ.R. LY294002, an inhibitor of phosphatidylinositol-3-kinase, causes preferential induction of apoptosis in human multidrug resistant cells.Cancer Lett.20031901313610.1016/S0304‑3835(02)00615‑812536074
    [Google Scholar]
  37. JoshiS. SinghA.R. DurdenD.L. Pan-PI-3 kinase inhibitor SF1126 shows antitumor and antiangiogenic activity in renal cell carcinoma.Cancer Chemother. Pharmacol.201575359560810.1007/s00280‑014‑2639‑x25578041
    [Google Scholar]
  38. MahadevanD. ChioreanE.G. HarrisW.B. Von HoffD.D. Stejskal-BarnettA. QiW. AnthonyS.P. YoungerA.E. RensvoldD.M. CordovaF. SheltonC.F. BeckerM.D. GarlichJ.R. DurdenD.L. RamanathanR.K. Phase I pharmacokinetic and pharmacodynamic study of the pan-PI3K/mTORC vascular targeted pro-drug SF1126 in patients with advanced solid tumours and B-cell malignancies. Eur J Cancer201248183319332710.1016/j.ejca.2012.06.027
    [Google Scholar]
  39. OliveiraM. SauraC. NuciforoP. CalvoI. AndersenJ. Passos-CoelhoJ.L. Gil GilM. BermejoB. PattD.A. CiruelosE. de la PeñaL. XuN. WongchenkoM. ShiZ. SingelS.M. IsakoffS.J. FAIRLANE, a double-blind placebo-controlled randomized phase II trial of neoadjuvant ipatasertib plus paclitaxel for early triple-negative breast cancer.Ann. Oncol.20193081289129710.1093/annonc/mdz17731147675
    [Google Scholar]
  40. WuX. XuY. LiangQ. YangX. HuangJ. WangJ. ZhangH. ShiJ. Recent advances in dual PI3K/mTOR inhibitors for tumour treatment.Front. Pharmacol.20221387537210.3389/fphar.2022.87537235614940
    [Google Scholar]
  41. CermaK. PiacentiniF. MoscettiL. BarboliniM. CaninoF. TornincasaA. CaggiaF. CerriS. MolinaroA. DominiciM. OmariniC. Targeting PI3K/AKT/mTOR pathway in breast cancer: From biology to clinical challenges.Biomedicines202311110910.3390/biomedicines1101010936672617
    [Google Scholar]
  42. SobandeF. DušekL. MatějkováA. RozkošT. LacoJ. RyškaA. EGFR in triple negative breast carcinoma: Significance of protein expression and high gene copy number.Cesk. Patol.2015512808625970719
    [Google Scholar]
  43. SabbahD.A. HajjoR. SweidanK. Review on epidermal growth factor receptor (EGFR) structure, signaling pathways, interactions, and recent updates of EGFR inhibitors.Curr. Top. Med. Chem.2020201081583410.2174/156802662066620030312310232124699
    [Google Scholar]
  44. CorkeryB. CrownJ. ClynesM. O’DonovanN. Epidermal growth factor receptor as a potential therapeutic target in triple-negative breast cancer.Ann. Oncol.200920586286710.1093/annonc/mdn71019150933
    [Google Scholar]
  45. BaselgaJ. GómezP. GreilR. BragaS. ClimentM.A. WardleyA.M. KaufmanB. StemmerS.M. PêgoA. ChanA. GoeminneJ.C. GraasM.P. KennedyM.J. Ciruelos GilE.M. SchneeweissA. ZubelA. GroosJ. MelezínkováH. AwadaA. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer.J. Clin. Oncol.201331202586259210.1200/JCO.2012.46.240823733761
    [Google Scholar]
  46. TangA.H. HoeferR.A. GuyeM.L. BearH.D. Persistent EGFR/K-RAS/SIAH pathway activation drives chemo-resistance and early tumor relapse in triple-negative breast cancer.Cancer Drug Resist.20225369170210.20517/cdr.2022.3136176751
    [Google Scholar]
  47. HegdeP.S. WallinJ.J. MancaoC. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics.Semin. Cancer Biol.201852Pt 211712410.1016/j.semcancer.2017.12.00229229461
    [Google Scholar]
  48. RobertN.J. DiérasV. GlaspyJ. BrufskyA.M. BondarenkoI. LipatovO.N. PerezE.A. YardleyD.A. ChanS.Y.T. ZhouX. PhanS.C. O’ShaughnessyJ. RIBBON-1: Randomized, double-blind, placebo-controlled, phase III trial of chemotherapy with or without bevacizumab for first-line treatment of human epidermal growth factor receptor 2-negative, locally recurrent or metastatic breast cancer.J. Clin. Oncol.201129101252126010.1200/JCO.2010.28.098221383283
    [Google Scholar]
  49. GerberB. von MinckwitzG. EidtmannH. RezaiM. FaschingP. TeschH. EggemannH. SchraderI. KittelK. HanuschC. SolbachC. JackischC. KunzG. BlohmerJ.U. HuoberJ. HauschildM. NekljudovaV. LoiblS. UntchM. Surgical outcome after neoadjuvant chemotherapy and bevacizumab: Results from the GeparQuinto study (GBG 44).Ann. Surg. Oncol.20142182517252410.1245/s10434‑014‑3606‑924740826
    [Google Scholar]
  50. LoiblS. WeberK.E. TimmsK.M. ElkinE.P. HahnenE. FaschingP.A. LedererB. DenkertC. SchneeweissA. BraunS. SalatC.T. RezaiM. BlohmerJ.U. ZahmD.M. JackischC. GerberB. KlareP. KümmelS. SchemC. PaepkeS. SchmutzlerR. RhiemK. PennS. ReidJ. NekljudovaV. HartmanA.R. von MinckwitzG. UntchM. Survival analysis of carboplatin added to an anthracycline/taxane-based neoadjuvant chemotherapy and HRD score as predictor of response—final results from GeparSixto.Ann. Oncol.201829122341234710.1093/annonc/mdy46030335131
    [Google Scholar]
  51. BellR. BrownJ. ParmarM. ToiM. SuterT. StegerG.G. PivotX. MackeyJ. JackischC. DentR. HallP. XuN. MoralesL. ProvencherL. HeggR. VanlemmensL. KirschA. SchneeweissA. MasudaN. OverkampF. CameronD. Final efficacy and updated safety results of the randomized phase III BEATRICE trial evaluating adjuvant bevacizumab-containing therapy in triple-negative early breast cancer.Ann. Oncol.201728475476010.1093/annonc/mdw66527993816
    [Google Scholar]
  52. HowardF.M. PearsonA.T. NandaR. Clinical trials of immunotherapy in triple-negative breast cancer.Breast Cancer Res. Treat.2022195111510.1007/s10549‑022‑06665‑635834065
    [Google Scholar]
  53. KimmickG. RatainM.J. BerryD. WoolfS. NortonL. MussH.B. Subcutaneously administered recombinant human interleukin-2 and interferon alfa-2a for advanced breast cancer: A phase II study of the Cancer and Leukemia Group B (CALGB 9041).Invest. New Drugs2004221838910.1023/B:DRUG.0000006178.32718.2214707498
    [Google Scholar]
  54. LoiS. SirtaineN. PietteF. SalgadoR. VialeG. Van EenooF. RouasG. FrancisP. CrownJ.P.A. HitreE. de AzambujaE. QuinauxE. Di LeoA. MichielsS. PiccartM.J. SotiriouC. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98.J. Clin. Oncol.201331786086710.1200/JCO.2011.41.090223341518
    [Google Scholar]
  55. AdamsS. GrayR.J. DemariaS. GoldsteinL. PerezE.A. ShulmanL.N. MartinoS. WangM. JonesV.E. SaphnerT.J. WolffA.C. WoodW.C. DavidsonN.E. SledgeG.W. SparanoJ.A. BadveS.S. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199.J. Clin. Oncol.201432272959296610.1200/JCO.2013.55.049125071121
    [Google Scholar]
  56. Cancer Genome Atlas NetworkComprehensive molecular portraits of human breast tumours.Nature20124907418617010.1038/nature1141223000897
    [Google Scholar]
  57. NandaR. ChowL.Q.M. DeesE.C. BergerR. GuptaS. GevaR. PusztaiL. PathirajaK. AktanG. ChengJ.D. KarantzaV. BuisseretL. Pembrolizumab in patients with advanced triple-negative breast cancer: Phase Ib KEYNOTE-012 study.J. Clin. Oncol.201634212460246710.1200/JCO.2015.64.893127138582
    [Google Scholar]
  58. AdamsS. SchmidP. RugoH.S. WinerE.P. LoiratD. AwadaA. CesconD.W. IwataH. CamponeM. NandaR. HuiR. CuriglianoG. ToppmeyerD. O’ShaughnessyJ. LoiS. Paluch-ShimonS. TanA.R. CardD. ZhaoJ. KarantzaV. CortésJ. Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: Cohort A of the phase II KEYNOTE-086 study.Ann. Oncol.201930339740410.1093/annonc/mdy51730475950
    [Google Scholar]
  59. CortésJ. LipatovO. ImS.A. GonçalvesA. LeeK.S. SchmidP. TamuraK. TestaL. WitzelI. OhtaniS. ZambelliS. HarbeckN. AndréF. DentR. ZhouX. KarantzaV. MejiaJ.A. WinerE.P. KEYNOTE-119: Phase III study of pembrolizumab (pembro) versus single-agent chemotherapy (chemo) for metastatic triple negative breast cancer (mTNBC).Ann. Oncol.201930v859v86010.1093/annonc/mdz394.010
    [Google Scholar]
  60. SchmidP. HaideraliA. MejiaJ. GuoZ. ZhouX. Martin-NguyenA. CortésJ. WinerE. 141P Impact of pembrolizumab versus chemotherapy on health-related quality of life in patients with metastatic triple negative breast cancer.Ann. Oncol.202031S65S6610.1016/j.annonc.2020.03.242
    [Google Scholar]
  61. CortesJ. CesconD.W. RugoH.S. NoweckiZ. ImS.A. YusofM.M. GallardoC. LipatovO. BarriosC.H. HolgadoE. IwataH. MasudaN. OteroM.T. GokmenE. LoiS. GuoZ. ZhaoJ. AktanG. KarantzaV. SchmidP. LuisF. GonzaloG.A. DiegoK. RubenK. MatiasM. MirtaV. SallyB.H. StephenB. PhilipC. ShereneL. DhanushaS. AndreaG. DonatienneT. CarlosB. LeandroB. FabianoC. RuffoF.J. RobertoH. CarvalhoD.L. ToniazziF.C.L. OdebrechtR.R. OrlandoA.S.N. FelipeS. DavidC. DanielleC. CristianoF. XinniS. JoanneY. AlejandroA. CarlosG. ClaudioS. CesarS. EduardoY. AlvaroG.D. JesusS. PetraH. ZdenekK. BohuslavM. KatarinaP. JanaP. VesnaG. ErikJ. JeanetteJ. SorenL. TamasL. HerveB. IsabelleD. AnthonyG. Anne-ClaireH.B. LuisT. Jens-UweB. PeterF. DirkF. NadiaH. JensH. AnnaK.F.S. ChristianK. SibylleL. DianaL. Tjoung-WonP.S. Raquel VonS. PaulineW. LouisC. AvaK. RogerK.C.N. PeterA. TiborC. ZsuzsannaK. LaszloL. KarolyM. GaborR. JohnC. CatherineK. SeamusO.R. SaverioC. AntoniettaD.A. EnricoR. TomoyukiA. TakaakiF. KenichiI. TakashiI. YoshinoriI. TsutomuI. HirojiI. YoshimasaK. KojiM. YasuoM. HirofumiM. SeigoN. NaokiN. ShoichiroO. AkihikoO. YasuakiS. EijiS. MasatoT. YukoT. KenjiT. KoichiroT. JunichiroW. NaohitoY. YutakaY. TeruoY. AnitaB. MasturaM.Y. AngelG.V. AlejandroJ.R. JorgeM.R. FlaviaM.V. JessicaR.C. KarinB. VivianneT.H. DavidP. EwaC. EwaN.Z. ZbigniewN. BarbaraR. JoannaS. CezaryS. RafalT. BogdanZ. AlexanderA. NataliaF. OlegL. AndreyM. VladimirM. GuzelM. Jin HeeA. Seock-AhI. Keun SeokL. Kwong HwaP. Yeon HeeP. BegonaB.H. JavierC. JosefinaC.J. LuisC.M. JoseG.S. MariaG. EstherH. EstherZ.A. Chien-TingL. Mei-ChingL. Chiun-ShengH. Chao-JungT. Ling-MingT. CagatayA. GulB. IrfanC. ErhanG. SeydaG. NilM.M. MustafaO. OzgurO. SinanY. SteveC. JanineG. IainM. PeterS. NicholasT. MarkT. ChristopherT. DuncanW. HryhoriyA. OleksandrB. IgorB. OleksiiK. OlenaK. HannaK. AnnaK. IuriiL. AllaN. NatalyaO. OlgaP. AndriiR. SergiiS. YaroslavS. DmytroT. GrygoriiU. IhorV. SibelB. MadhuC. MichaelC. PatrickC. ScottC. JenniferD. KeerthiG. JeffreyH. KentH. WilliamI. RandaL. JaniceL. RaulM. SusanM. RitaN. IraO. CoralO. TimothyP. AmitP. BrianP. HopeR. IrinaR. MichaelS. RobertS. MichaelS. LauraS. BradleyS. MichaelaT. FrancesV.A. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial.Lancet2020396102651817182810.1016/S0140‑6736(20)32531‑933278935
    [Google Scholar]
  62. ShahA.N. FlaumL. HelenowskiI. Santa-MariaC.A. JainS. RademakerA. NelsonV. TsarwhasD. CristofanilliM. GradisharW. Phase II study of pembrolizumab and capecitabine for triple negative and hormone receptor-positive, HER2−negative endocrine-refractory metastatic breast cancer.J. Immunother. Cancer202081e00017310.1136/jitc‑2019‑00017332060053
    [Google Scholar]
  63. BaarJ. AbrahamJ. BuddG.T. SilvermanP. MonteroA. MooreH. FuP. VaradanV. LadaikaK. HricikL. Abstract PS12-16: Pilot study of carboplatin, nab-paclitaxel and pembrolizumab for metastatic triple-negative breast cancer.Cancer Res.202181S4PS12-1610.1158/1538‑7445.SABCS20‑PS12‑16
    [Google Scholar]
  64. WangG.L. JiangB.H. RueE.A. SemenzaG.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension.Proc. Natl. Acad. Sci.199592125510551410.1073/pnas.92.12.55107539918
    [Google Scholar]
  65. LiS. WeiQ. LiQ. ZhangB. XiaoQ. Down-regulating HIF-1α by lentivirus-mediated shRNA for therapy of triple negative breast cancer.Cancer Biol. Ther.201516686687510.1080/15384047.2015.104095825920936
    [Google Scholar]
  66. PrabhakarN.R. SemenzaG.L. Adaptive and maladaptive cardiorespiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2.Physiol. Rev.2012923967100310.1152/physrev.00030.201122811423
    [Google Scholar]
  67. MinetE. MottetD. MichelG. RolandI. RaesM. RemacleJ. MichielsC. Hypoxia-induced activation of HIF-1: Role of HIF-1α-Hsp90 interaction.FEBS Lett.1999460225125610.1016/S0014‑5793(99)01359‑910544245
    [Google Scholar]
  68. AganiF. JiangB.H. Oxygen-independent regulation of HIF-1: Novel involvement of PI3K/AKT/mTOR pathway in cancer.Curr. Cancer Drug Targets201313324525110.2174/156800961131303000323297826
    [Google Scholar]
  69. El GuerrabA. ZegrourR. NemlinC.C. VigierF. CayreA. Penault-LlorcaF. RossignolF. BignonY.J. Differential impact of EGFR-targeted therapies on hypoxia responses: Implications for treatment sensitivity in triple-negative metastatic breast cancer.PLoS One201169e2508010.1371/journal.pone.002508021966417
    [Google Scholar]
  70. SemenzaG.L. Cancer–stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis.Oncogene201332354057406310.1038/onc.2012.57823222717
    [Google Scholar]
  71. SemenzaG.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations.J. Clin. Invest.201312393664367110.1172/JCI6723023999440
    [Google Scholar]
  72. ForsytheJ.A. JiangB.H. IyerN.V. AganiF. LeungS.W. KoosR.D. SemenzaG.L. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1.Mol. Cell. Biol.19961694604461310.1128/MCB.16.9.46048756616
    [Google Scholar]
  73. CeradiniD.J. KulkarniA.R. CallaghanM.J. TepperO.M. BastidasN. KleinmanM.E. CaplaJ.M. GalianoR.D. LevineJ.P. GurtnerG.C. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1.Nat. Med.200410885886410.1038/nm107515235597
    [Google Scholar]
  74. ChaturvediP. GilkesD.M. WongC.C.L. Kshitiz LuoW. ZhangH. WeiH. TakanoN. SchitoL. LevchenkoA. SemenzaG.L. Hypoxia-inducible factor–dependent breast cancer–mesenchymal stem cell bidirectional signaling promotes metastasis.J. Clin. Invest.2012123118920510.1172/JCI6499323318994
    [Google Scholar]
  75. ChoiJ.Y. JangY.S. MinS.Y. SongJ.Y. Overexpression of MMP-9 and HIF-1α in breast cancer cells under hypoxic conditions.J. Breast Cancer2011142889510.4048/jbc.2011.14.2.8821847402
    [Google Scholar]
  76. WongC.C.L. GilkesD.M. ZhangH. ChenJ. WeiH. ChaturvediP. FraleyS.I. WongC.M. KhooU.S. NgI.O.L. WirtzD. SemenzaG.L. Hypoxia-inducible factor 1 is a master regulator of breast cancer metastatic niche formation.Proc. Natl. Acad. Sci.201110839163691637410.1073/pnas.111348310821911388
    [Google Scholar]
  77. LiuQ. GuanC. LiuC. LiH. WuJ. SunC. Targeting hypoxia-inducible factor-1alpha: A new strategy for triple-negative breast cancer therapy.Biomed. Pharmacother.202215611386110.1016/j.biopha.2022.11386136228375
    [Google Scholar]
  78. EsfandiaryA. Taherian-EsfahaniZ. Abedin-DoA. MirfakhraieR. ShirzadM. Ghafouri-FardS. MotevaseliE. Lactobacilli modulate hypoxia-inducible factor (HIF)-1 regulatory pathway in triple negative breast cancer cell line.Cell J.201618223724410.22074/cellj.2016.431927540529
    [Google Scholar]
  79. WangF. MiaomiaoC ShiY. JiangL. ZhaoJ. HaiL. SharenG. DuH. Down-regulation of hypoxia-inducible factor-1 suppresses malignant biological behavior of triple-negative breast cancer cells.Int J Clin Exp Med.n.d.201471139333940
    [Google Scholar]
  80. KimD.H. SungB. KimJ.A. KangY.J. HwangS.Y. HwangN.L. SuhH. ChoiY.H. ImE. ChungH.Y. KimN.D. HS-1793, a resveratrol analogue, downregulates the expression of hypoxia-induced HIF-1 and VEGF and inhibits tumor growth of human breast cancer cells in a nude mouse xenograft model.Int. J. Oncol.201751271572310.3892/ijo.2017.405828656256
    [Google Scholar]
  81. YurasakpongL. ApisawetakanS. PranweerapaiboonK. SobhonP. ChaithirayanonK. Holothuria scabra extract induces cell apoptosis and suppresses warburg effect by down-regulating Akt/mTOR/HIF-1 axis in MDA-MB-231 breast cancer cells.Nutr. Cancer202173101964197510.1080/01635581.2020.181482532878490
    [Google Scholar]
  82. BaileyC.M. LiuY. PengG. ZhangH. HeM. SunD. ZhengP. LiuY. WangY. Liposomal formulation of HIF-1α inhibitor echinomycin eliminates established metastases of triple-negative breast cancer.Nanomedicine20202910227810.1016/j.nano.2020.10227832738299
    [Google Scholar]
  83. WangK.L. HsiaS.M. ChanC.J. ChangF.Y. HuangC.Y. BauD.T. WangP.S. Inhibitory effects of isoliquiritigenin on the migration and invasion of human breast cancer cells.Expert Opin. Ther. Targets201317433734910.1517/14728222.2013.75686923327692
    [Google Scholar]
  84. Mir HassaniZ. NabiuniM. ParivarK. AbdiradS. KarimzadehL. Melittin inhibits the expression of key genes involved in tumor microenvironment formation by suppressing HIF-1α signaling in breast cancer cells.Med. Oncol.20213877710.1007/s12032‑021‑01526‑634076777
    [Google Scholar]
  85. JinJ. QiuS. WangP. LiangX. HuangF. WuH. ZhangB. ZhangW. TianX. XuR. ShiH. WuX. Cardamonin inhibits breast cancer growth by repressing HIF-1α-dependent metabolic reprogramming.J. Exp. Clin. Cancer Res.201938137710.1186/s13046‑019‑1351‑431455352
    [Google Scholar]
  86. WuS. ZhangL. LiH. XuJ. JiangC. SunT. Combined use of apatinib mesylate and vinorelbine versus vinorelbine alone in recurrent or metastatic triple-negative breast cancer: Study protocol for a randomized controlled clinical trial.Trials202021142010.1186/s13063‑020‑04342‑x32448335
    [Google Scholar]
  87. GuoC.H. HsiaS. ChungC.H. LinY.C. ShihM.Y. ChenP.C. PengC.L. HenningS.M. HsuG.S.W. LiZ. Nutritional supplements in combination with chemotherapy or targeted therapy reduces tumor progression in mice bearing triple-negative breast cancer.J. Nutr. Biochem.20218710850410.1016/j.jnutbio.2020.10850432956826
    [Google Scholar]
  88. NaikA. DecockJ. Lactate metabolism and immune modulation in breast cancer: A focused review on triple negative breast tumors.Front. Oncol.20201059862610.3389/fonc.2020.59862633324565
    [Google Scholar]
  89. ParksW.C. WilsonC.L. López-BoadoY.S. Matrix metalloproteinases as modulators of inflammation and innate immunity.Nat. Rev. Immunol.20044861762910.1038/nri141815286728
    [Google Scholar]
  90. StamenkovicI. Extracellular matrix remodelling: The role of matrix metalloproteinases.J. Pathol.2003200444846410.1002/path.140012845612
    [Google Scholar]
  91. ChienY-C. LiuL-C. YeH-Y. WuJ-Y. YuY-L. EZH2 promotes migration and invasion of triple-negative breast cancer cells via regulating TIMP2-MMP-2/-9 pathway.Am. J. Cancer Res.20188342243429636998
    [Google Scholar]
  92. KessenbrockK. PlaksV. WerbZ. Matrix metalloproteinases: Regulators of the tumor microenvironment.Cell20101411526710.1016/j.cell.2010.03.01520371345
    [Google Scholar]
  93. GialeliC. TheocharisA.D. KaramanosN.K. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting.FEBS J.20112781162710.1111/j.1742‑4658.2010.07919.x21087457
    [Google Scholar]
  94. RadiskyE.S. RadiskyD.C. Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer.J. Mammary Gland Biol. Neoplasia201015220121210.1007/s10911‑010‑9177‑x20440544
    [Google Scholar]
  95. MehnerC. HocklaA. MillerE. RanS. RadiskyD.C. RadiskyE.S. Tumor cell-produced matrix metalloproteinase 9 (MMP-9) drives malignant progression and metastasis of basal-like triple negative breast cancer.Oncotarget2014592736274910.18632/oncotarget.193224811362
    [Google Scholar]
  96. GuoH. LiR. ZuckerS. TooleB.P. EMMPRIN (CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface.Cancer Res.200060488889110706100
    [Google Scholar]
  97. DeryuginaE.I. QuigleyJ.P. Matrix metalloproteinases and tumor metastasis.Cancer Metastasis Rev.200625193410.1007/s10555‑006‑7886‑916680569
    [Google Scholar]
  98. ZuckerS. HymowitzM. RolloE.E. MannR. ConnerC.E. CaoJ. FodaH.D. TompkinsD.C. TooleB.P. Tumorigenic potential of extracellular matrix metalloproteinase inducer.Am. J. Pathol.200115861921192810.1016/S0002‑9440(10)64660‑311395366
    [Google Scholar]
  99. van ’t VeerL.J. DaiH. van de VijverM.J. HeY.D. HartA.A.M. MaoM. PeterseH.L. van der KooyK. MartonM.J. WitteveenA.T. SchreiberG.J. KerkhovenR.M. RobertsC. LinsleyP.S. BernardsR. FriendS.H. Gene expression profiling predicts clinical outcome of breast cancer.Nature2002415687153053610.1038/415530a11823860
    [Google Scholar]
  100. YousefE.M. TahirM.R. St-PierreY. GabouryL.A. MMP-9 expression varies according to molecular subtypes of breast cancer.BMC Cancer201414160910.1186/1471‑2407‑14‑60925151367
    [Google Scholar]
  101. VenugopalA. Uma MaheswariT.N. Expression of matrix metalloproteinase-9 in oral potentially malignant disorders: A systematic review.J. Oral Maxillofac. Pathol.201620347447910.4103/0973‑029X.19095127721614
    [Google Scholar]
  102. GongL. WuD. ZouJ. ChenJ. ChenL. ChenY. NiC. YuanH. Prognostic impact of serum and tissue MMP-9 in non-small cell lung cancer: A systematic review and meta-analysis.Oncotarget2016714184581846810.18632/oncotarget.760726918342
    [Google Scholar]
  103. VermaS. KeshK. GuptaA. SwarnakarS. An overview of matrix metalloproteinase 9 polymorphism and gastric cancer risk.Asian Pac. J. Cancer Prev.201516177393740010.7314/APJCP.2015.16.17.739326625734
    [Google Scholar]
  104. DasariS. WudayagiriR. ValluruL. Cervical cancer: Biomarkers for diagnosis and treatment.Clin. Chim. Acta201544571110.1016/j.cca.2015.03.00525773118
    [Google Scholar]
  105. YuD.C. ChenJ. DingY.T. Hypoxic and highly angiogenic non-tumor tissues surrounding hepatocellular carcinoma: The ‘niche’ of endothelial progenitor cells.Int. J. Mol. Sci.20101182901290910.3390/ijms1108290121152281
    [Google Scholar]
  106. OkamotoT. SandaT. AsamitsuK. NF-kappa B signaling and carcinogenesis.Curr. Pharm. Des.200713544746210.2174/13816120778016294417348842
    [Google Scholar]
  107. BatraJ. RobinsonJ. MehnerC. HocklaA. MillerE. RadiskyD.C. RadiskyE.S. PEGylation extends circulation half-life while preserving in vitro and in vivo activity of tissue inhibitor of metalloproteinases-1 (TIMP-1).PLoS One2012711e5002810.1371/journal.pone.005002823185522
    [Google Scholar]
  108. ZhaoS. MaW. ZhangM. TangD. ShiQ. XuS. ZhangX. LiuY. SongY. LiuL. ZhangQ. High expression of CD147 and MMP-9 is correlated with poor prognosis of triple-negative breast cancer (TNBC) patients.Med. Oncol.201330133510.1007/s12032‑012‑0335‑423263825
    [Google Scholar]
  109. RenF. TangR. ZhangX. MadushiW.M. LuoD. DangY. LiZ. WeiK. ChenG. Overexpression of MMP family members functions as prognostic biomarker for breast cancer patients: A systematic review and meta-analysis.PLoS One2015108e013554410.1371/journal.pone.013554426270045
    [Google Scholar]
  110. ReggianiF. LabancaV. MancusoP. RabascioC. TalaricoG. OrecchioniS. ManconiA. BertoliniF. Adipose progenitor cell secretion of GM-CSF and MMP9 promotes a stromal and immunological microenvironment that supports breast cancer progression.Cancer Res.201777185169518210.1158/0008‑5472.CAN‑17‑091428754674
    [Google Scholar]
  111. TanH. ZhangM. XuL. ZhangX. ZhaoY. Gypensapogenin H suppresses tumor growth and cell migration in triple-negative breast cancer by regulating PI3K/AKT/NF-κB/MMP-9 signaling pathway.Bioorg. Chem.202212610591310.1016/j.bioorg.2022.10591335671647
    [Google Scholar]
  112. GautamJ. BanskotaS. LeeH. LeeY.J. JeonY.H. KimJ.A. JeongB.S. Down-regulation of cathepsin S and matrix metalloproteinase-9 via Src, a non-receptor tyrosine kinase, suppresses triple-negative breast cancer growth and metastasis.Exp. Mol. Med.201850911410.1038/s12276‑018‑0135‑930185799
    [Google Scholar]
  113. ZhouR. XuL. YeM. LiaoM. DuH. ChenH. Formononetin inhibits migration and invasion of MDA-MB-231 and 4T1 breast cancer cells by suppressing MMP-2 and MMP-9 through PI3K/AKT signaling pathways.Horm. Metab. Res.2014461175376010.1055/s‑0034‑137697724977660
    [Google Scholar]
  114. ZhuX. WangK. ZhangK. XuF. YinY. ZhuL. ZhouF. Galectin-1 knockdown in carcinoma-associated fibroblasts inhibits migration and invasion of human MDA-MB-231 breast cancer cells by modulating MMP-9 expression.Acta Biochim. Biophys. Sin.201648546246710.1093/abbs/gmw01927025601
    [Google Scholar]
  115. KwonM.J. Matrix metalloproteinases as therapeutic targets in breast cancer.Front. Oncol.202312110869510.3389/fonc.2022.110869536741729
    [Google Scholar]
  116. JuricV. O’SullivanC. StefanuttiE. KovalenkoM. GreensteinA. Barry-HamiltonV. MikaelianI. DegenhardtJ. YueP. SmithV. Mikels-VigdalA. MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors.PLoS One20181311e020725510.1371/journal.pone.020725530500835
    [Google Scholar]
  117. LingB. WattK. BanerjeeS. NewstedD. TruesdellP. AdamsJ. SidhuS.S. CraigA.W.B. A novel immunotherapy targeting MMP-14 limits hypoxia, immune suppression and metastasis in triple-negative breast cancer models.Oncotarget2017835583725838510.18632/oncotarget.1770228938563
    [Google Scholar]
  118. MatsumotoH. KooS. DentR. TanP.H. IqbalJ. Role of inflammatory infiltrates in triple negative breast cancer: Table 1.J. Clin. Pathol.201568750651010.1136/jclinpath‑2015‑20294425750267
    [Google Scholar]
  119. CarswellE.A. OldL.J. KasselR.L. GreenS. FioreN. WilliamsonB. An endotoxin-induced serum factor that causes necrosis of tumors.Proc. Natl. Acad. Sci.19757293666367010.1073/pnas.72.9.36661103152
    [Google Scholar]
  120. BradleyJ.R. TNF‐mediated inflammatory disease.J. Pathol.2008214214916010.1002/path.228718161752
    [Google Scholar]
  121. Van HerrewegheF. FestjensN. DeclercqW. VandenabeeleP. Tumor necrosis factor-mediated cell death: to break or to burst, that’s the question.Cell. Mol. Life Sci.201067101567157910.1007/s00018‑010‑0283‑020198502
    [Google Scholar]
  122. LiH.H. ZhuH. LiuL.S. HuangY. GuoJ. LiJ. SunX.P. ChangC.X. WangZ.H. ZhaiK. Tumour necrosis factor-α gene polymorphism is associated with metastasis in patients with triple negative breast cancer.Sci. Rep.2015511024410.1038/srep1024426165253
    [Google Scholar]
  123. SpriggsDR DeutschS. KufeD.W. Genomic structure, induction, and production of TNF-alpha.Immunol Ser199256334
    [Google Scholar]
  124. YuM. ZhouX. NiuL. LinG. HuangJ. ZhouW. GanH. WangJ. JiangX. YinB. LiZ. Targeting transmembrane TNF-α suppresses breast cancer growth.Cancer Res.201373134061407410.1158/0008‑5472.CAN‑12‑394623794706
    [Google Scholar]
  125. SzlosarekP.W. BalkwillF.R. Tumour necrosis factor α: A potential target for the therapy of solid tumours.Lancet Oncol.20034956557310.1016/S1470‑2045(03)01196‑312965278
    [Google Scholar]
  126. PileczkiV. BraicuC. GhermanC. Berindan-NeagoeI. TNF-α gene knockout in triple negative breast cancer cell line induces apoptosis.Int. J. Mol. Sci.201214141142010.3390/ijms1401041123263670
    [Google Scholar]
  127. Martínez-RezaI. DíazL. BarreraD. Segovia-MendozaM. Pedraza-SánchezS. Soca-ChafreG. LarreaF. García-BecerraR. Calcitriol inhibits the proliferation of triple-negative breast cancer cells through a mechanism involving the proinflammatory cytokines IL-1 β and TNF- α.J. Immunol. Res.2019201911110.1155/2019/638427831093512
    [Google Scholar]
  128. ZhengM. WuZ. WuA. HuangZ. HeN. XieX. MiR-145 promotes TNF-α-induced apoptosis by facilitating the formation of RIP1-FADDcaspase-8 complex in triple-negative breast cancer.Tumour Biol.20163778599860710.1007/s13277‑015‑4631‑426733177
    [Google Scholar]
  129. GreishK. TaurinS. MorsyM.A. The effect of adjuvant therapy with TNF-α on animal model of triple-negative breast cancer.Ther. Deliv.20189533334210.4155/tde‑2017‑010129681232
    [Google Scholar]
  130. MendoncaP. AlghamdiS. MessehaS. SolimanK.F.A. Pentagalloyl glucose inhibits TNF-α-activated CXCL1/GRO-α expression and induces apoptosis-related genes in triple-negative breast cancer cells.Sci. Rep.2021111564910.1038/s41598‑021‑85090‑z33707603
    [Google Scholar]
  131. LiuJ. WangP. HuangB. ChengQ. DuanY. ChenL. MaT. ZhuC. LiD. FanW. YuM. Effective suppression of triple negative breast cancer by paclitaxel nanoparticles conjugated with transmembrane TNF-α monoclonal antibody.Int. J. Pharm.202262412196910.1016/j.ijpharm.2022.12196935803533
    [Google Scholar]
  132. Martínez-RezaI. DíazL. García-BecerraR. Preclinical and clinical aspects of TNF-α and its receptors TNFR1 and TNFR2 in breast cancer.J. Biomed. Sci.20172419010.1186/s12929‑017‑0398‑929202842
    [Google Scholar]
  133. WuX. WuM.Y. JiangM. ZhiQ. BianX. XuM.D. GongF.R. HouJ. TaoM. ShouL.M. DuanW. ChenK. ShenM. LiW. TNF-α sensitizes chemotherapy and radiotherapy against breast cancer cells.Cancer Cell Int.20171711310.1186/s12935‑017‑0382‑128127258
    [Google Scholar]
  134. LibuttiS.K. PaciottiG.F. ByrnesA.A. AlexanderH.R.Jr GannonW.E. WalkerM. SeidelG.D. YuldashevaN. TamarkinL. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine.Clin. Cancer Res.201016246139614910.1158/1078‑0432.CCR‑10‑097820876255
    [Google Scholar]
  135. TesarováP. KvasnickaJ. UmlaufováA. HomolkováH. JirsaM. TesarV. SolubleT.N.F. Soluble TNF and IL-2 receptors in patients with breast cancer.Med. Sci. Monit.20006466166711208388
    [Google Scholar]
  136. KeslerS. JanelsinsM. KoovakkattuD. PaleshO. MustianK. MorrowG. DhabharF.S. Reduced hippocampal volume and verbal memory performance associated with interleukin-6 and tumor necrosis factor-alpha levels in chemotherapy-treated breast cancer survivors.Brain Behav. Immun.201330S109S11610.1016/j.bbi.2012.05.01722698992
    [Google Scholar]
  137. HayslipJ. DresslerE.V. WeissH. TaylorT.J. ChambersM. NoelT. MiriyalaS. KeeneyJ.T.R. RenX. SultanaR. VoreM. ButterfieldD.A. St ClairD. MoscowJ.A. Plasma TNF-α and soluble TNF receptor levels after doxorubicin with or without Co-administration of mesna—a randomized, cross-over clinical study.PLoS One2015104e012498810.1371/journal.pone.012498825909710
    [Google Scholar]
  138. LebrecH. PonceR. PrestonB.D. IlesJ. BornT.L. HooperM. Tumor necrosis factor, tumor necrosis factor inhibition, and cancer risk.Curr. Med. Res. Opin.201531355757410.1185/03007995.2015.101177825651481
    [Google Scholar]
  139. SloanE.K. PricemanS.J. CoxB.F. YuS. PimentelM.A. TangkanangnukulV. ArevaloJ.M.G. MorizonoK. KaranikolasB.D.W. WuL. SoodA.K. ColeS.W. The sympathetic nervous system induces a metastatic switch in primary breast cancer.Cancer Res.2010707042705210.1158/0008‑5472.CAN‑10‑0522
    [Google Scholar]
  140. PerronL. BairatiI. HarelF. MeyerF. Antihypertensive drug use and the risk of prostate cancer (Canada).Cancer Causes Control200415653554110.1023/B:CACO.0000036152.58271.5e15280632
    [Google Scholar]
  141. AlgaziM. Plu-BureauG. FlahaultA. DondonM.G. LêM.G. Could β-blocker treatments be associated with a reduction in cancer risk?Rev. Epidemiol. Sante Publique2004521536510.1016/S0398‑7620(04)99022‑015107693
    [Google Scholar]
  142. PoweD.G. VossM.J. ZänkerK.S. HabashyH.O. GreenA.R. EllisI.O. EntschladenF. Beta-blocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival.Oncotarget20101762863810.18632/oncotarget.19721317458
    [Google Scholar]
  143. BarronT.I. ConnollyR.M. SharpL. BennettK. VisvanathanK. Beta blockers and breast cancer mortality: A population- based study.J. Clin. Oncol.201129192635264410.1200/JCO.2010.33.542221632503
    [Google Scholar]
  144. Melhem-BertrandtA. Chavez-MacGregorM. LeiX. BrownE.N. LeeR.T. Meric-BernstamF. SoodA.K. ConzenS.D. HortobagyiG.N. Gonzalez-AnguloA.M. Beta-blocker use is associated with improved relapse-free survival in patients with triple-negative breast cancer.J. Clin. Oncol.201129192645265210.1200/JCO.2010.33.444121632501
    [Google Scholar]
  145. ChoyC. RaytisJ.L. SmithD.D. DuenasM. NemanJ. JandialR. LewM.W. Inhibition of β2-adrenergic receptor reduces triple-negative breast cancer brain metastases: The potential benefit of perioperative β-blockade.Oncol. Rep.20163563135314210.3892/or.2016.471027035124
    [Google Scholar]
  146. PoweD.G. EntschladenF. Using β-blockers to inhibit breast cancer progression.Nat. Rev. Clin. Oncol.20118951151210.1038/nrclinonc.2011.12321808268
    [Google Scholar]
  147. GanzP.A. ColeS.W. Expanding our therapeutic options: Beta blockers for breast cancer?J. Clin. Oncol.201129192612261610.1200/JCO.2011.35.882021632500
    [Google Scholar]
  148. RogerS.Ã. GilletL. Le GuennecJ.Y. BessonP. Voltage-gated sodium channels and cancer: Is excitability their primary role?Front. Pharmacol.2015615210.3389/fphar.2015.0015226283962
    [Google Scholar]
  149. LuoQ. WuT. WuW. ChenG. LuoX. JiangL. TaoH. RongM. KangS. DengM. The functional role of voltage-gated sodium channel Nav1.5 in metastatic breast cancer.Front. Pharmacol.202011111110.3389/fphar.2020.0111132792949
    [Google Scholar]
  150. BrackenburyW.J. ChioniA.M. DissJ.K.J. DjamgozM.B.A. The neonatal splice variant of Nav1.5 potentiates in vitro invasive behaviour of MDA-MB-231 human breast cancer cells.Breast Cancer Res. Treat.2007101214916010.1007/s10549‑006‑9281‑116838113
    [Google Scholar]
  151. YangM. KozminskiD.J. WoldL.A. ModakR. CalhounJ.D. IsomL.L. BrackenburyW.J. Therapeutic potential for phenytoin: Targeting Nav1.5 sodium channels to reduce migration and invasion in metastatic breast cancer.Breast Cancer Res. Treat.2012134260361510.1007/s10549‑012‑2102‑922678159
    [Google Scholar]
  152. FairhurstC. WattI. MartinF. BlandM. BrackenburyW.J. Sodium channel-inhibiting drugs and survival of breast, colon and prostate cancer: A population-based study.Sci. Rep.2015511675810.1038/srep1675826577038
    [Google Scholar]
  153. NeliganA. BellG.S. JohnsonA.L. GoodridgeD.M. ShorvonS.D. SanderJ.W. The long-term risk of premature mortality in people with epilepsy.Brain2011134238839510.1093/brain/awq37821278406
    [Google Scholar]
  154. OnkalR. DjamgozM.B.A. Molecular pharmacology of voltage-gated sodium channel expression in metastatic disease: Clinical potential of neonatal Nav1.5 in breast cancer.Eur. J. Pharmacol.20096251-320621910.1016/j.ejphar.2009.08.04019835862
    [Google Scholar]
  155. DjamgozM.B.A. OnkalR. Persistent current blockers of voltage-gated sodium channels: A clinical opportunity for controlling metastatic disease.PRA201381668410.2174/157489281130801006623116083
    [Google Scholar]
  156. ZhaC. BrownG.B. BrouilletteW.J. A highly predictive 3D-QSAR model for binding to the voltage-gated sodium channel: Design of potent new ligands.Bioorg. Med. Chem.20142219510410.1016/j.bmc.2013.11.04924332655
    [Google Scholar]
  157. DuttaS. Lopez CharcasO. TannerS. GradekF. DriffortV. RogerS. SelanderK. VeluS.E. BrouilletteW. Discovery and evaluation of nNav1.5 sodium channel blockers with potent cell invasion inhibitory activity in breast cancer cells.Bioorg. Med. Chem.20182692428243610.1016/j.bmc.2018.04.00329673714
    [Google Scholar]
  158. CazalesM. SchmittE. MontembaultE. DozierC. PrigentC. DucommunB. CDC25B phosphorylation by Aurora-A occurs at the G2/M transition and is inhibited by DNA damage.Cell Cycle2005491233123810.4161/cc.4.9.196416082213
    [Google Scholar]
  159. KrystyniakA. Garcia-EcheverriaC. PrigentC. FerrariS. Inhibition of Aurora A in response to DNA damage.Oncogene200625333834810.1038/sj.onc.120905616158051
    [Google Scholar]
  160. CimprichK.A. CortezD. ATR: an essential regulator of genome integrity.Nat. Rev. Mol. Cell Biol.20089861662710.1038/nrm245018594563
    [Google Scholar]
  161. FooteK.M. LauA. M NissinkJ.W. DruggingA.T.R. Drugging ATR: Progress in the development of specific inhibitors for the treatment of cancer.Future Med. Chem.20157787389110.4155/fmc.15.3326061106
    [Google Scholar]
  162. BarniehF.M. LoadmanP.M. FalconerR.A. Progress towards a clinically-successful ATR inhibitor for cancer therapy.CRPHAR2021210001710.1016/j.crphar.2021.10001734909652
    [Google Scholar]
  163. WilsonZ. OdedraR. WallezY. WijnhovenP.W.G. HughesA.M. GerrardJ. JonesG.N. Bargh-DawsonH. BrownE. YoungL.A. O’ConnorM.J. LauA. InhibitorA.T.R. ATR inhibitor AZD6738 (Ceralasertib) exerts antitumor activity as a monotherapy and in combination with chemotherapy and the PARP inhibitor olaparib.Cancer Res.20228261140115210.1158/0008‑5472.CAN‑21‑299735078817
    [Google Scholar]
  164. TuX. KahilaM.M. ZhouQ. YuJ. KalariK.R. WangL. HarmsenW.S. YuanJ. BougheyJ.C. GoetzM.P. SarkariaJ.N. LouZ. MutterR.W. ATR inhibition is a promising radiosensitizing strategy for triple negative breast cancer.Mol. Cancer Ther.201817112462247210.1158/1535‑7163.MCT‑18‑047030166399
    [Google Scholar]
  165. Al-SubhiN. AliR. Abdel-FatahT. MoseleyP.M. ChanS.Y.T. GreenA.R. EllisI.O. RakhaE.A. MadhusudanS. Targeting ataxia telangiectasia-mutated- and Rad3-related kinase (ATR) in PTEN-deficient breast cancers for personalized therapy.Breast Cancer Res. Treat.2018169227728610.1007/s10549‑018‑4683‑429396668
    [Google Scholar]
  166. de NonnevilleA. FinettiP. BirnbaumD. MamessierE. BertucciF. WEE1 dependency and pejorative prognostic value in triple‐negative breast cancer.Adv. Sci.2021817210103010.1002/advs.20210103034227743
    [Google Scholar]
  167. JinJ. FangH. YangF. JiW. GuanN. SunZ. ShiY. ZhouG. GuanX. Combined inhibition of ATR and WEE1 as a novel therapeutic strategy in triple-negative breast cancer.Neoplasia201820547848810.1016/j.neo.2018.03.00329605721
    [Google Scholar]
  168. ChenX. LowK.H. AlexanderA. JiangY. KarakasC. HessK.R. CareyJ.P.W. BuiT.N. VijayaraghavanS. EvansK.W. YiM. EllisD.C. CheungK.L. EllisI.O. FuS. Meric-BernstamF. HuntK.K. KeyomarsiK. CyclinE. Cyclin E overexpression sensitizes triple-negative breast cancer to wee1 kinase inhibition.Clin. Cancer Res.201824246594661010.1158/1078‑0432.CCR‑18‑144630181387
    [Google Scholar]
  169. YangG.J. ZhongH.J. KoC.N. WongS.Y. VellaisamyK. YeM. MaD.L. LeungC.H. Identification of a rhodium( iii ) complex as a Wee1 inhibitor against TP53 -mutated triple-negative breast cancer cells.Chem. Commun.201854202463246610.1039/C7CC09384E29367998
    [Google Scholar]
  170. HwangS.Y. ParkS. KwonY. Recent therapeutic trends and promising targets in triple negative breast cancer.Pharmacol. Ther.2019199305710.1016/j.pharmthera.2019.02.00630825473
    [Google Scholar]
  171. ChenP. LuoC. DengY. RyanK. RegisterJ. MargosiakS. Tempczyk-RussellA. NguyenB. MyersP. LundgrenK. KanC.C. O’ConnorP.M. The 1.7 A crystal structure of human cell cycle checkpoint kinase Chk1: implications for Chk1 regulation.Cell2000100668169210.1016/S0092‑8674(00)80704‑710761933
    [Google Scholar]
  172. LiuQ. GuntukuS. CuiX.S. MatsuokaS. CortezD. TamaiK. LuoG. Carattini-RiveraS. DeMayoF. BradleyA. DonehowerL.A. ElledgeS.J. Chk1 is an essential kinase that is regulated by Atr and required for the G 2 /M DNA damage checkpoint.Genes Dev.200014121448145910.1101/gad.14.12.144810859164
    [Google Scholar]
  173. BryantC. RawlinsonR. MasseyA.J. Chk1 Inhibition as a novel therapeutic strategy for treating triple-negative breast and ovarian cancers.BMC Cancer201414157010.1186/1471‑2407‑14‑57025104095
    [Google Scholar]
  174. AlbigesL. GoubarA. ScottV. VicierC. LefèbvreC. AlsafadiS. CommoF. SaghatchianM. LazarV. DessenP. DelalogeS. AndréF. QuidvilleV. Chk1 as a new therapeutic target in triple-negative breast cancer.Breast201423325025810.1016/j.breast.2014.02.00424636978
    [Google Scholar]
  175. RundleS. BradburyA. DrewY. CurtinN. Targeting the ATR-CHK1 axis in cancer therapy.Cancers20179124110.3390/cancers905004128448462
    [Google Scholar]
  176. BennettC.N. TomlinsonC.C. MichalowskiA.M. ChuI.M. LugerD. MitterederL.R. AprelikovaO. ShouJ. Piwinica-WormsH. CaplenN.J. HollingsheadM.G. GreenJ.E. Cross-species genomic and functional analyses identify a combination therapy using a CHK1 inhibitor and a ribonucleotide reductase inhibitor to treat triple-negative breast cancer.Breast Cancer Res.2012144R10910.1186/bcr323022812567
    [Google Scholar]
  177. InfanteJ.R. HollebecqueA. Postel-VinayS. BauerT.M. BlackwoodE.M. EvangelistaM. MahrusS. PealeF.V. LuX. SahasranamanS. ZhuR. ChenY. DingX. MurrayE.R. SchutzmanJ.L. LauchleJ.O. SoriaJ.C. LoRussoP.M. PhaseI. Phase I study of GDC-0425, a checkpoint kinase 1 inhibitor, in combination with gemcitabine in patients with refractory solid tumors.Clin. Cancer Res.201723102423243210.1158/1078‑0432.CCR‑16‑178227815358
    [Google Scholar]
  178. TentlerJ.J. IonkinaA.A. TanA.C. NewtonT.P. PittsT.M. GlogowskaM.J. KabosP. SartoriusC.A. SullivanK.D. EspinosaJ.M. EckhardtS.G. DiamondJ.R. p53 family members regulate phenotypic response to aurora kinase A inhibition in triple-negative breast cancer.Mol. Cancer Ther.20151451117112910.1158/1535‑7163.MCT‑14‑0538‑T25758253
    [Google Scholar]
  179. KaiK. KondoK. WangX. XieX. PitnerM.K. ReyesM.E. Torres-AdornoA.M. MasudaH. HortobagyiG.N. BartholomeuszC. SayaH. TripathyD. SenS. UenoN.T. Antitumor activity of KW-2450 against triple-negative breast cancer by inhibiting aurora A and B kinases.Mol. Cancer Ther.201514122687269910.1158/1535‑7163.MCT‑15‑009626443806
    [Google Scholar]
  180. RomanelliA. ClarkA. AssayagF. Chateau-JoubertS. PouponM.F. ServelyJ.L. FontaineJ.J. LiuX. SpoonerE. GoodstalS. de CremouxP. BiècheI. DecaudinD. MarangoniE. Inhibiting aurora kinases reduces tumor growth and suppresses tumor recurrence after chemotherapy in patient-derived triple-negative breast cancer xenografts.Mol. Cancer Ther.201211122693270310.1158/1535‑7163.MCT‑12‑0441‑T23012245
    [Google Scholar]
  181. BushT.L. PaytonM. HellerS. ChungG. HanestadK. RottmanJ.B. LobergR. FribergG. KendallR.L. SaffranD. RadinskyR. AMG 900, a small-molecule inhibitor of aurora kinases, potentiates the activity of microtubule-targeting agents in human metastatic breast cancer models.Mol. Cancer Ther.201312112356236610.1158/1535‑7163.MCT‑12‑117823990115
    [Google Scholar]
/content/journals/ccdt/10.2174/0115680096280750240123054936
Loading
/content/journals/ccdt/10.2174/0115680096280750240123054936
Loading

Data & Media loading...

This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test