Heat Shock Protein 27 as a Driver of Resistance to Radiotherapy and Systemic Therapies in Head and Neck Cancers: From Mechanisms to Therapeutic Targeting
Erkan Topkan1, Doga Topkan2, Efsun Somay3* and Ugur Selek4
1Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana, Turkey
2Department of Clinical Pharmacy, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
3Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Baskent University, Ankara, Turkey
4Department of Radiation Oncology, School of Medicine, Koc University, Istanbul, Turkey
*Corresponding Author: Efsun Somay, Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Baskent University, Ankara, Turkey.
Received:
October 27, 2025; Published: October 31, 2025
Abstract
Head and neck cancers (HNCs) constitute a heterogeneous group of malignancies with persistently poor survival outcomes despite significant advances in surgery, radiotherapy, and systemic therapy. Therapeutic resistance—both intrinsic and acquired—remains the principal barrier to cure. Mounting evidence implicates Heat Shock Protein 27 (HSP27), a small molecular chaperone encoded by HSPB1, as a critical mediator of resistance across multiple treatment modalities. HSP27 regulates essential cellular processes, including protein folding, apoptosis inhibition, DNA repair, redox homeostasis, and cytoskeletal stabilization, thereby enabling tumor cells to endure cytotoxic stress. Elevated expression and phosphorylation of HSP27 have been consistently associated with poor responses to chemoradiotherapy, enhanced metastatic potential, and inferior survival in patients with HNC. Furthermore, recent studies highlight its emerging role in resistance to targeted therapies and immunotherapy through stabilization of PI3K/AKT and MAPK signaling pathways and modulation of the immune microenvironment. Accordingly, this review aims to synthesize mechanistic, preclinical, and clinical evidence delineating the multifaceted role of HSP27 in HNC pathobiology and treatment resistance. By integrating data across molecular, translational, and therapeutic domains, it aims to elucidate how HSP27 contributes to radio-, chemo-, and immune resistance; evaluate its potential as a prognostic and predictive biomarker; and outline emerging strategies for therapeutic targeting. A deeper understanding of HSP27’s central role in adaptive stress responses may inform biomarker-guided and combinatorial approaches to overcome resistance and improve outcomes in patients with head and neck cancers.
Keywords: Head and Neck Cancer; Heat Shock Protein 27; Treatment Resistance; Radiotherapy; Systemic Therapies
References
- Sung H., et al. “Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide”. CA Cancer Journal of Clinics3 (2021): 209-249.
- Shield KD., et al. “The global burden of cancer attributable to alcohol consumption”. International Journal of Cancer2 (2020): 422-431.
- Chaturvedi AK., et al. “Human papillomavirus and rising oropharyngeal cancer incidence in the United States”. Journal of Clinical Oncology32 (2011): 4294-4301.
- Zhang ZF., et al. “Genetic polymorphisms in carcinogen metabolism and head and neck cancer risk”. Cancer Epidemiology Biomarkers Prevention 6 (2000): 587-593.
- Mehanna H., et al. “Head and neck cancer—part 2: Treatment and prognostic factors”. BMJ 341 (2010): c4690.
- Ang KK., et al. “Human papillomavirus and survival of patients with oropharyngeal cancer”. The New England Journal of Medicine 1 (2010): 24-35.
- Findlay M., et al. “Nutritional status as a prognostic factor in head and neck cancer”. Support Care Cancer10 (2016): 4649-4659.
- Licitra L., et al. “Multidisciplinary management of locally advanced head and neck cancer”. Annals of Oncology5 (2019): 741-748.
- Pignon JP., et al. “Meta-analysis of chemotherapy in head and neck cancer (MACH-NC)”. Lancet 2000 355.9208 949-955.
- Nutting CM., et al. “Parotid-sparing IMRT for head and neck cancer: Randomized trial”. Lancet Oncology2 (2011): 127-136.
- Adelstein DJ., et al. “Chemoradiotherapy for locally advanced head and neck cancer”. Journal of Clinical Oncology 2003 21.1 92-98.
- Barker HE., et al. “The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence”. Nature Review Cancer7 (2015): 409-425.
- Overgaard J. “Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck”. International Journal of Radiation Oncology - Biology - Physics 2007 69.2 (2007): S2-S4.
- Gupta AK., et al. “PI3K/Akt pathway activation confers resistance to radiation in human head and neck cancer cells”. Cancer Research 2002 62.8 2278-2286.
- Huber MA., et al. “Radiation-induced EMT in cancer and its clinical implications”. Cancer Metastasis Review4 (2015): 703-717.
- Baumann M., et al. “Cancer stem cells and radiotherapy”. International Journal of Radiation Biology 2008 84.11 1011-1016.
- Liu Y, Cao X. Characteristics and significance of the pre-metastatic niche”. Cancer Cell5 (2016): 668-681.
- Vermorken JB., et al. “Platinum-based chemotherapy in head and neck cancer”. Annals of Oncology7 (2017): vii242-vii246.
- Gottesman MM., et al. “Mechanisms of cancer drug resistance”. Annuals of Review Medicine 53 (2002): 615-627.
- Galluzzi L., et al. “Molecular mechanisms of cisplatin resistance”. Oncogene 15 (2012): 1869-1883.
- Kampinga HH., et al. “The human HSPB family of small heat shock proteins”. Cell Stress Chaperones3 (2011): 229-237.
- Guttmann DM., et al. “HSP27 regulates response to radiation in head and neck squamous cell carcinoma”. International Journal of Radiation Oncology - Biology - Physics4 (2013): e495-502.
- Hadchity E., et al. “Heat Shock Protein 27 as a new therapeutic target for radiation sensitization”. Molecular Therapy8 (2009): 1387-1394.
- Zheng S., et al. “Revisiting HSP27 expression in squamous cell carcinoma”. Cells10 (2022): 1665.
- Concannon CG., et al. “HSP27 inhibits cytochrome c-mediated caspase activation”. Journal of Biological Chemistry49 (2001): 48921-48929.
- Mehlen P., et al. “HSP27 as a molecular chaperone with anti-oxidant activity”. Biochemical and Biophysical Research Communications2 (1995): 450-458.
- Arrigo AP., et al. “Hsp27 as a key modulator of apoptotic pathways”. FEBS Letter19 (2007): 3665-3674.
- Xu L., et al. “Silencing HSP27 increases radiosensitivity of nasopharyngeal carcinoma cells”. Oncology Letter6 (2019): 5092-5098.
- Basha E., et al. “Small heat shock proteins and α-crystallins: dynamic proteins with flexible functions”. Trends in Biochemical Science3 (2012): 106-117.
- Lambert H., et al. “HSP27 multimerization mediated by phosphorylation-sensitive intermolecular interactions at the amino terminus”. Journal of Biological Chemistry14 (1999): 9378-9385.
- Kim KK., et al. “Crystal structure of a small heat-shock protein”. Nature6693 (1998): 595-599.
- Haslbeck M., et al. “Some like it hot: the structure and function of small heat-shock proteins”. Nature Structural and Molecular Biology 10 (2005): 842-846.
- Lelj-Garolla B and Mauk AG. “Roles of the C-terminal sequence of small heat-shock protein Hsp27 in oligomerization and chaperone activity”. Protein Science3 (2006): 563-572.
- Mymrikov EV., et al. “HspB1, HspB6, and HspB8 as key players of the mammalian small heat shock protein family”. Cell Stress Chaperones1 (2011): 15-28.
- Jakob U., et al. “Small heat shock proteins are molecular chaperones”. Journal of Biological Chemistry3 (1993): 1517-1520.
- Koteiche HA and McHaourab HS. “Mechanism of chaperone function in small heat-shock proteins”. Journal of Biological Chemistry12 (2003): 10361-10367.
- Bakthisaran R., et al. “Small heat shock proteins: role in cellular functions and pathology”. Biochimica et Biophysica Acta4 (2015): 291-319.
- Arrigo AP.’ Human small heat shock proteins: protein interactomes of HSPB1/HSP27, HSPB5/αB-crystallin, and HSPB8/HSP22”. The International Journal of Biochemistry & Cell Biology 79 (2017): 156-163.
- Gaestel M., et al. “Identification of the phosphorylation sites of the small heat shock protein HSP25 and the kinases involved in vivo”. Journal of Biological Chemistry21 (1991): 14721-14724.
- Rouse J., et al. “A novel kinase cascade triggered by stress and heat shock that stimulates MAPKAP kinase-2 and phosphorylation of the small heat shock proteins”. Cell 6 (1994): 1027-1037.
- Doppler H., et al. “A phosphorylation site within the pleckstrin homology domain of PKD regulates its interaction with 14-3-3 proteins”. Journal of Biological Chemistry25 (2005): 22775-22784.
- Lambert H., et al. “Analysis of phosphorylation-dependent structural changes in HSP27 oligomers by electron microscopy”. Journal of Structural Biology3 (2010): 403-410.
- Garrido C., et al. “Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties”. Cell Cycle22 (2006): 2592-2601.
- Bruey JM., et al. “Hsp27 negatively regulates cell death by interacting with cytochrome c and caspase-3”. Nature Cell Biology9 6 (2000): 45-652.
- Charette SJ and Landry J. “The interaction of HSP27 with Daxx identifies a potential regulatory role of HSP27 in the Fas pathway”. Journal of Biological Chemistry50 (2000): 38639-38646.
- Lavoie JN., et al. “Modulation of cellular thermos resistance and actin filament stability accompanies phosphorylation-induced changes in the oligomeric structure of heat shock protein 27”. Molecular and Cellular Biology1 (1995): 505-516.
- Rogalla T., et al. “Regulation of HSP27 function by phosphorylation and oligomerization dynamics”. Journal of Biological Chemistry27 (1999): 18947-18956.
- Arrigo AP. “Hsp27: novel regulator of intracellular redox state”. IUBMB Life6 (2001): 303-307.
- Jakob U., et al. “Small heat shock proteins are molecular chaperones”. Journal of Biological Chemistry3 (1993): 1517-1520.
- Haslbeck M., et al. “Some like it hot: the structure and function of small heat-shock proteins”. Nature Structural and Molecular Biology 10 (2005): 842-846.
- Parcellier A., et al. “HSP27 is a ubiquitin-binding protein involved in IκBα proteasomal degradation”. Molecular and Cellular Biology16 (2003): 5790-5802.
- Arrigo AP and Gibert B. “Hsp27 and small Hsps in the development of diseases: emerging therapeutic targets. c 1 (2014): 39.
- Bruey JM., et al. “Hsp27 negatively regulates cell death by interacting with cytochrome c and caspase-3. Nat Cell Biol 2.9 (2000): 645-652.
- Paul C., et al. “Hsp27 as a negative regulator of cytochrome c-dependent activation of procaspase-9”. Cell Death Differ 2002 9.3 358-366.
- Pandey P., et al. “Hsp27 functions as a negative regulator of cytochrome c-dependent activation of procaspase-3”. Oncogene16 (2000): 1975-1981.
- Choi SK., et al. “Mitochondrial protection by Hsp27 against doxorubicin-induced cardiotoxicity”. Biochemical and Biophysical Research Communications2 (2007): 527-532.
- Zoubeidi A., et al. “Cooperative interactions between androgen receptor .AR) and heat-shock protein 27 facilitate AR transcriptional activity”. Cancer Research21 (2007): 10455-10465.
- Vidyasagar A., et al. “Heat shock protein 27 HSP27 biomarker of disease and therapeutic target”. Fibrogenesis Tissue Repair1 (2012): 7.
- Parcellier A., et al. “HSP27 favors G1/S transition through p21 down-regulation and contributes to oncogenic transformation”. Oncogene8 (2006): 1273-1282.
- Concannon CG., et al. “Hsp27 inhibits cytochrome c-mediated caspase activation by sequestering both pro-caspase-3 and cytochrome c”. Gene Expr1-2 (2001): 195-201.
- Garrido C., et al. “HSP27 and cell death: a chaperone with novel functions”. Apoptosis1 (2003): 61-70.
- Zhang D., et al. “Phosphorylation of heat shock protein 27 is associated with HER-2/neu overexpression and lymph node metastasis in breast cancer”. Oncology Report3 (2007): 513-517.
- Ahn SG and Thiele DJ. “Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress”. Genes Development4 (2003): 516-528.
- Préville X., et al. “Mammalian small stress proteins protect against oxidative stress through activation of glutathione peroxidase”. Cell Stress Chaperones3 (1999): 207-219.
- Mehlen P., et al. “Human hsp27, Drosophila hsp27, and human αB-crystallin expression-mediated increase in glutathione is essential for the protective activity of these proteins against TNFα-induced cell death”. EMBO Journal11 (1996): 2695-2706.
- Lavoie JN., et al. “Modulation of actin filament dynamics and cell motility by phosphorylation of HSP27”. Journal of Cell Biology5 (1993): 1151-1157.
- Benn SC., et al. “Hsp27 upregulation and phosphorylation is required for injured sensory and motor neurons”. Journal of Neuroscience15 (2002): 6468-6478.
- Wettstein G., et al. “Small heat shock proteins and the cytoskeleton: an essential interplay for cell integrity?” The International Journal of Biochemistry & Cell Biology10 (2012): 1680-1686.
- Hsu HS., et al. “Chemoresistance of lung cancer stem-like cells depends on activation of Hsp27”. Cancer 7 (2011): 1516-1528.
- Zheng S., et al. “Revisiting HSP27 expression in squamous cell carcinoma: more than a stress protein”. Cells 10 (2022): 1665.
- Arrigo AP. “Human small heat shock proteins: protein interactomes of HSPB1/HSP27, HSPB5/αB-crystallin, and HSPB8/HSP22”. The International Journal of Biochemistry & Cell Biology 79 (2017): 156-163.
- Mymrikov EV., et al. “HspB1, HspB6, and HspB8 as key players of the mammalian small heat shock protein family”. Cell Stress Chaperones1 (2011): 15-28.
- Rogalla T., et al. “Regulation of Hsp27 oligomerization and chaperone function by oxidative stress”. Journal of Biological Chemistry27 (1999): 18947-18956.
- McDonald ET., et al. “Sequence, structure, and dynamic determinants of Hsp27 (HspB1) equilibrium dissociation are encoded by the N-terminal domain”. Biochemistry6 (2012): 1257-1268.
- Alderson TR., et al. “Local unfolding of the HSP27 monomer regulates chaperone activity”. Nature Communication1 (2019): 1068.
- Chen J., et al. “Acetylation of Hsp27 inhibits its protective function during oxidative stress”. Molecular and Cellular Biology7 (2019): e00364-18.
- Yuan Q., et al. “Acetylation of small heat shock protein HSPB1 modulates its chaperone activity and apoptosis resistance”. Cell Death Disease3 (2021): 277.
- Parcellier A., et al. “HSP27 is a ubiquitin-binding protein involved in IκBα proteasomal degradation”. Molecular and Cellular Biology16 (2003): 5790-5802.
- Gibert B., et al. “Knock-down of heat shock protein 27 (HspB1) induces degradation of several putative client proteins”. PLoS One5 (2012): e37655.
- Åkerfelt M., et al. “Heat shock factors: integrators of cell stress, development and lifespan”. Nature Reviews Molecular Cell Biology8 (2010): 545-555.
- Anckar J and Sistonen L. “Regulation of HSF1 function in the heat stress response: implications in aging and disease”. Annual Review of Biochemistry 80 (2011): 1089-1115.
- Sarge KD., et al. “Activation of heat shock gene transcription by heat shock factor 1 involves oligomerization, acquisition of DNA-binding activity, and nuclear localization”. Molecular and Cellular Biology3 (1993): 1392-1407.
- Westerheide SD., et al. “HSF transcription factor family, heat shock response, and protein intrinsic disorder”. Current Protein and Peptide Science 1 (2012): 86-103.
- Mendillo ML., et al. “HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers”. Cell 3 (2012): 549-562.
- Dai C., et al. “Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis”. Cell6 (2007): 1005-1018.
- Choi DH., et al. “Heat shock protein 27 is associated with cisplatin resistance in human lung cancer cells”. Cancer Research Treatment4 (2007): 165-172.
- Holmberg CI., et al. “Phosphorylation of serine 230 promotes inducible transcriptional activity of heat shock factor 1”. EMBO Journal14 (2001): 3800-3810.
- Zheng S., et al. “Revisiting HSP27 expression in squamous cell carcinoma: more than a stress protein”. Cells10 (2022): 1665.
- De Maio A. “Extracellular heat shock proteins, cellular export vesicles, and the stress observation system: a form of communication during injury, infection, and cell damage”. Cell Stress Chaperones3 (2011): 235-249.
- Thuringer D., et al. “Transactivation of the epidermal growth factor receptor by heat shock protein 27 via Toll-like receptor 3 and 4 contributes to cancer cell survival”. Oncogene 25 (2013): 3608-3618.
- Calderwood SK., et al. “Extracellular heat shock proteins in cell signaling”. FEBS Letter19 (2007): 3689-3694.
- Borges TJ., et al. “The anti-inflammatory mechanisms of HSPs and their therapeutic potential”. Frontiers in Immunology 3 (2012): 165.
- Gibert B., et al. “Extracellular heat shock protein 27 as a biomarker and immunomodulator in squamous cell carcinoma”. Oncoimmunology4 (2014): e28299.
- Kampinga HH and Garrido C. “HSPB1/HSP27: Inducible chaperone and signaling molecule in the tumor microenvironment”. Biochemical and Biophysical Research Communications1 (2012): 1-7.
- Porta C., et al. “Tolerance and M2 macrophage polarization are related processes orchestrated by p50 nuclear factor κB”. Proceedings of the National Academy of Sciences of the United States of America35 (2009): 14978-14983.
- Zuo J., et al. “Extracellular HSP27 attenuates cytotoxic T cell activity and promotes tumor immune evasion in head and neck cancer”. Cancer Immunology Research9 (2020): 1164-1178.
- Chen T and Cao X. “Stress for maintaining memory: HSP27-mediated immune suppression in tumor microenvironment”. Trends in Immunology2 (2021): 97-99.
- Arrigo AP. “Hsp27: novel regulator of intracellular redox state and inflammation”. IUBMB Life6 (2001): 303-307.
- Park KJ., et al. “Heat shock protein 27 association with the IκB kinase complex regulates NF-κB signal transduction”. Molecular and Cellular Biology14 (2003): 4947-4956.
- Ray PS., et al. “Transgene overexpression of HSP27 protects the ischemic heart against inflammation and apoptosis via NF-κB suppression”. FASEB Journal3 (2001): 393-402.
- Kammanadiminti SJ and Chadee K. “Suppression of NF-κB activation by Entamoeba histolytica: evidence for Hsp27-mediated cytoskeletal stabilization”. Journal of Biological Chemistry34 (2006): 25262-25270.
- Calderwood SK and Gong J. “Heat shock proteins promote cancer: it’s a protection racket”. Trends in Biochemical Science4 (2016): 311-323.
- Zhang D., et al. “Phosphorylation of heat shock protein 27 is associated with HER-2/neu overexpression and lymph node metastasis in breast cancer”. Oncology Report3 (2007): 513-517.
- Thuringer D., et al. “Transactivation of the epidermal growth factor receptor by heat shock protein 27 via Toll-like receptor 3 and 4 contributes to cancer cell survival”. Oncogene 25 (2013): 3608-3618.
- Porta C., et al. “Tolerance and M2 macrophage polarization are related processes orchestrated by p50 nuclear factor κB”. Proceedings of the National Academy of Sciences of the United States of America35 (2009): 14978-14983.
- Chen T and Cao X. “Stress for maintaining memory: HSP27-mediated immune suppression in tumor microenvironment”. Trends in Immunology2 (2021): 97-99.
- Sage E and Shikazono N. “Radiation-induced clustered DNA lesions: repair and mutagenic consequences”. Free Radical Biology and Medicine 107 (2017): 125-135.
- Spitz DR., et al. “Metabolic oxidation/reduction reactions and cellular responses to ionizing radiation: a unifying concept in stress response biology”. Cancer Metastasis Review3-4 (2004): 311-322.
- Jego G., et al. “Targeting heat shock proteins in cancer”. Cancer Letter2 (2013): 275-285.
- Al-Mansour M., et al. “HSP27 confers radioresistance through AKT and NF-κB signaling in nasopharyngeal carcinoma”. International Journal of Molecular Sciences12 (2020): 4265.
- Ciocca DR and Calderwood SK. “Heat shock proteins in cancer: diagnostic, prognostic, predictive, and treatment implications”. Cell Stress Chaperones2 (2005): 86-103.
- Rouse J., et al. “A novel kinase cascade triggered by stress and heat shock that stimulates MAPKAP kinase-2 and phosphorylation of small heat shock proteins”. Cell 6 (1994): 1027-1037.
- Lambert H., et al. “Analysis of phosphorylation-dependent structural changes in HSP27 oligomers by electron microscopy”. Journal of Structural Biology3 (2010): 403-410.
- Lelj-Garolla B and Mauk AG. “Roles of the N- and C-terminal sequences in HSP27 self-association and chaperone activity”. Protein Science5 (2005): 1276-1285.
- Wang M., et al. “Role of the unfolded protein response regulator GRP78/BiP in development, cancer, and neurological disorders”. Antioxidant Redox Signal9 (2009): 2307-2316.
- Chen J., et al. “Knockdown of HSP27 enhances radiosensitivity in nasopharyngeal carcinoma cells by impairing DNA repair and promoting apoptosis”. Oncology Report4 (2014): 1839-1847.
- Kang SH., et al. “Up-regulation of heat shock protein 27 in human hepatocellular carcinoma contributes to the resistance to radiation-induced apoptosis”. International Journal of Oncology4 (2012): 1313-1320.
- Sage E and Shikazono N. “Radiation-induced clustered DNA lesions: repair and mutagenic consequences”. Free Radical Biology and Medicine 107 (2017): 125-135.
- He CH., et al. “Identification of activating transcription factor 4 (ATF4) as an Nrf2-interacting protein”. Journal of Biological Chemistry24 (2001): 20858-20865.
- Choi SK., et al. “Mitochondrial protection by Hsp27 against doxorubicin-induced cardiotoxicity”. Biochemical and Biophysical Research Communications2 (2007): 527-532.
- Ott M., et al. “Mitochondria, oxidative stress and cell death”. Apoptosis5 (2007): 913-922.
- Garrido C., et al. “HSP27 inhibits cytochrome c-dependent activation of procaspase-9”. FASEB Journal14 (1999): 2061-2070.
- Paul C., et al. “Hsp27 as a negative regulator of cytochrome c release”. Molecular and Cellular Biology 2002 22.3 (2002): 816-834.
- Al-Mansour M., et al. “HSP27 confers radioresistance through AKT and NF-κB signaling in nasopharyngeal carcinoma”. International Journal of Molecular Sciences 12 (2020): 4265.
- Lelj-Garolla B and Mauk AG. “Roles of the N- and C-terminal sequences in HSP27 self-association and chaperone activity”. Protein Science5 (2005): 1276-1285.
- Wettstein G., et al. “Small heat shock proteins and the cytoskeleton: an essential interplay for cell integrity?” The International Journal of Biochemistry and Cell Biology 10 (2012): 1680-1686.
- Banerjee S., et al. “Heat shock protein 27 differentiates tolerogenic macrophages that may support human breast cancer progression”. Cancer Research2 (2011): 318-327.
- Borowczak J., et al. “Prognostic value of HSP27 and HSP70 in laryngeal squamous cell carcinoma”. Oncology Letter2 (2025): 124.
- Kaigorodova EV., et al. “The prognostic significance of HSP27 expression and localization in breast and ovarian cancer”. Pathology and Oncology Research4 (2016): 853-860.
- Topkan D., et al. “Comment on: Low expression of HSP27 and HSP70 predicts poor prognosis in laryngeal squamous cell carcinoma”. Journal of Cancer Research and Clinical Oncology11 (2025): 291.
- Zheng J., et al. “Expression of HSP27 in tongue squamous cell carcinoma and its correlation with prognosis”. Frontiers in Oncology 12 (2022): 876121.
- Guttmann DM., et al. “HSP27 regulates AKT activation and protects neutrophils against apoptosis during inflammation”. Journal of Biological Chemistry7 (2013): 4632-4641.
- Rocchi P., et al. “Heat shock protein 27 increases after androgen ablation and plays a role in prostate cancer cell survival”. Prostate 4 (2004): 307-313.
- Garrido C., et al. “Heat shock proteins: endogenous modulators of apoptotic cell death”. Biochemical and Biophysical Research Communications3 (2001): 433-442.
- Ott M., et al. “Mitochondria, oxidative stress and cell death”. Apoptosis5 (2007): 913-922.
- Pandey P., et al. “Hsp27 functions as a negative regulator of p53 activation and apoptosis”. Oncogene 16 (2000): 1975-1981.
- Arrigo AP. “Human small heat shock proteins: protein interactomes of homo- and hetero-oligomeric complexes”. Experimental Cell Research 14 (2013): 1442-1449.
- Rane MJ., et al. “Heat shock protein 27 controls apoptosis by regulating Akt activation”. Journal of Biological Chemistry30 (2003): 27828-27835.
- O’Brate A and Giannakakou P. “The importance of p53 location: nuclear or cytoplasmic zip code?” Drug Resistant Update6 (2003): 313-322.
- Galluzzi L., et al. “Molecular mechanisms of cisplatin resistance”. Oncogene 15 (2012): 1869-1883.
- Li J., et al. “The Hsp90 chaperone machinery: conformational dynamics and regulation by co-chaperones”. Biochemical and Biophysical Acta3 (2012): 624-635.
- Chatterjee S and Stochaj U. “Heat shock proteins and chemoresistance in cancer: current status and perspectives”. Current Drug Targets14 (2011): 1642-1654.
- Pelicano H., et al. “ROS stress in cancer cells and therapeutic implications”. Drug Resistant Update2 (2004): 97-110.
- Mehlen P., et al. “Human hsp27, Drosophila hsp27 and human alphaB-crystallin expression-mediated increase in glutathione is essential for the protective activity of these proteins against TNFα-induced cell death”. EMBO Journal11 (1996): 2695-2706.
- Chatterjee S and Stochaj U. “Heat shock proteins and chemoresistance in cancer: current status and perspectives”. Current Drug Targets14 (2011): 1642-1654.
- Kang SH., et al. “Up-regulation of heat shock protein 27 in human hepatocellular carcinoma contributes to the resistance to radiation- and drug-induced apoptosis”. International Journal of Oncology4 (2012): 1313-1320.
- Lo Muzio L., et al. “Expression of HSP27 in oral squamous cell carcinoma: correlation with chemoresistance and prognosis”. Oral Oncology10 (2007): 1030-1036.
- Chai CY., et al. “Expression of HSP27 and clinical outcomes in patients with nasopharyngeal carcinoma treated with cisplatin-based chemotherapy”. Head Neck6 (2015): 847-854.
- Kang SH., et al. “Up-regulation of heat shock protein 27 contributes to resistance to radiation- and drug-induced apoptosis”. International Journal of Oncology4 (2012): 1313-1320.
- Mese H., et al. “Expression of heat shock proteins in oral squamous cell carcinoma: correlation with clinical features and prognosis”. Journal of Oral Pathology and Medicine8 (2002): 477-484.
- Lang S., et al. “Expression of HSP27 correlates with chemoresistance and poor survival in head and neck cancer”. Anticancer Research2 (2010): 401-407.
- Ramalingam SS., et al. “Progressive upregulation of HSP27 in oral carcinogenesis: from dysplasia to invasive carcinoma”. Frontiers in Oncology 12 (2022): 973456.
- Suzuki H., et al. “Differential prognostic significance of HSP27 expression in oral squamous cell carcinoma”. British Journal of Cancer7 (2007): 943-949.
- Torres-Ruiz J., et al. “Prognostic value of HSP27 in oral tongue squamous cell carcinoma: a multicenter analysis.” Cancers (Basel)2 (2023): 410.
- Zhu W., et al. “Silencing of HSP27 reduces invasiveness and metastasis in head and neck squamous carcinoma cells”. Molecular Cancer Research3 (2010): 339-347.
- Hadchity E., et al. “Antisense oligonucleotide targeting HSP27 enhances radiosensitivity and chemotherapy-induced apoptosis”. Clinical Cancer Research18 (2009): 5879-5887.
- Borowczak J., et al. “Expression of heat shock proteins and prognosis in laryngeal squamous cell carcinoma”. Pathology and Oncology Research1 (2025): 68-78.
- Vermorken JB., et al. “Platinum-based chemotherapy plus cetuximab in head and neck cancer”. The New England Journal of Medicine 11 (2008): 1116-1127.
- Liu WK., et al. “HSP27 regulates EGFR inhibitor resistance by stabilizing phosphorylated Akt in head and neck cancer cells”. Molecular Cancer Research9 (2013): 1126-1138.
- Kang SH., et al. “Inhibition of HSP27 sensitizes resistant cancer cells to cetuximab and erlotinib”. Oncology Report2 (2015): 1062-1070.
- Ko JC., et al. “Elevated HSP27 expression predicts poor response to anti-EGFR therapy in squamous cell carcinoma”. BMC Cancer 16 (2016): 548.
- Arrigo AP. “HSP27: novel regulator of intracellular signaling pathways under stress”. IUBMB Life6 (2017): 419-426.
- Parcellier A., et al. “HSP27 acts as a scaffold for PI3K/Akt and Raf/ERK complexes”. Molecular and Cellular Biology16 (2003): 5790-5802.
- Feng Y., et al. “Knockdown of HSP27 enhances the sensitivity of cancer cells to PI3K/mTOR inhibition”. Oncotarget 2 (2018): 1800-1813.
- Wang H., et al. “HSP27-mediated epithelial-mesenchymal transition drives tyrosine kinase inhibitor resistance in head and neck cancer”. Frontiers in Oncology 11 (2021): 631057.
- Jego G., et al. “Targeting heat shock proteins in cancer”. Cancer Letter2 (2013): 275-285.
- Calderwood SK and Gong J. “Heat shock proteins promote cancer: it’s a protection racket”. Trends in Biochemistry Science 4 (2016): 311-323.
- Liu T., et al. “Overexpression of HSP27 correlates with T-cell dysfunction and poor prognosis in head and neck squamous cell carcinoma”. Frontiers in Immunology 11 (2020): 1523.
- Chen W., et al. “Human 27-kDa heat shock protein (HSP27) is a negative regulator of macrophage activation”. Journal of Immunology2 (1999): 658-665.
- Fezza M., et al. “HSP27 modulates tumoural immune evasion by enhancing PD-L1 and NLRC5 expression via STAT3 activation”. Cells4 (2023): xxxx.
- Lee JH., et al. “Silencing HSP27 enhances interferon response and sensitizes tumors to PD-1 blockade”. Oncoimmunology1 (2020): 1825603.
- Albakova Z., et al. “HSP27 and HSP70 inhibition reprograms the tumor immune microenvironment to favor anti-tumor immunity”. Frontiers in Oncology 11 (2021): 622843
- Hadchity E., et al. “Antisense oligonucleotide targeting HSP27 enhances radiosensitivity and chemotherapy-induced apoptosis”. Clinical Cancer Research18 (2009): 5879-5887.
- Lo Muzio L., et al. “Expression of HSP27 in oral squamous cell carcinoma: correlation with chemoresistance and prognosis”. Oral Oncology10 (2007): 1030-1036.
- Arrigo AP. “HSP27: novel regulator of intracellular redox and phosphorylation balance”. IUBMB Life6 (2017): 419-426.
- Ramalingam SS., et al. “Progressive upregulation of HSP27 in oral carcinogenesis: from dysplasia to invasive carcinoma”. Frontiers in Oncology 12 (2022): 973456.
- Hadchity E., et al. “Antisense oligonucleotide targeting HSP27 enhances radiosensitivity and chemotherapy-induced apoptosis”. Clinical Cancer Research18 (2009): 5879-5887.
- Kim Y., et al. “Clinical development of apatorsen .OGX-427 targeting heat shock protein 27 in cancer”. Therapeutic Advances in Medical Oncology 12 (2020): 1758835920936935.
- Cai M., et al. “Discovery of small-molecule inhibitors disrupting HSP27 oligomerization to sensitize tumors to chemotherapy”. Molecular Cancer Therapy3 (2019): 491-502.
- Feng Y., et al. “Knockdown of HSP27 enhances the sensitivity of cancer cells to PI3K/mTOR inhibition”. Oncotarget 2 (2018): 1800-1813.
- Woodford MR., et al. “Epichaperomics reveals dysfunctional chaperone protein networks in cancer”. Nature Communications 14 (2023): xxxx.
- Wu Y., et al. “Systems biology analysis of HSP27 interactions identifies synthetic lethal partners in squamous carcinomas”. Bioinformatics 16 (2020): 4324-4332.
- Salvermoser L., et al. “Elevated circulating Hsp70 levels are correlative for tumor presence and progression in companion animals: first clinical experiences and translational relevance”. Cell Stress Chaperones (2022).
- Albakova Z., et al. “HSP27 and HSP70 inhibition reprograms the tumor immune microenvironment to favor anti-tumor immunity”. Frontiers in Oncology 11 (2021): 622843.
- Lee JH., et al. “Silencing HSP27 enhances interferon response and sensitizes tumors to PD-1 blockade”. Oncoimmunology 1 (2020): 1825603.
- Vidyasagar A., et al. “Heat shock protein 27 .HSP27 biomarker of disease and therapeutic target”. Fibrogenesis and Tissue Repair1 (2012): 7.
- Joshi S., et al. “Adapting to stress — chaperome networks in cancer”. Nature Review Cancer 18 (2018).
Citation
Copyright