Next Article in Journal
From Cells to Organoids: Approaches, Regulatory Mechanisms, Applications, and Challenges of Organoids
Previous Article in Journal
MicroRNA-221: A Context-Dependent Mediator in Human Diseases—Highlights from Molecular Mechanisms to Clinical Translation
Previous Article in Special Issue
Generation and Characterization of Cisplatin-Resistant Oral Squamous Cell Carcinoma Cells Displaying an Epithelial–Mesenchymal Transition Signature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Heat Shock Proteins in Head and Neck Squamous Cell Carcinoma

1
Department of Maxillofacial Surgery, The Ludwik Rydygier Specialist Hospital, Osiedle Zlotej Jesieni 1, 31-826 Krakow, Poland
2
Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
*
Authors to whom correspondence should be addressed.
Cells 2025, 14(23), 1897; https://doi.org/10.3390/cells14231897
Submission received: 18 October 2025 / Revised: 22 November 2025 / Accepted: 25 November 2025 / Published: 28 November 2025

Abstract

Heat shock proteins (HSPs) are produced in response to stressful conditions, such as temperature, inflammation, infection, or exposure to environmental factors. HSPs are overexpressed in some malignancies, where they modulate the tumor microenvironment and influence cancer cell behavior and survival. Clinical trials for breast, prostate, colon, and lung cancers exist, but not for head and neck squamous cell carcinomas (HNSCCs). Nonetheless, clinical studies on HSPs in HNSCC are still lacking. We review the role of HSPs with regard to physiology and as potential targets for molecular therapy in HNSCC.

1. Introduction

Heat shock proteins (HSPs) are a family of molecular chaperones that play a role in protein synthesis, secretion, folding, stabilization, translocation, and proteolysis [1,2]. Although the precise role of HSPs in tumor biology is unclear, HSPs appear to be involved in carcinogenesis and drug resistance [2,3,4].
This paper is a summary of the current knowledge on HSPs (especially HSP27, HSP70, and HSP90) in head and neck squamous cell carcinoma (HNSCC). We also consider the potential role of HSPs in their pathogenesis and the possible use of HSP inhibitors in therapy.

2. The HSP Family: Characteristics and Classification of HSPs

HSPs play a regulatory role as chaperones in protein-folding homeostasis and are involved in proliferation, differentiation, apoptosis, and responses to stress [3,5,6,7,8]. HSPs are activated by stressors such as high temperature, hypoxia, lack of nutrients, ischemia, infection, and inflammation [2,4]. They prevent protein aggregation by binding to misfolded proteins and helping them refold when the stress subsides [2]. HSPs prevent the accumulation of damaged proteins by regulating the ubiquitin–proteasome system, which is involved in the degradation of aggregated or improperly folded proteins [4]. HSPs constitute 5–10% of all proteins in normal cells and increase in response to stress [4,9]. They range in size from 10 kDa to >100 kDa and are divided into small HSPs [sHSPs—HSP20 and HSP27], HSP40, HSP60, HSP70, and HSP90, and large HSPs [HSP105 and HSP110] [4,10,11].
Ten sHSPs [HSPB1 to HSPB10] are first responders to cellular stress and do not use ATP [3,4,6,12]. Their α-crystallin domain (ACD) sequence is located near the C-terminus [11]. HSPB1, also called HSP27, is its most prominent member [6].
The HSP40 family is divided into three subclasses: Type I (DNAJA), Type II (DNAJB), and Type III (DNAJC) [8]. They have a highly conserved J-domain composed of four α-helices. They may interact with HSP70 by stimulating ATPase activity [6,8]. HSP40 assists in processes concerned with folding, unfolding, translocation, and protein degradation [6].
HSP60 (also known as chaperonin or Cpn60) is highly conserved [4]. Structurally, it comprises a double ring shape with three domains: equatorial, intermediate, and apical [13]. Its activity is controlled by the binding of ATP to the equatorial domain [8]. It is responsible for the transport and folding of mitochondrial proteins [14] and regulating apoptosis in response to stressors [13].
HSP70, a large family of ATP-dependent, highly conserved proteins, has two main domains: the nucleotide-binding domain (NBD) and the substrate-binding domain (SBD) [4,15]. NBD binds to and hydrolyzes ATP, thereby regulating its activity [8]. The SBD interacts with client proteins, assisting in folding and preventing aggregation [8]. HSP70 activity is controlled by the J-domain protein (JDP) and nucleotide exchange factor (NEF) [16].
HSP90 is the most-studied HSP, which regulates many signaling pathways involved in intracellular protein stability [17]. It is highly conserved, ATP-dependent, and expressed by six genes in humans [4,8]. It has three domains: C-terminal (CTD), middle (MD), and N-terminal nucleotide-binding (NTD) [4,8]. Its activity is based on ATP-dependent conformation changes that may be regulated by binding co-chaperones such as HSP40, HSP70, p23, Csc37, Aha1, Hip, and Hop [8].
The large HSPs (including HSP105, HSP110, and Grp170) are a subfamily that prevents substrate aggregation [4]. As they are homologous to the HSP70 family, they are often described as the “HSP70 superfamily” [4]. HSP110 serves as a co-chaperone for HSP70 to improve its folding capability [8].

3. The Role of HSPs in Cancer

Dysregulated HSP expression is associated with several diseases, including cancer [1,18,19]. It is generally accepted that HSPs play a key role in tumor initiation and progression (Figure 1). Dysregulation of the chaperonage machinery inhibits the targeting of oncoproteins and signaling pathways participating in tumor development and progression [6,18,19,20,21]. Overexpression of HSPs induces tumorigenesis in HNSCC (or cell lines) involved in carcinogenesis, epithelial–mesenchymal transition (EMT), metastasis, and resistance to irradiation and chemotherapy [3,5,20,22,23]. This review focuses on HSP27, HSP70, and HPS90 as there is less information available for other HSPs. These three HSPs are overexpressed or upregulated in tumors, but their specific roles and correlations with other proteins are not well defined [24].

3.1. HSP27

HSP27 plays a key role in cell proliferation, invasion, metastasis, and apoptosis. It is expressed in normal cells at basal levels. Conformational changes are induced by phosphorylation at one of the specific sites (Serine 15, Serine 78, Serine 82, or Threonine 143) through MAPKAP kinase 2/3 [6]. The phosphorylation status is closely related to cancer disease [1,25]. HSP27 levels increase, and there is a different function than in normal cells [26]. HSP27 interacts with different oncoproteins, receptors, and signaling pathway components (Figure 1). Its overexpression reduces cell death by apoptosis in response to various stressful factors such as hypoxia and cytotoxic drugs.
Phosphorylated HSP27 participates in extrinsic and intrinsic pathways of apoptosis, suppresses caspase-3 activity, and inactivates Bax and Daxx proteins [4,27]. HSP27 blocks caspase activity by blocking the release of SMAC Diablo and cytochrome c from mitochondria, resulting in a lack of interaction between cytochrome c and apoptotic protease-activating factor-1 (APAF-1) and procaspase-9 [1,4,6]. Phosphorylation of AKT by HSP27 prevents apoptosome formation and induces cell survival through Bax inhibition [6]. HSP27 also suppresses other apoptosis pathways such as FAS, TNF, and TRAIL [1,4,27]. HSP27 may regulate cellular senescence by HDM2 destabilization and p53 stabilization [6]. It may also inhibit several p53-related functions [6,18], such as p21 protein-inhibiting senescence in response to p53 activator nutlin-3.
HSP27 is involved in EMT, which allows epithelial cells to leave their polarized organization and intercellular adhesion, and to increase tumor invasiveness and migratory properties [4,28,29]. HSP27 triggers the phosphorylation of STAT3, which then binds to the Twist promoter and enhances EMT [30]. HSP27 overexpression might result in epidermal growth factor (EGF) signaling pathway/β-catenin activity: β-catenin binding to the SNAIL 2 (Slug) promoter, increasing metallopeptidases (MMPs) [4,26,28]. HSP27 induces the suppression of E-cadherin with concurrent activation of other EMT markers, such as vimentin, Slug, and fibronectin [4,28]. HSP27 influences EMT by c-Myc and p53 protein loss [31,32].
HSP27 may suppress apoptosis in oral cancer and facilitate tumor immune rejection by modulating the β-catenin/MMP-3 signaling cascade and by the upregulation of NF-ĸB signaling pathways [27,31,33]. HSP27 involvement in oral squamous cell carcinoma (OSCC) is unclear: it is overexpressed in dysplasia and OSCC [27], and in HNSCC lymph nodes, but not at primary sites [32]; and tumor grade correlates with HSP27 [31,32,34]. A HSP27 role in HNSCC progression has been reported by some authors who indicated that HSP27 overexpression could be attributed to the overexpression of HER receptors, c-Myc, and the loss of the p53 protein [31,32].
HSP27 plays a role in the angiogenesis of many solid tumors, including HNSCC [28]. It facilitates angiogenesis by triggering calcium entry via TLR3 and activating NF-κB in human endothelial cells, leading to the release of the vascular endothelial growth factor (VEGF), a primary controller of tumor neoangiogenesis [1,24,35]. This also results in the enhancement of vascular endothelial growth factor receptor type 2 (VEGFR2) and the activation of interleukin-8 (IL-8), both of which are proangiogenic factors in the endothelium of blood vessels [1,24,35]. In breast cancer cells, HSP27 upregulates VEGF gene transcription and stimulates VEGFR2, promoting angiogenesis and cell migration [6].

3.2. HSP70

The HSP70 family of 13 proteins play cytoprotective and anti-apoptotic roles in cell differentiation, gene expression, immune system control, cellular senescence, and programmed cell death [1,22], and protects cells from a variety of stressors such as ischemia, infection, inflammation, heavy metals, and hyperthermia [22]. HSP70 overexpression might cause cell transformation by suppressing oncogene-induced p53-dependent and independent senescence as well as blocking myc-induced apoptosis [1]. HSP70 may enhance tumor development through the immune escape mechanism [36]. HSP70 overexpression has been reported in experimental models to enhance the tumorigenicity of transformed cells; its downregulation reduced tumorigenicity [37]. HSP70 induces carcinogenesis by stabilizing cyclin D1 expression, decreasing p53 protein, and suppressing apoptosis [1,36]. HSP70 may suppress p53 and allow the proliferation of cells with the upregulation of different oncogenes [38]. Oncogenes of the RAS signaling pathway activate two parallel pathways, including proliferation response and p53 pathway-dependent inhibition [38].
HSP70 inhibits intrinsic and extrinsic apoptosis pathways [39]. In the intrinsic mechanism, it inhibits the activation of Bax, which stabilizes the permeability of the mitochondrial membrane and protects it from the activity of factors inducing apoptosis [22]. When the extrinsic mechanism is present, HSP70 inhibits death-inducing signaling complex (DISC) [24]. Like HSP27, it is involved in caspase-dependent and caspase-independent mechanisms of apoptosis, inhibiting cell death by cathepsin modulation [1,36]. HSP70 blocks the apoptosis of cancer cells by the inhibition of caspases-3/9 activation and apoptosome formation [1]. The overexpression of HSP70 protects carcinoma cells from apoptosis by increasing the intracellular expression of the anti-apoptotic BCL-2 protein and decreasing the expression of cytochrome c [1,36,40]. HSP70 also facilitates apoptosis evasion by inhibiting PI3K/AKT signaling and initiating downstream AKT/mTOR signaling [1].
HSP70 is involved in EMT regulation [41]. Mortalin mitochondria-resident HSP70 isoforms activate EMT in different tumors, facilitating tumor invasion and metastasis [42]. Active HSP70 protein mediates invasion and EMT and comprises assisting HSP90-dependent activation of MMP-2-enhanced cell migration and metastasis [42]. The inhibition of HSP70 function reduces MMP-2 activity in cell lines and suppresses invasiveness [42].
HSP70 stabilizes the transcription factor H1F-1α, the main regulator of cancer cell hypoxia, and regulates angiogenesis [43]. A human umbilical vein endothelial cell (HUVEC) in vitro model noted that the IL-5 angiogenic activator is regulated by HSP70. An HSP70 knockdown led to the dysfunction of IL-5-induced cell proliferation, colony formation, and migration [44]. HSP70 promotes neovascularization via HIF-1/VEGF. Like VEGF, IL-5 induced HUVEC migration, colony formation in vitro, and microvessel formation in vivo [45,46]. HSP70 has a role in neoangiogenesis by the activation of HIF-1/VEGF by cancer cells and the activation of stromal endothelial cells with IL-5 help [44,45,46].

3.3. HSP90

As it interacts with ~400 client proteins (such as transcription factors and kinases), HSP90 has been studied in carcinogenesis [5,6,21,47]. HSP90-dependent proteins are divided into protein kinases (e.g., c-SRC/v-SRC, c RAF-1/v-RAF-1, HER2, EGFR, AKT, BRAF, MOK, and MET), transcription factors (e.g., p53 and HIF-1), and other proteins (e.g., hTERT) involved in the regulation of numerous cellular processes [5,6,19,48,49]. Some are crucial oncoproteins responsible for tumor cell survival and progression [5,6,12]. HSP90 is also involved in the evasion of apoptosis, immortalization, angiogenesis, and therapy resistance [1,19,21]. The interaction between HSP90 and components of the PI3K/AKT/mTOR signaling pathway suggests a role in the regulation of tumor cell survival [1,19]. The regulation of HIF and human telomerase reverse transcriptase (hTERT) activity by HSP90 suggests that it might have a role in tumorigenesis [19]. HSP90 may induce the immortalization of cancer cells due to the interaction with the hTERT promoter, increasing the level of telomerase in cancer cells [19]. The mechanisms through which HSP90 exerts its anti-apoptotic effects include a reduction in the activity of caspase-3 and caspase-8, a decrease in the quantity of tumor necrosis factor and FAS receptors, alterations in the levels of transcription factors p53 and NF-κB, and an imbalance in the pro- and anti-apoptotic proteins of the Bc1-2 family, favoring the latter [49,50]. This dominance of anti-apoptotic protein expression inhibits a decrease in the mitochondrial transmembrane potential [48,49]. HSP90 can block apoptosis by connecting with APAF-1, which inhibits the oligomerization of APAF-1 mediated by cytochrome c, and activates procaspase-9 [48]. HSP90 also blocks apoptosis through the formation of a triple complex with the pro-apoptotic kinase ASK1 and AKT. HSP 90 clients, such as AKT, may promote the anti-apoptotic influence of tumor cells through the downregulation of members belonging to the apoptotic machinery [19,48]. HSP90 also inhibits intracellular transport of the apoptosis-inducing factor (AIF) and endonuclease G from the mitochondrion to cytosol [51,52]. HSP90 inhibits apoptosis by p53 protein stabilization in both wild-type and mutated p53, protecting it from proteasome degradation [49].
HSP90 binds with some of cytoskeletal proteins, such as NCK-associated protein 1 (NCKAP1) and Wiskott–Aldrich syndrome protein family member 3 (WASF3), which are involved in the regulation of actin polymerization and are responsible for cell motility [53]. The mechanism of metastasis in various tumors, including HNSCC, is induced by the cooperation of extracellular HSP90 with MMP-2 and MMP-9, which disrupts the extracellular matrix and leads to the dissemination of tumor cells and formation of metastasis [2,54]. HSP90 is secreted by tumor cells, interacting with MMP-2 and MMP-9 [49]. The direct activation of MMP-2 by HSP90 increases tumor cell motility and invasiveness [2,5,54]. HSP90 engages with various receptors (including HER2, EGFR, and LPR1) to facilitate downstream signaling pathways related to tumor proliferation and metastasis, resembling the EMT phenotype [49].
HSP90 expression in OSCC is associated with nodal metastasis and an advanced tumor stage, suggesting that HSP90 may play a role in tumor progression. This may reflect HSP90 interaction with the NF-ĸB and PI3K/AKT pathways [6,12,47,51]. The association between HSP90 and AKT and hypoxia-inducible factor 1-alpha (HIF-1α) contributes to the functional stabilization of PI3K/AKT signaling vital for glycolysis-dependent tumor cell survival and participates in the control of metabolic reprograming [2] that enables tumor cells to sustain elevated proliferation rates and endure stressors. Radiotherapy may change the metabolism of tumor cells, leading to alterations in levels of metabolites, inducing oxidative stress and increasing the production of reactive oxygen species (ROS) that modify the network of cellular signaling pathways responsible for cell survival and apoptosis [6,12,52].

4. The Clinicopathological and Prognostic Significance of HSPs in HNSCC

The prognostic value of HSPs in HNSCC is unclear, although there are some correlations that medium and large HSPs correlate with more advanced disease and worse overall survival (OS). Previous clinical studies have been limited, usually single-center with fewer than 100 cases, have not distinguished HPV+ from HPV- tumors, and have used heterogenous methods, although they have usually used immunohistochemistry. Table 1 summarizes the significant (p < 0.05) clinicopathological findings for each HSP.
The most controversial findings are related to HSP 27, where some studies show that HSP27 overexpression is correlated with a lower pT stage, lower tumor grade, better prognosis, and longer OS [55,56,57,58,59,60,61]. In contrast, other studies suggest that HSP27 may be responsible for tumor growth and progression. Its increased expression was associated with more severe dysplasia (suggesting the role of HSP27 in EMT and tumor initiation) [34], a higher tumor grade [62], higher pT stage [59], higher clinical stage [63], lymph node metastasis [59,64], and worse OS [64].
HSP47 expression in HNSCC has yielded inconsistent findings. Its overexpression was reported to correlate with better OS [65] and with worse OS [66,67].
Only a few studies have reported on HSP60 in HNSCC. Worse OS, as well as a higher clinical stage, poor prognosis, and the presence of nodal metastasis have been reported to be associated with HSP60 overexpression [66,68].
Several studies have reported correlations between HSP70 and advanced HNSCC, including a higher tumor grade and stage, nodal metastasis, worse OS, and shorter DFS [22,40,69,70,71,72], although one study reported better OS [59].
HSP90 overexpression in HNSCC is associated with a worse stage, lymph node metastasis, tumor progression [12,73,74,75], and worse OS [50,66,74,75,76]. Multivariate Cox regression showed that HSP90 is a poor prognostic biomarker in HNSCC [73].
Similar observations were noted for HSP105 in HNSCC. Increased HSP105 expression is correlated with an advanced clinical stage [61], poor prognosis [77], and worse OS [50,66,77].
One report showed that HSP110 overexpression is associated with worse OS in patients with HNSCC [66].
In summary, although clinical studies have been limited, most studies that show consistency suggest that the overexpression of medium and large HSPs (HSP60, HSP70, HSP90, HSP105, and HSP110) is significantly associated with worse patient OS and related factors, such as nodal status and advanced stage [12,22,50,61,66,67,68,69,71,72,73,74,76,77]. An opposite association was observed for sHSPs, such as HSP27, where its increased expression was correlated with better survival [55,56,57,58]. Similar observations were reported for other clinicopathological features of HNSCC.
Table 1. The summary of studies evaluating HSP overexpression and clinicopathological findings in head and neck squamous cell carcinoma.
Table 1. The summary of studies evaluating HSP overexpression and clinicopathological findings in head and neck squamous cell carcinoma.
HSPFindings *MethodSampleSource
HSP27Tumor site, lower pT stageIHC50Gandour-Edwards et al., 1998 [60]
Better survivalIHC40Mese et al., 2002 [55]
Better survival, lower tumor grade, older ageIHC, WB79Muzio et al., 2004 [56]
Better OS, better prognosisIHC57Muzio et al., 2006 [57]
Better OS, better prognosis, lower tumor gradeIHC80Wang et al., 2009 [58]
Early clinical stage, lower tumor gradeIHC56Mohtasham et al., 2011 [61]
Worse OS, lymph node metastasisIHC50Kaigorodova et al., 2016 [64]
Higher clinical stageIHC, PCR44Karam et al., 2017 [63]
Higher tumor gradeIHC30Ajalyakeen et al., 2020 [62]
Better OS, better prognosis, older age, higher pT stage, lymph node metastasisIHC/TCGA158/112Borowczak et al., 2025 [59]
HSP47Better OS, lower tumor gradeIHC, WB50Song et al., 2017 [65]
Worse OSTCGA504Fan et al., 2020 [66]
Worse survival, shorter DFSIHC339Da Costa et al., 2023 [67]
HSP60Worse OSTCGA504Fan et al., 2020 [66]
Worse OS, poor prognosis, higher clinical stage,
lymph node metastasis
IHC79Zhou et al., 2023 [68]
HSP70Tumor size, tumor gradeIHC, WB38Kaur et al., 1995 [78]
Lower pT stageIHC50Gandour-Edwards et al., 1998 [60]
Shorter DFS, higher tumor grade, shorter transition timeIHC125Kaur et al., 1998 [69]
Higher tumor grade, no lymph node metastasisIHC41Lee et al., 2008 [70]
Shorter DFS, tumor location, lymph node metastasis, tumor gradeIHC90Choi et al., 2015 [71]
Lower clinical stage, no lymph node metastasis, smaller tumor sizeIHC50Taghavi et al., 2018 [79]
Higher tumor gradeIHC15Priyanka et al., 2019 [40]
Older ageIHC117Venugopal et al., 2022 [80]
Worse survival, poor prognosis, higher clinical stage, pT stageIHC104Ceylan et al., 2022 [72]
Higher TNM stage, higher tumor grade, lymph node metastasis, higher recurrence rate, shorter DFS, larger tumor sizeIHC50Elhendawy et al., 2023 [22]
Better OS, higher tumor gradeIHC/TCGA158/112Borowczak et al., 2025 [59]
HSP90Lymph node metastasis, worse survivalIHC36Chang et al., 2017 [74]
Worse survivalWB499Ono et al., 2018 [50]
Worse OSTCGA504Fan et al., 2020 [66]
Lymph node metastasisIHC58Shiraishi et al., 2021 [12]
Worse survivalIHC/TCGA97/98Santos et al., 2021 [76]
Higher pT stage, survival status, poor prognosisIHC56Bar et al., 2021 [73]
Worse OS, higher pT stage, lymph node metastasisIHC/TCGA68/499Zhang et al., 2022 [75]
Worse OS, higher pT stage, clinical stage, lymph node metastasisIHC, WB, PCR/TCGA59/419Tang et al., 2023 [20]
HSP105Advanced clinical stageIHC56Mohtasham et al., 2011 [61]
Worse survivalWB499Ono et al., 2018 [50]
Worse OSTCGA504Fan et al., 2020 [66]
Worse survival, poor prognosisIHC70Arvanitidou et al., 2020 [77]
HSP110Worse OSTCGA504Fan et al., 2020 [66]
Abbreviations: OS—overall survival, DFS—disease-free survival, IHC—immunohistochemistry, WB—Western blot, TCGA—The Cancer Genome Atlas, PCR—polymerase chain reaction. * This table presents only statistically significant correlations found in the cited studies (p < 0.05).

5. HSPs and Cancer Stem Cells

Cancer stem cells (CSCs) in HNSCC and their role in tumorigenesis, tumor progression, metastasis, tumor recurrence, and resistance to therapy are well-discussed, including their origin and facilitation by EMT [81,82,83,84]. Various genetic and epigenetic factors regulate their biological behavior, such as transcription factors, signaling pathways, and molecular chaperones [82,85]. The roles of transcription factors and signaling pathways are also well-discussed. The role of HSPs in CSCs is unknown [82,85].

5.1. HSP27

HSP27 is involved in tumorigenesis and the resistance of the tumor to chemotherapy and radiotherapy [86]. Because the phosphorylation of HSP27 facilitates the ubiquitination and proteasomal breakdown of proteins that drive stemness (including NANOG, OCT4, c-Myc, SOX2, and KLF4) in non-small-cell lung cancer (NSCLC) cells, the inactivation of p38 promotes the expression of CSC characteristics [87]. Phosphorylated HSP27 may act in various functions in cell biology, depending on the specific tumor [87].
Colorectal CD133+ CSCs with HSP27 expression prevent caspase-9 and caspase-3 cleavage in the cell death cascade, and the inhibition of HSP27 may enhance apoptosis in response to hypoxia [88]. Phosphorylation of HSP27 is enhanced in ALDH1+ breast cancer stem cells and is required for the functioning of CSCs in different cancers [1]. Experimental findings indicate that HSP27 and its phosphorylation play a role in the vasculogenic activity induced by EGF in breast cancer stem/progenitor cells [1,89]. Phosphorylated HSP27 induces EMT and activates NF-ĸB, thereby taking part in the maintenance of stem cells [46].
Oral CSC-like cells with increased HSP27 expression are associated with resistance to apoptosis, oxidative stress, and cytotoxic drugs [90]. Phosphorylated HSP27 appeared to provide protection against caspase-dependent apoptosis triggered by a lack of oxygen or serum depletion in CD133+ CSCs derived from various tumor, including OSCC [82,88]. This mechanism was linked to the p38 MAPK/MAPKAPK2/HSP27 pathway, which is inhibited by the protein phosphatase PP2A which is responsible for the dephosphorylation of HSP27 [82,88]. There is a report showing that the p38/HSP27 pathway enhances EMT induction in oral cancer cells [90].

5.2. HSP70

Increased expression of HSP70 in CSC-like cells has been reported in medulloblastoma and gastric and breast cancer [91,92,93]. HSP70 expression in breast cancer cells is associated with the expression of stem cell markers (CD44 and Sca1) and the high metastatic potential of tumor cells [92]. EMT is an important feature of CSCs, which determines their biological behavior. HSP70 plays a role in cancer stemness by increasing the expression of cancer stemness-associated proteins (such as vimentin, N-cadherin, MMP-2, MMP-9, Snail, Slug, Twist, and others) [94]. HSP70 is also associated with high CSC resistance to apoptosis [82]. HSP70 is expressed in the hypoxic regions of tumor tissue where EMT is present, indicating an association between HSP70 and the development and maintenance of cancer stemness [82]. GRP75 (mortalin) may contribute to EMT and cancer stemness, and correlate with the aggressive behavior of mammary gland carcinomas [82,95]. Tumor-initiating CSCs in breast cancer tissue may be a result of oncogenic mutations in normal stem cells or progenitors; increased levels of HSP70 are necessary to provide their ‘stemness’, contributing to tumor progression [82,96]. There may be a role of HSP70 in CSC resistance to therapy. An experimental study in lung and breast cancer showed that HSP chaperonage is enhanced in CSCs by the transcriptional upregulation of proteins in the HSP family, including HSP70 [97,98].

5.3. HSP90

HSP90 regulates the CSC phenotype and maintenance by interacting with proteins expressed by stem cells [82]. HSP90 cooperates with various transcriptional factors (including OCT4, NANOG, STAT3, and JAKs) which influence stem cell pluripotency [49]. For example, HSP90 abnormally activates EGFR, AKT, Hedgehog, WNT/β catenin, and Src, components of signaling pathways which promote the self-renewal of CSCs in malignant tissue [82,99,100]. WNT and Hedgehog signaling are strongly activated in HNSCC and are associated with CD133, SOX2, and CXCR4 expression, and most of the analyzed HNSCCs presented high expressions of HSP90 [73,101,102]. The direct interaction between HSP90 function and BMI1 gene transcription expression and CSC self-renewal in HNSCC was reported in one study [103]. HSP90 was reported to promote EMT via the activation of HIF1α and NF-қB and the enhancement of CSC accumulation [99,100]. The inhibition of HSP90 by Ku711 and Ku757 in HNSCC decreased stem cell markers (such as CD44 and ALDH) but increased E-cadherin [103]. Targeted therapy with HSP90 inhibitors may lead to a reduction in the population of ALDH+/CD44+ cells and downregulation of certain microRNAs related to the resistance of CSCs to chemotherapy [103]. Data on HSP90 chaperone function concerning activation of the AKT/MEK/ERK/JAK2/STAT3 signaling network in triple-negative breast cancer showed the generation of CSC-like cells, which demonstrated CD44 expression and high ALDH1 activity [104]. It has been proposed that HSP90 activates CSC mechanisms which upregulate the expression of MMP-2 and MMP-9 and that interaction with secreted MMPs induces local degradation of the extracellular matrix and enhances tumor cells migration, especially those with the CSC phenotype [82]. This is rational because MMPs increase the degradation of selected immunoreactive proteins at the surface of CSCs, thereby making CSCs less visible to the immunological system [82]. Many reports have demonstrated that molecular chaperones are determinants of cancer stemness development, which define the formation of the CSC phenotype and mediate the tumorigenic properties of CSCs [82,90,91]. Most CSC features are linked to the activity of molecular chaperones (especially HSP27, HSP70, and HSP90), which determine the migration, invasion, and resistance to radiotherapy and chemotherapy in solid tumors, including HNSCC [1,49,82,90,92,103].

6. HSP Inhibitors

HSPs are targetable as anticancer therapy. HSP inhibitors have been explored in treating some malignancies, including breast, lung, pancreatic, and gastrointestinal cancers, and melanoma [6,12,19,24,86,105,106]. However, studies on HNSCC are limited. HSP inhibitors are classified according to the specific HSP targeted: HSP27, HSP70, or HSP90. Frequently tested HSP inhibitors, potentially useful for future HNSCC therapy, are listed below:
(1)
HSP27 inhibitors: quercetin, RP101 (brivudine), PA11, PA50, OGX-427 (apatorsen), and ivermectin.
(2)
HSP70 inhibitors: apoptozole, triptolide, minnelide, artesunate, PES (pifithrin-μ), MKT-077, YM-01, and YM-08.
(3)
HSP90 inhibitors: geldanamycin, 17-AAG (tanespimycin), 17-DMAG (alvespimycin), IPI-504 (retaspimycin), IP-493, WK881, radicicol, NVP-AUY922 (luminespib), AT13387 (onalespib), GRP94 (glucose-regulated protein 94), STA-9090 (ganetespib), CNF-2024 (BIIB021), CUDC-305 (Debio 0932), PU-H71 (zelavespib), XL888, NVP-BEP80, sansalvamide, novobiocin, TAS-116 (pimitespib), HS-196, SNX-5422, Ku363, Ku711, and Ku757.

6.1. HSP27 Inhibitors

Three approaches have been investigated for targeting HSP27 in cancer cells [6,24,86]:
(1)
Small-molecule inhibitors—quercetin and RP101—which are under in vitro and in vivo evaluation; they increase anticancer impact in several cancer cell lines, including HNSCC [24,86,105,107,108].
(2)
Peptide aptamers—PA11 and PA50.
(3)
Antisense oligonucleotides—such as OGX-427 (apatorsen)—which interact directly with HSP27 have been investigated.
The most-studied HSP27 inhibitor is quercetin, a bioflavonoid with anticancer properties that targets heat shock transcriptional factor 1-related HSPs in many tumor cell lines [6,24,86,109]. There are several in vitro studies evaluating its use in HNSCC, especially in OSCC. It was reported that quercetin significantly inhibits cell proliferation and induces apoptosis in OSCC cell lines [110,111]. The mechanism of quercetin activity is complex and not yet fully understood. Studies have focused on the mechanisms underlying its anticancer effects, which may include the following:
(a)
The inhibition of cell invasion, migration, and colony formation by suppressing MMP-2 and MMP-9 activity [112].
(b)
The inhibition of the progression of OSCC by the activation of the miR- 1254/CD36 pathway [113].
(c)
The inhibition of glycolysis and cell proliferation of OSCC cells by suppressing the G3BP1/YWHAZ 919 axis [110].
(d)
Ferroptosis (programmed cell death characterized by iron dependency] may also be a mechanism [114].
(e)
The provision of an unfavorable microenvironment for HPV+ HNSCC by increasing reactive oxygen species production, decreasing tumor pH, and inhibiting tumor growth [115].
Quercetin provides synergy with chemotherapy in HNSCC. Quercetin with cisplatin or gemcitabine increases the chemosensitivity of oral tumor cells [109]. A similar effect was observed when quercetin was used with erlotinib (EGFR tyrosine kinase inhibitor) [116]. HNSCC often overexpresses EGFR, but resistance to anti-EGFR drugs develops, limiting the clinical efficacy of erlotinib. The addition of quercetin may block the development of resistance in HNSCC cells and enhance erlotinib efficacy in HNSCC [116]. However, quercetin use is limited by its low water solubility, instability under physiological conditions, and poor bioavailability [117]. Some researchers have proposed the use of photodynamic therapy to increase its bioavailability [117].
RP101 is a nucleoside unstudied in HNSCC, which may inhibit the function of HSP27 [105,118]. When it is bound to HSP27, the connection between HSP27 and AKT1, procaspase-3, and cytochrome c is reduced, impacting apoptosis [24]. RP101 is more effective when used in combination with other cytotoxic drugs. An in vivo study on rats with AH13r sarcoma cells showed that using it with cisplatin or cyclophosphamide had better antiproliferative efficacy compared to monotherapy [118].
Protein aptamers (such as PA11 and PA50) may impact HSP27 activity, increasing apoptosis in tumor cells [86]. PA11 and PA50 are negative mediators of HSP27 activity, binding to and disrupting the dimerization and oligomerization of HSP27, thereby suppressing its anti-apoptotic effect [119]. These peptide aptamers inhibit the growth of HNSCC tumors in vivo through cell cycle arrest [6,24,109,120]. They are ineffective as a monotherapy [24,86,119]; similar to small-molecule inhibitors of HSP27, protein aptamers demonstrate better efficacy when combined with other anticancer agents [86].
OGX-427 (apatorsen) is an antisense oligonucleotide. It inhibits HSP27 expression by increasing the apoptosis of tumor cells [6,121]. Combining it with conventional chemotherapy (eg gemcitabine and docetaxel) showed strong therapeutic activity, as well as with other HSP inhibitors [121,122]. The inhibition of HSP27 leads to better sensitization of NSCLC cells to erlotinib and standard chemotherapy [123]. Treatment of the multiple myeloma U266 cell line with OGX-427 or bortezomib decreased cell proliferation, enhanced apoptosis, and reduced HSP27 expression [124]. It was also found that HSP27 inhibition decreased BCL-2 expression and increased Bax expression [124]. Using OGX-427 as a radiosensitizer led to decreased neoangiogenesis and the inactivation of the AKT pathway [86]. HSP27 inhibition was also observed in the radioresistant HNSCC cell line SQ20B [125] and increased the cytotoxic effects of radiation on HNSCC cells [125].
Ivermectin is an antiparasitic drug [29]. Studies have reported that its use as an HSP27 inhibitor that may improve the targeting of oncogenes in cancer models [126]. It blocks the phosphorylation of HSP27 mediated by MAPKAPK2, disturbs the inhibition of SHPTP1 mediated by HSP27, and decreases the resistance to erlotinib [126]. It inhibited the growth of ESCC cells in culture. One study reported that it induced apoptosis in ESCC [127], while another reported that it suppressed tumor growth, esophageal metastasis, and improved chemosensitivity to cisplatin and 5-fluorouracil [128].
There is one active clinical trial evaluating HSP27 inhibitors in HNSCC (NCT05724329): quercetin use in combination with tislelizumab (anti-PD-1 antibody) and dasatinib in patients prior to tumor resection. In 2011, RP101 was noted to extend survival in pancreatic cancer patients in a phase II trial when used with gemcitabine. OGX-427 results were reported between 2016 and 2018 for patients with bladder, urothelial, prostate, and breast cancers [122,129,130].

6.2. HSP70 Inhibitors

HSP70 inhibitors may be classified into three elementary categories according to the target of binding: C-terminal peptide or substrate-binding domain (SBD), N-terminal ATP-binding domain (ABD), and HSP70 co-chaperones [2,24,131].
Apoptozole inhibits the ATPase activity of HSP70 by interacting with the ATP-binding site, inhibiting tumor cell proliferation and migration, and inducing cell apoptosis, with resultant decreased tumor growth in a xenograft model [131]. It promotes caspase-dependent apoptosis by inhibiting the connection between HSP70 and APAF-1, induces lysosome-mediated apoptosis, and decreases autophagy [131,132,133]. Its toxicity has been reported in a variety of tumor cells, including OSCC and breast and liver cancer [132,133].
Triptolide also decreases HSP70 expression. It induces apoptosis in HNSCC cell lines and inhibits the growth of tumors [134]. It inhibits the growth, invasion, migration, and angiogenesis of OSCC cells. It increases the radiosensitivity of nasopharyngeal cancer cells by inhibiting HSPA5 and inducing apoptosis [135]. It also inhibits the growth of OSCCs in tumor xenografts by suppressing the PD-L1 pathway [136]. Minnelide is a triptolide derivative that inhibits HSP70 activity. It reduces tumor growth in xenograft and metastasis models as well as reduces the expression of pro-survival proteins (e.g., c-Myc and survivin) and targets the NF-κB signaling pathway [137,138]. Its antitumor activity was reported in 2014 in two HPV+ HNSCC cell lines, inducing apoptosis and inhibiting tumor progression [139].
Artesunate is another inhibitor that decreases the expression and activity of HSP70 and has been reported to have an antitumor effect against HNSCC cells [140]. A study using cell lines showed an inhibitory effect on the growth and proliferation of HNSCC cells, causing cell cycle arrest [141]. Its cytotoxic effect was increased when used with cisplatin, and decreased levels of Rb and p-Rb were reported (directing the cell cycle to the G1/S phase, which is considered more sensitive for cisplatin) [141].
MKT-077 is an inhibitor obtained from rhodacyanine dye that impacts the interaction of nucleotide exchange factors (NEFs) with HSPA8 and HSPA9, inhibiting tumor growth and inducing cell senescence [142]. It inhibited tumor progression by releasing wild-type p53 from the HSP70–p53 complex and decreased tau protein levels in the cells [142]. The efficacy of derivatives of MKT-077, such as YM-01 and YM-08, have also been studied—YM-01 inhibited breast tumor and melanoma growth in xenograft models, and YM-08 reduced anti-tau activity in cancer cells [143].
Pifithrin-µ (PES) inhibits HSP70 by interacting with its substrate-binding domain [24]. In an animal model, PES was observed to inhibit HSP70 (HSPA1A/B)–client interaction, thereby suppressing tumor cell proliferation and migration, inducing apoptosis and cell cycle arrest, inhibiting tumor growth, downregulating the phosphorylation of AKT and ERK, dysregulating autophagy, and impairing lysosomal function. The potential of combining HSP70 inhibitors with radiotherapy, chemotherapy, or molecular-targeted therapy have occasionally been studied [135,144,145]. The combination of PES with oxaliplatin and cisplatin showed enhanced therapeutical responses in colorectal and cervical cancers [144,146].
The inhibition of HSP70 has been analyzed in clinical trials for various tumors; however, no clinical studies have been performed on HNSCC patients [145,147,148].

6.3. HSP90 Inhibitors

HSP90 is the most-studied target for HSP inhibitors. The first generation of HSP90 inhibitors was developed based on two potent natural inhibitors (geldanamycin and radicicol) with various subsequent derivatives (17-AAG, 17 DMAG, IPI-504, and WK881) [130,149]. A second generation of HSP90 inhibitors was derived from radicicol-based derivatives (e.g., luminespib, onalespib, glucose-regulated protein 94, and ganetespib) and purine or purine-like analogs that use X-ray crystallography to block HSP90 (e.g., BIIB021, Debio 0932, and zelavespib) [149].
Geldanamycin is the first HSP90 inhibitor. Its analogs (17-AAG, 17-DMAG, IPI-504, and IPI-493) are safer and better tolerated in clinical trials [106,130,149,150]. Geldanamycin and 17-AAG were tested in vitro on HNSCC cell lines and presented a suppressive function on tumor growth [151,152]. 17-AAG has been used in combination with radiotherapy as it enhances the sensitivity of OSCC cells to irradiation, thereby increasing the overall cytotoxic effect of therapy [153]. 17-AAG also has a synergistic effect when combined with cisplatin and induced apoptosis in cisplatin-resistant ESCC cell lines [154]. 17-AAG and retaspimycin hydrochloride (IPI-504) were also reported to be effective in glioma cell lines, where the suppression of tumor growth in immunocompromised mice was documented [106,155]. In the melanoma cell line, geldanamycin derivatives, such as 17-AAG and 17-DMAG, induced apoptosis by the activation of poly-ADP ribose polymerase (PARP) and several caspases (caspase-2, caspase-7, and caspase-9) [156]. Preclinical findings from various tumor cell lines and tumor xenograft models showed that HSP90 inhibitors, such as 17-AAG, are synergistic with taxanes in targeting NSCLC and breast cancer [19]. 17-DMAG and ganetespib reduce the viability and migration of OSCC cells [12]. Ganetespib inhibits glycolytic flux in HNSCC cells, resulting in metabolic failure and better antitumor effects [53]. It also enhances the cytotoxic effects of radiotherapy on HNSCC cells [53]. In myeloid cell lines (MCL-1) exposed to ganetespib, low expressions of anti-apoptotic proteins such as surviving and BCL-2 were observed, suggesting an apoptotic effect [157]. Ku363 is one of the novel HSP90 inhibitors that showed good in vivo efficacy in HNSCC therapy with lower toxicity than cisplatin [152]. Onalespib (AT13387) demonstrated radiosensitization in HNSCC both in vitro and in vivo [158]. It inhibits radiation-induced vasculogenesis, a typical process involved in tumor recurrence [158]. The radiosensitive effect of low doses of onalespib was confirmed in other studies on HNSCC xenografts [159,160]. Luminespib (NVP-AUY922) with cisplatin and radiotherapy was noted to be synergistic in HNSCC [161] as well as BIIB021, another HSP90 inhibitor [162]. BIIB021 demonstrated a strong antitumor effect by increasing apoptosis and enhancing cell cycle arrest [162]. A similar effect of combined therapy with HSP90 inhibition was observed with SNX5422, which increased cytotoxicity, chemosensitivity, and radiosensitivity in HNSCC [163].
NVP-BEP800 is a novel, synthetic, orally bioavailable N-terminal HSP90 inhibitor with potential selective antineoplastic activity in cancer cell lines [164]. Sansalvamide binds to N-terminal fragments of the middle domain of HSP90. These agents inhibited proliferation in melanoma cell lines [106]. Novobiocin is an aminocoumarin antibiotic that interacts with the C-terminal domain of HSP90 [130,164]. Analogs of novobiocin exhibit significant anti-proliferation influence on prostate cancer cell lines [165]. Novobiocin triggered the degradation of HSP90 client proteins such as CRAF, HER2, mutated p53, and SRC [164]. In vitro and in vivo studies demonstrated that HSP90 inhibitors suppress EMT and reduce cancer stem cells (CSCs) [82]. The inhibition of intracellular HSP90 activity by geldanamycin, 17-AAG, and 17-DMAG suppressed the proliferation and growth of CSCs [106,111,166,167]. HSP90 inhibitors can inhibit EMT together with EMT-evoked CSC accumulation, and may reverse the stemness of cancer cells by MET promotion [82]. CSCs in HNSCC are decreased by two novel HSP90 inhibitors, Ku711 and Ku757 [103]. An analysis of the signaling pathways demonstrated that these novel HSP90 inhibitors inhibit CSC pathways involved in migration, invasion, and proliferation [103].
In summary, novel HSP90 inhibitors are able to target CSCs in HNSCC and overcome resistance to radio/chemotherapy. There are several clinical trials using HSP90 inhibitors in breast, lung, and prostate cancer, melanoma, and GIST [106,168,169,170,171]. Only sporadic clinical trials have focused on HNSCC. Ganetespib was piloted in a study terminated (by the sponsor because of funding) in 2016 after four patients had received it prior to tumor resection [53]. A small phase I trial from 2015 to 2020 for patients with locoregionally advanced HNSCC used onalespib combined with cisplatin and radiotherapy (NCT02381535); however, the results are unpublished.

7. Summary

This is a comprehensive review of the HSP family, with particular attention to their roles in malignancy. This review includes an overview of the available information related to HNSCC. In vitro studies have shown that HSPs are involved in carcinogenesis, including tumor initiation, angiogenesis, inhibition of apoptosis, and tumor progression. HSPs are expressed in many malignancies, including HNSCC, and their expression is correlated with clinical behavior. The overexpression of many HSPs in HNSCC is associated with advanced tumors and, hence, worse survival and poor prognosis.
There are a few HSP inhibitors which have been evaluated in clinical trials not involving HNSCC, and no meaningful ones involving HNSCC. Most of the studies have not involved patients. No HSP inhibitors have been approved by the Food and Drug Administration (FDA), although pimitespib, an oral HSP90 inhibitor, was registered in Japan in 2022 for patients with GIST [172]. HSP inhibitors as a monotherapy are less useful than when used in lower doses combined with established anticancer agents or irradiation.
HSP inhibitors have potential as anticancer therapeutic agents; however, there are many reasons that attention is focused on elsewhere. Genomic alterations, epigenetic regulators, microRNAs, antibody–drug conjugates, advances in immunotherapy beyond checkpoint inhibitors, and the prospects of AI-assisted drug development are all more promising.

Author Contributions

Conceptualization, P.C.; writing—original draft preparation, P.C. and J.B.; writing—review and editing, P.C. and J.B.; visualization, P.C.; supervision, J.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The authors would like to express their appreciation to Piotr Grelewski for his help in preparing Figure 1.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Alberti, G.; Vergilio, G.; Paladino, L.; Barone, R.; Cappello, F.; Conway de Macario, E.; Macario, A.J.L.; Bucchieri, F.; Rappa, F. The Chaperone System in Breast Cancer: Roles and Therapeutic Prospects of the Molecular Chaperones Hsp27, Hsp60, Hsp70, and Hsp90. Int. J. Mol. Sci. 2022, 23, 7792. [Google Scholar] [CrossRef] [PubMed]
  2. Yang, S.; Xiao, H.; Cao, L. Recent Advances in Heat Shock Proteins in Cancer Diagnosis, Prognosis, Metabolism and Treatment. Biomed. Pharmacother. 2021, 142, 112074. [Google Scholar] [CrossRef] [PubMed]
  3. Wu, J.; Liu, T.; Rios, Z.; Mei, Q.; Lin, X.; Cao, S. Heat Shock Proteins and Cancer. Trends Pharmacol. Sci. 2017, 38, 226–256. [Google Scholar] [CrossRef]
  4. Hu, C.; Yang, J.; Qi, Z.; Wu, H.; Wang, B.; Zou, F.; Mei, H.; Liu, J.; Wang, W.; Liu, Q. Heat Shock Proteins: Biological Functions, Pathological Roles, and Therapeutic Opportunities. MedComm 2022, 3, e161. [Google Scholar] [CrossRef]
  5. Abdul, N.S.; Ahmad Alrashed, N.; Alsubaie, S.; Albluwi, H.; Badr Alsaleh, H.; Alageel, N.; Ghaleb Salma, R. Role of Extracellular Heat Shock Protein 90 Alpha in the Metastasis of Oral Squamous Cell Carcinoma: A Systematic Review. Cureus 2023, 15, e38514. [Google Scholar] [CrossRef]
  6. Chatterjee, S.; Burns, T.F. Targeting Heat Shock Proteins in Cancer: A Promising Therapeutic Approach. Int. J. Mol. Sci. 2017, 18, 1978. [Google Scholar] [CrossRef]
  7. Mikhailova, E.; Sokolenko, A.; Combs, S.E.; Shevtsov, M. Modulation of Heat Shock Proteins Levels in Health and Disease: An Integrated Perspective in Diagnostics and Therapy. Cells 2025, 14, 979. [Google Scholar] [CrossRef]
  8. Zuo, W.-F.; Pang, Q.; Zhu, X.; Yang, Q.-Q.; Zhao, Q.; He, G.; Han, B.; Huang, W. Heat Shock Proteins as Hallmarks of Cancer: Insights from Molecular Mechanisms to Therapeutic Strategies. J. Hematol. Oncol. 2024, 17, 81. [Google Scholar] [CrossRef]
  9. Hartl, F.U.; Bracher, A.; Hayer-Hartl, M. Molecular Chaperones in Protein Folding and Proteostasis. Nature 2011, 475, 324–332. [Google Scholar] [CrossRef]
  10. Jee, H. Size Dependent Classification of Heat Shock Proteins: A Mini-Review. J. Exerc. Rehabil. 2016, 12, 255–259. [Google Scholar] [CrossRef]
  11. Gu, C.; Fan, X.; Yu, W. Functional Diversity of Mammalian Small Heat Shock Proteins: A Review. Cells 2023, 12, 1947. [Google Scholar] [CrossRef]
  12. Shiraishi, N.; Onda, T.; Hayashi, K.; Onidani, K.; Watanabe, K.; Sekikawa, S.; Shibahara, T. Heat Shock Protein 90 as a Molecular Target for Therapy in Oral Squamous Cell Carcinoma: Inhibitory Effects of 17-DMAG and Ganetespib on Tumor Cells. Oncol. Rep. 2021, 45, 448–458. [Google Scholar] [CrossRef] [PubMed]
  13. Singh, M.K.; Shin, Y.; Han, S.; Ha, J.; Tiwari, P.K.; Kim, S.S.; Kang, I. Molecular Chaperonin HSP60: Current Understanding and Future Prospects. Int. J. Mol. Sci. 2024, 25, 5483. [Google Scholar] [CrossRef] [PubMed]
  14. Lianos, G.D.; Alexiou, G.A.; Mangano, A.; Mangano, A.; Rausei, S.; Boni, L.; Dionigi, G.; Roukos, D.H. The Role of Heat Shock Proteins in Cancer. Cancer Lett. 2015, 360, 114–118. [Google Scholar] [CrossRef] [PubMed]
  15. Ambrose, A.J.; Chapman, E. Function, Therapeutic Potential, and Inhibition of Hsp70 Chaperones. J. Med. Chem. 2021, 64, 7060–7082. [Google Scholar] [CrossRef]
  16. Wickramaratne, A.C.; Liao, J.-Y.; Doyle, S.M.; Hoskins, J.R.; Puller, G.; Scott, M.L.; Alao, J.P.; Obaseki, I.; Dinan, J.C.; Maity, T.K.; et al. J-Domain Proteins Form Binary Complexes with Hsp90 and Ternary Complexes with Hsp90 and Hsp70. J. Mol. Biol. 2023, 435, 168184. [Google Scholar] [CrossRef]
  17. Wei, H.; Zhang, Y.; Jia, Y.; Chen, X.; Niu, T.; Chatterjee, A.; He, P.; Hou, G. Heat Shock Protein 90: Biological Functions, Diseases, and Therapeutic Targets. MedComm 2024, 5, e470. [Google Scholar] [CrossRef]
  18. Wawrzynow, B.; Zylicz, A.; Zylicz, M. Chaperoning the Guardian of the Genome. The Two-Faced Role of Molecular Chaperones in P53 Tumor Suppressor Action. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 161–174. [Google Scholar] [CrossRef]
  19. Ren, X.; Li, T.; Zhang, W.; Yang, X. Targeting Heat-Shock Protein 90 in Cancer: An Update on Combination Therapy. Cells 2022, 11, 2556. [Google Scholar] [CrossRef]
  20. Tang, F.; Li, Y.; Pan, M.; Wang, Z.; Lu, T.; Liu, C.; Zhou, X.; Hu, G. HSP90AA1 Promotes Lymphatic Metastasis of Hypopharyngeal Squamous Cell Carcinoma by Regulating Epithelial-Mesenchymal Transition. Oncol. Res. 2023, 31, 787–803. [Google Scholar] [CrossRef]
  21. Lang, B.J.; Guerrero-Giménez, M.E.; Prince, T.L.; Ackerman, A.; Bonorino, C.; Calderwood, S.K. Heat Shock Proteins Are Essential Components in Transformation and Tumor Progression: Cancer Cell Intrinsic Pathways and Beyond. Int. J. Mol. Sci. 2019, 20, 4507. [Google Scholar] [CrossRef] [PubMed]
  22. Elhendawy, H.A. Clinical Implications of Heat Shock Protein 70 in Oral Carcinogenesis and Prediction of Progression and Recurrence in Oral Squamous Cell Carcinoma Patients: A Retrospective Clinicopathological Study. Eur. J. Med. Res. 2023, 28, 464. [Google Scholar] [CrossRef] [PubMed]
  23. Muschter, D.; Geyer, F.; Bauer, R.; Ettl, T.; Schreml, S.; Haubner, F. A Comparison of Cell Survival and Heat Shock Protein Expression after Radiation in Normal Dermal Fibroblasts, Microvascular Endothelial Cells, and Different Head and Neck Squamous Carcinoma Cell Lines. Clin. Oral Investig. 2018, 22, 2251–2262. [Google Scholar] [CrossRef]
  24. Somu, P.; Mohanty, S.; Basavegowda, N.; Yadav, A.K.; Paul, S.; Baek, K.-H. The Interplay between Heat Shock Proteins and Cancer Pathogenesis: A Novel Strategy for Cancer Therapeutics. Cancers 2024, 16, 638. [Google Scholar] [CrossRef]
  25. Konda, J.D.; Olivero, M.; Musiani, D.; Lamba, S.; Di Renzo, M.F. Heat-Shock Protein 27 (HSP27, HSPB1) Is Synthetic Lethal to Cells with Oncogenic Activation of MET, EGFR and BRAF. Mol. Oncol. 2017, 11, 599–611. [Google Scholar] [CrossRef]
  26. Wei, L.; Liu, T.-T.; Wang, H.-H.; Hong, H.-M.; Yu, A.L.; Feng, H.-P.; Chang, W.-W. Hsp27 Participates in the Maintenance of Breast Cancer Stem Cells through Regulation of Epithelial-Mesenchymal Transition and Nuclear Factor-κB. Breast Cancer Res. 2011, 13, R101. [Google Scholar] [CrossRef]
  27. Ramalingam, K.; Chawla, G.; Puri, A.; Krishnan, M.; Aneja, T.; Gill, K. Heat Shock Protein 27 (HSP27) as a Potential Prognostic Marker: Immunohistochemical Analysis of Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Cureus 2022, 14, e33020. [Google Scholar] [CrossRef]
  28. Asgharzadeh, F.; Moradi-Marjaneh, R.; Marjaneh, M.M. The Role of Heat Shock Protein 27 in Carcinogenesis and Treatment of Colorectal Cancer. Curr. Pharm. Des. 2022, 28, 2677–2685. [Google Scholar] [CrossRef]
  29. Lampros, M.; Vlachos, N.; Voulgaris, S.; Alexiou, G.A. The Role of Hsp27 in Chemotherapy Resistance. Biomedicines 2022, 10, 897. [Google Scholar] [CrossRef]
  30. Shiota, M.; Bishop, J.L.; Nip, K.M.; Zardan, A.; Takeuchi, A.; Cordonnier, T.; Beraldi, E.; Bazov, J.; Fazli, L.; Chi, K.; et al. Hsp27 Regulates Epithelial Mesenchymal Transition, Metastasis, and Circulating Tumor Cells in Prostate Cancer. Cancer Res. 2013, 73, 3109–3119. [Google Scholar] [CrossRef]
  31. Lu, W.; Wang, Y.; Gan, M.; Duan, Q. Prognosis and Predictive Value of Heat-Shock Proteins Expression in Oral Cancer: A PRISMA-Compliant Meta-Analysis. Medicine 2021, 100, e24274. [Google Scholar] [CrossRef]
  32. Zhu, Z.; Xu, X.; Yu, Y.; Graham, M.; Prince, M.E.; Carey, T.E.; Sun, D. Silencing Heat Shock Protein 27 Decreases Metastatic Behavior of Human Head and Neck Squamous Cell Cancer Cells In Vitro. Mol. Pharm. 2010, 7, 1283–1290. [Google Scholar] [CrossRef] [PubMed]
  33. Fang, Z.; Liang, W.; Luo, L. HSP27 Promotes Epithelial-Mesenchymal Transition through Activation of the β-Catenin/MMP3 Pathway in Pancreatic Ductal Adenocarcinoma Cells. Transl. Cancer Res. 2019, 8, 1268–1278. [Google Scholar] [CrossRef] [PubMed]
  34. Karri, R.L.; Subramanyam, R.V.; Venigella, A.; Babburi, S.; Pinisetti, S.; Rudraraju, A. Differential Expression of Heat Shock Protein 27 in Oral Epithelial Dysplasias and Squamous Cell Carcinoma. J. Microsc. Ultrastruct. 2020, 8, 62–68. [Google Scholar] [CrossRef] [PubMed]
  35. Zheng, S.; Liang, Y.; Li, L.; Tan, Y.; Liu, Q.; Liu, T.; Lu, X. Revisiting the Old Data of Heat Shock Protein 27 Expression in Squamous Cell Carcinoma: Enigmatic HSP27, More Than Heat Shock. Cells 2022, 11, 1665. [Google Scholar] [CrossRef]
  36. Vostakolaei, M.A.; Hatami-Baroogh, L.; Babaei, G.; Molavi, O.; Kordi, S.; Abdolalizadeh, J. Hsp70 in Cancer: A Double Agent in the Battle between Survival and Death. J. Cell Physiol. 2021, 236, 3420–3444. [Google Scholar] [CrossRef]
  37. Das, J.K.; Xiong, X.; Ren, X.; Yang, J.-M.; Song, J. Heat Shock Proteins in Cancer Immunotherapy. J. Oncol. 2019, 2019, 3267207. [Google Scholar] [CrossRef]
  38. Karri, R.L.; Subramanyam, R.V.; Venigella, A.; Babburi, S.; Pinisetti, S.; Amrutha, R.; Nelakurthi, H. Expression of Heat Shock Protein 70 in Oral Epithelial Dysplasia and Squamous Cell Carcinoma. J. Cancer Res. Ther. 2023, 19, 1939–1944. [Google Scholar] [CrossRef]
  39. Albakova, Z.; Armeev, G.A.; Kanevskiy, L.M.; Kovalenko, E.I.; Sapozhnikov, A.M. HSP70 Multi-Functionality in Cancer. Cells 2020, 9, 587. [Google Scholar] [CrossRef]
  40. Priyanka, K.P.; Majumdar, S.; Kotina, S.; Uppala, D.; Balla, H. Expression of Heat Shock Protein 70 in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma: An Immunohistochemical Study. Contemp. Clin. Dent. 2019, 10, 185–190. [Google Scholar] [CrossRef]
  41. Kasioumi, P.; Vrazeli, P.; Vezyraki, P.; Zerikiotis, S.; Katsouras, C.; Damalas, A.; Angelidis, C. Hsp70 (HSP70A1A) Downregulation Enhances the Metastatic Ability of Cancer Cells. Int. J. Oncol. 2019, 54, 821–832. [Google Scholar] [CrossRef]
  42. Perconti, G.; Maranto, C.; Romancino, D.P.; Rubino, P.; Feo, S.; Bongiovanni, A.; Giallongo, A. Pro-Invasive Stimuli and the Interacting Protein Hsp70 Favour the Route of Alpha-Enolase to the Cell Surface. Sci. Rep. 2017, 7, 3841. [Google Scholar] [CrossRef]
  43. Kabakov, A.E.; Yakimova, A.O. Hypoxia-Induced Cancer Cell Responses Driving Radioresistance of Hypoxic Tumors: Approaches to Targeting and Radiosensitizing. Cancers 2021, 13, 1102. [Google Scholar] [CrossRef]
  44. Park, S.L.; Chung, T.-W.; Kim, S.; Hwang, B.; Kim, J.M.; Lee, H.M.; Cha, H.-J.; Seo, Y.; Choe, S.Y.; Ha, K.-T.; et al. HSP70-1 Is Required for Interleukin-5-Induced Angiogenic Responses through eNOS Pathway. Sci. Rep. 2017, 7, 44687. [Google Scholar] [CrossRef]
  45. Kim, T.-K.; Na, H.J.; Lee, W.R.; Jeoung, M.H.; Lee, S. Heat Shock Protein 70-1A Is a Novel Angiogenic Regulator. Biochem. Biophys. Res. Commun. 2016, 469, 222–228. [Google Scholar] [CrossRef]
  46. Jang, J.; Kim, M.R.; Kim, T.-K.; Lee, W.R.; Kim, J.H.; Heo, K.; Lee, S. CLEC14a-HSP70-1A Interaction Regulates HSP70-1A-Induced Angiogenesis. Sci. Rep. 2017, 7, 10666. [Google Scholar] [CrossRef]
  47. Freilich, R.; Arhar, T.; Abrams, J.L.; Gestwicki, J.E. Protein-Protein Interactions in the Molecular Chaperone Network. Acc. Chem. Res. 2018, 51, 940–949. [Google Scholar] [CrossRef] [PubMed]
  48. Sumi, M.P.; Ghosh, A. Hsp90 in Human Diseases: Molecular Mechanisms to Therapeutic Approaches. Cells 2022, 11, 976. [Google Scholar] [CrossRef]
  49. Zhang, J.; Li, H.; Liu, Y.; Zhao, K.; Wei, S.; Sugarman, E.T.; Liu, L.; Zhang, G. Targeting HSP90 as a Novel Therapy for Cancer: Mechanistic Insights and Translational Relevance. Cells 2022, 11, 2778. [Google Scholar] [CrossRef] [PubMed]
  50. Ono, K.; Eguchi, T.; Sogawa, C.; Calderwood, S.K.; Futagawa, J.; Kasai, T.; Seno, M.; Okamoto, K.; Sasaki, A.; Kozaki, K.-I. HSP-Enriched Properties of Extracellular Vesicles Involve Survival of Metastatic Oral Cancer Cells. J. Cell Biochem. 2018, 119, 7350–7362. [Google Scholar] [CrossRef] [PubMed]
  51. Basset, C.A.; Rappa, F.; Barone, R.; Florena, A.M.; Porcasi, R.; Conway de Macario, E.; Macario, A.J.L.; Leone, A. The Chaperone System in Salivary Glands: Hsp90 Prospects for Differential Diagnosis and Treatment of Malignant Tumors. Int. J. Mol. Sci. 2022, 23, 9317. [Google Scholar] [CrossRef]
  52. Forman, H.J.; Zhang, H. Targeting Oxidative Stress in Disease: Promise and Limitations of Antioxidant Therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef]
  53. Chen, F.; Tang, C.; Yang, F.; Ekpenyong, A.; Qin, R.; Xie, J.; Momen-Heravi, F.; Saba, N.F.; Teng, Y. HSP90 Inhibition Suppresses Tumor Glycolytic Flux to Potentiate the Therapeutic Efficacy of Radiotherapy for Head and Neck Cancer. Sci. Adv. 2024, 10, eadk3663. [Google Scholar] [CrossRef]
  54. Costa, T.E.M.M.; Raghavendra, N.M.; Penido, C. Natural Heat Shock Protein 90 Inhibitors in Cancer and Inflammation. Eur. J. Med. Chem. 2020, 189, 112063. [Google Scholar] [CrossRef] [PubMed]
  55. Mese, H.; Sasaki, A.; Nakayama, S.; Yoshioka, N.; Yoshihama, Y.; Kishimoto, K.; Matsumura, T. Prognostic Significance of Heat Shock Protein 27 (HSP27) in Patients with Oral Squamous Cell Carcinoma. Oncol. Rep. 2002, 9, 341–344. [Google Scholar] [CrossRef] [PubMed]
  56. Muzio, L.L.; Leonardi, R.; Mariggiò, M.A.; Mignogna, M.D.; Rubini, C.; Vinella, A.; Pannone, G.; Giannetti, L.; Serpico, R.; Testa, N.F.; et al. HSP 27 as Possible Prognostic Factor in Patients with Oral Squamous Cell Carcinoma. Histol. Histopathol. 2004, 19, 119–128. [Google Scholar] [CrossRef] [PubMed]
  57. Muzio, L.L.; Campisi, G.; Farina, A.; Rubini, C.; Ferrari, F.; Falaschini, S.; Leonardi, R.; Carinci, F.; Stalbano, S.; De Rosa, G. Prognostic Value of HSP27 in Head and Neck Squamous Cell Carcinoma: A Retrospective Analysis of 57 Tumours. Anticancer Res. 2006, 26, 1343–1349. [Google Scholar]
  58. Wang, A.; Liu, X.; Sheng, S.; Ye, H.; Peng, T.; Shi, F.; Crowe, D.L.; Zhou, X. Dysregulation of Heat Shock Protein 27 Expression in Oral Tongue Squamous Cell Carcinoma. BMC Cancer 2009, 9, 167. [Google Scholar] [CrossRef]
  59. Borowczak, J.; Łaszczych, D.; Czyżnikiewicz, A.; Marszałek, A.; Szylberg, Ł.; Bodnar, M. Low Expression of HSP27 and HSP70 Predicts Poor Prognosis in Laryngeal Squamous Cell Carcinoma. J. Cancer Res. Clin. Oncol. 2025, 151, 264. [Google Scholar] [CrossRef]
  60. Gandour-Edwards, R.; Trock, B.J.; Gumerlock, P.; Donald, P.J. Heat Shock Protein and P53 Expression in Head and Neck Squamous Cell Carcinoma. Otolaryngol. Head Neck Surg. 1998, 118, 610–615. [Google Scholar] [CrossRef]
  61. Mohtasham, N.; Babakoohi, S.; Montaser-Kouhsari, L.; Memar, B.; Salehinejad, J.; Rahpeyma, A.; Khageh-Ahmady, S.; Marouzi, P.; Firooz, A.; Pazoki-Toroudi, H.; et al. The Expression of Heat Shock Proteins 27 and 105 in Squamous Cell Carcinoma of the Tongue and Relationship with Clinicopathological Index. Med. Oral Patol. Oral Cir. Bucal 2011, 16, e730–e735. [Google Scholar] [CrossRef]
  62. Ajalyakeen, H.; Almohareb, M.; Al-Assaf, M. Overexpression of Heat Shock Protein 27 (HSP-27) Is Associated with Bad Prognosis in Oral Squamous Cell Carcinoma. Dent. Med. Probl. 2020, 57, 227–231. [Google Scholar] [CrossRef]
  63. Karam, J.; Fadous-Khalifé, M.C.; Tannous, R.; Fakhreddine, S.; Massoud, M.; Hadchity, J.; Aftimos, G.; Hadchity, E. Role of Krüppel-like Factor 4 and Heat Shock Protein 27 in Cancer of the Larynx. Mol. Clin. Oncol. 2017, 7, 808–814. [Google Scholar] [CrossRef] [PubMed]
  64. Kaigorodova, E.V.; Zavyalova, M.V.; Bychkov, V.A.; Perelmuter, V.M.; Choynzonov, E.L. Functional State of the Hsp27 Chaperone as a Molecular Marker of an Unfavorable Course of Larynx Cancer. Cancer Biomark. 2016, 17, 145–153. [Google Scholar] [CrossRef] [PubMed]
  65. Song, X.; Liao, Z.; Zhou, C.; Lin, R.; Lu, J.; Cai, L.; Tan, X.; Zeng, W.; Lu, X.; Zheng, W.; et al. HSP47 Is Associated with the Prognosis of Laryngeal Squamous Cell Carcinoma by Inhibiting Cell Viability and Invasion and Promoting Apoptosis. Oncol. Rep. 2017, 38, 2444–2452. [Google Scholar] [CrossRef] [PubMed][Green Version]
  66. Fan, G.; Tu, Y.; Wu, N.; Xiao, H. The Expression Profiles and Prognostic Values of HSPs Family Members in Head and Neck Cancer. Cancer Cell Int. 2020, 20, 220. [Google Scholar] [CrossRef]
  67. da Costa, B.C.; Dourado, M.R.; de Moraes, E.F.; Panini, L.M.; Elseragy, A.; Téo, F.H.; Guimarães, G.N.; Machado, R.A.; Risteli, M.; Gurgel Rocha, C.A.; et al. Overexpression of Heat-Shock Protein 47 Impacts Survival of Patients with Oral Squamous Cell Carcinoma. J. Oral Pathol. Med. 2023, 52, 601–609. [Google Scholar] [CrossRef]
  68. Zhou, Y.; Tang, Y.; Luo, J.; Yang, Y.; Zang, H.; Ma, J.; Fan, S.; Wen, Q. High Expression of HSP60 and Survivin Predicts Poor Prognosis for Oral Squamous Cell Carcinoma Patients. BMC Oral Health 2023, 23, 629. [Google Scholar] [CrossRef]
  69. Kaur, J.; Srivastava, A.; Ralhan, R. Expression of 70-kDa Heat Shock Protein in Oral Lesions: Marker of Biological Stress or Pathogenicity. Oral Oncol. 1998, 34, 496–501. [Google Scholar] [CrossRef]
  70. Lee, S.-S.; Tsai, C.-H.; Ho, Y.-C.; Chang, Y.-C. The Upregulation of Heat Shock Protein 70 Expression in Areca Quid Chewing-Associated Oral Squamous Cell Carcinomas. Oral Oncol. 2008, 44, 884–890. [Google Scholar] [CrossRef]
  71. Choi, H.G.; Kim, J.-S.; Kim, K.H.; Kim, K.H.; Sung, M.-W.; Choe, J.-Y.; Kim, J.E.; Jung, Y.H. Expression of Hypoxic Signaling Markers in Head and Neck Squamous Cell Carcinoma and Its Clinical Significance. Eur. Arch. Otorhinolaryngol. 2015, 272, 219–228. [Google Scholar] [CrossRef] [PubMed]
  72. Ceylan, O.; Arslan, R. The Importance of Heat Shock-Related 70-kDa Protein 2 Expression in Laryngeal Squamous Cell Carcinomas. Eurasian J. Med. 2022, 54, 165–172. [Google Scholar] [CrossRef] [PubMed]
  73. Bar, J.K.; Cierpikowski, P.; Lis-Nawara, A.; Duc, P.; Hałoń, A.; Radwan-Oczko, M. Comparison of P53, HSP90, E-Cadherin and HPV in Oral Lichen Planus and Oral Squamous Cell Carcinoma. Acta Otorhinolaryngol. Ital. 2021, 41, 514–522. [Google Scholar] [CrossRef] [PubMed]
  74. Chang, W.-C.; Tsai, P.-T.; Lin, C.-K.; Shieh, Y.-S.; Chen, Y.-W. Expression Pattern of Heat Shock Protein 90 in Patients with Oral Squamous Cell Carcinoma in Northern Taiwan. Br. J. Oral Maxillofac. Surg. 2017, 55, 281–286. [Google Scholar] [CrossRef]
  75. Zhang, H.; Yin, X.; Zhang, X.; Zhou, M.; Xu, W.; Wei, Z.; Song, C.; Han, S.; Han, W. HSP90AB1 Promotes the Proliferation, Migration, and Glycolysis of Head and Neck Squamous Cell Carcinoma. Technol. Cancer Res. Treat. 2022, 21, 1–14. [Google Scholar] [CrossRef]
  76. Santos, E.M.; Fraga, C.A.d.C.; Xavier, A.R.E.d.O.; Xavier, M.A.d.S.; Souza, M.G.; Jesus, S.F.d.; Paula, A.M.B.d.; Farias, L.C.; Santos, S.H.S.; Santos, T.G.; et al. Prion Protein Is Associated with a Worse Prognosis of Head and Neck Squamous Cell Carcinoma. J. Oral Pathol. Med. 2021, 50, 985–994. [Google Scholar] [CrossRef]
  77. Arvanitidou, S.; Martinelli-Kläy, C.P.; Samson, J.; Lobrinus, J.A.; Dulguerov, N.; Lombardi, T. HSP105 Expression in Oral Squamous Cell Carcinoma: Correlation with Clinicopathological Features and Outcomes. J. Oral Pathol. Med. 2020, 49, 665–671. [Google Scholar] [CrossRef]
  78. Kaur, J.; Ralhan, R. Differential Expression of 70-kDa Heat Shock-Protein in Human Oral Tumorigenesis. Int. J. Cancer 1995, 63, 774–779. [Google Scholar] [CrossRef]
  79. Taghavi, N.; Mohsenifar, Z.; Baghban, A.A.; Arjomandkhah, A. CD20+ Tumor Infiltrating B Lymphocyte in Oral Squamous Cell Carcinoma: Correlation with Clinicopathologic Characteristics and Heat Shock Protein 70 Expression. Pathol. Res. Int. 2018, 2018, 4810751. [Google Scholar] [CrossRef]
  80. Venugopal, D.C.; Ravindran, S.; Shyamsundar, V.; Sankarapandian, S.; Krishnamurthy, A.; Sivagnanam, A.; Madhavan, Y.; Ramshankar, V. Integrated Proteomics Based on 2D Gel Electrophoresis and Mass Spectrometry with Validations: Identification of a Biomarker Compendium for Oral Submucous Fibrosis-An Indian Study. J. Pers. Med. 2022, 12, 208. [Google Scholar] [CrossRef]
  81. Fukumoto, C.; Uchida, D.; Kawamata, H. Diversity of the Origin of Cancer Stem Cells in Oral Squamous Cell Carcinoma and Its Clinical Implications. Cancers 2022, 14, 3588. [Google Scholar] [CrossRef] [PubMed]
  82. Kabakov, A.; Yakimova, A.; Matchuk, O. Molecular Chaperones in Cancer Stem Cells: Determinants of Stemness and Potential Targets for Antitumor Therapy. Cells 2020, 9, 892. [Google Scholar] [CrossRef] [PubMed]
  83. Zisis, V.; Venou, M.; Poulopoulos, A.; Andreadis, D. Cancer Stem Cells in Head and Neck Squamous Cell Carcinoma: Treatment Modalities. Balk. J. Dent. Med. 2021, 25, 73–79. [Google Scholar] [CrossRef]
  84. Hasegawa, T.; Nakashiro, K.-I.; Fukumoto, C.; Hyodo, T.; Sawatani, Y.; Shimura, M.; Kamimura, R.; Kuribayashi, N.; Fujita, A.; Uchida, D.; et al. Oral Squamous Cell Carcinoma May Originate from Bone Marrow-Derived Stem Cells. Oncol. Lett. 2021, 21, 170. [Google Scholar] [CrossRef]
  85. Lettini, G.; Lepore, S.; Crispo, F.; Sisinni, L.; Esposito, F.; Landriscina, M. Heat Shock Proteins in Cancer Stem Cell Maintenance: A Potential Therapeutic Target? Histol. Histopathol. 2020, 35, 25–37. [Google Scholar] [CrossRef]
  86. Choi, S.-K.; Kam, H.; Kim, K.-Y.; Park, S.I.; Lee, Y.-S. Targeting Heat Shock Protein 27 in Cancer: A Druggable Target for Cancer Treatment? Cancers 2019, 11, 1195. [Google Scholar] [CrossRef]
  87. Fang, Y.; Wang, J.; Wang, G.; Zhou, C.; Wang, P.; Zhao, S.; Zhao, S.; Huang, S.; Su, W.; Jiang, P.; et al. Inactivation of P38 MAPK Contributes to Stem Cell-like Properties of Non-Small Cell Lung Cancer. Oncotarget 2017, 8, 26702–26717. [Google Scholar] [CrossRef]
  88. Lin, S.-P.; Lee, Y.-T.; Wang, J.-Y.; Miller, S.A.; Chiou, S.-H.; Hung, M.-C.; Hung, S.-C. Survival of Cancer Stem Cells under Hypoxia and Serum Depletion via Decrease in PP2A Activity and Activation of P38-MAPKAPK2-Hsp27. PLoS ONE 2012, 7, e49605. [Google Scholar] [CrossRef]
  89. Lee, C.-H.; Wu, Y.-T.; Hsieh, H.-C.; Yu, Y.; Yu, A.L.; Chang, W.-W. Epidermal Growth Factor/Heat Shock Protein 27 Pathway Regulates Vasculogenic Mimicry Activity of Breast Cancer Stem/Progenitor Cells. Biochimie 2014, 104, 117–126. [Google Scholar] [CrossRef]
  90. Chen, S.-F.; Nieh, S.; Jao, S.-W.; Liu, C.-L.; Wu, C.-H.; Chang, Y.-C.; Yang, C.-Y.; Lin, Y.-S. Quercetin Suppresses Drug-Resistant Spheres via the P38 MAPK-Hsp27 Apoptotic Pathway in Oral Cancer Cells. PLoS ONE 2012, 7, e49275. [Google Scholar] [CrossRef]
  91. Yu, D.; Shin, H.-S.; Choi, G.; Lee, Y.C. Proteomic Analysis of CD44(+) and CD44(-) Gastric Cancer Cells. Mol. Cell Biochem. 2014, 396, 213–220. [Google Scholar] [CrossRef]
  92. Conner, C.; Lager, T.W.; Guldner, I.H.; Wu, M.-Z.; Hishida, Y.; Hishida, T.; Ruiz, S.; Yamasaki, A.E.; Gilson, R.C.; Belmonte, J.C.I.; et al. Cell Surface GRP78 Promotes Stemness in Normal and Neoplastic Cells. Sci. Rep. 2020, 10, 3474. [Google Scholar] [CrossRef] [PubMed]
  93. Ronci, M.; Catanzaro, G.; Pieroni, L.; Po, A.; Besharat, Z.M.; Greco, V.; Levi Mortera, S.; Screpanti, I.; Ferretti, E.; Urbani, A. Proteomic Analysis of Human Sonic Hedgehog (SHH) Medulloblastoma Stem-like Cells. Mol. Biosyst. 2015, 11, 1603–1611. [Google Scholar] [CrossRef] [PubMed]
  94. Gupta, N.; Jagadish, N.; Surolia, A.; Suri, A. Heat Shock Protein 70-2 (HSP70-2) a Novel Cancer Testis Antigen That Promotes Growth of Ovarian Cancer. Am. J. Cancer Res. 2017, 7, 1252–1269, Erratum in Am. J. Cancer Res. 2020, 10, 1919–1920. [Google Scholar]
  95. Wei, B.; Cao, J.; Tian, J.-H.; Yu, C.-Y.; Huang, Q.; Yu, J.-J.; Ma, R.; Wang, J.; Xu, F.; Wang, L.-B. Mortalin Maintains Breast Cancer Stem Cells Stemness via Activation of Wnt/GSK3β/β-Catenin Signaling Pathway. Am. J. Cancer Res. 2021, 11, 2696–2716. [Google Scholar]
  96. Kabakov, A.E.; Gabai, V.L. HSP70s in Breast Cancer: Promoters of Tumorigenesis and Potential Targets/Tools for Therapy. Cells 2021, 10, 3446. [Google Scholar] [CrossRef]
  97. Hyun, S.Y.; Le, H.T.; Min, H.-Y.; Pei, H.; Lim, Y.; Song, I.; Nguyen, Y.T.K.; Hong, S.; Han, B.W.; Lee, H.-Y. Evodiamine Inhibits Both Stem Cell and Non-Stem-Cell Populations in Human Cancer Cells by Targeting Heat Shock Protein 70. Theranostics 2021, 11, 2932–2952. [Google Scholar] [CrossRef]
  98. Tsai, C.-H.; Weng, J.-R.; Lin, H.-W.; Lu, M.-T.; Liu, Y.-C.; Chu, P.-C. Targeting Triple Negative Breast Cancer Stem Cells by Heat Shock Protein 70 Inhibitors. Cancers 2022, 14, 4898. [Google Scholar] [CrossRef]
  99. Prager, B.C.; Xie, Q.; Bao, S.; Rich, J.N. Cancer Stem Cells: The Architects of the Tumor Ecosystem. Cell Stem Cell 2019, 24, 41–53. [Google Scholar] [CrossRef]
  100. Jolly, M.K.; Celià-Terrassa, T. Dynamics of Phenotypic Heterogeneity Associated with EMT and Stemness during Cancer Progression. J. Clin. Med. 2019, 8, 1542. [Google Scholar] [CrossRef]
  101. Cierpikowski, P.; Lis-Nawara, A.; Bar, J. Prognostic Value of WNT1, NOTCH1, PDGFRβ, and CXCR4 in Oral Squamous Cell Carcinoma. Anticancer Res. 2023, 43, 591–602. [Google Scholar] [CrossRef]
  102. Cierpikowski, P.; Lis-Nawara, A.; Bar, J. SHH Expression Is Significantly Associated With Cancer Stem Cell Markers in Oral Squamous Cell Carcinoma. Anticancer Res. 2021, 41, 5405–5413. [Google Scholar] [CrossRef]
  103. Subramanian, C.; Kovatch, K.J.; Sim, M.W.; Wang, G.; Prince, M.E.; Carey, T.E.; Davis, R.; Blagg, B.S.J.; Cohen, M.S. Novel C-Terminal Heat Shock Protein 90 Inhibitors (KU711 and Ku757) Are Effective in Targeting Head and Neck Squamous Cell Carcinoma Cancer Stem Cells. Neoplasia 2017, 19, 1003–1011. [Google Scholar] [CrossRef]
  104. Cho, T.-M.; Kim, J.Y.; Kim, Y.-J.; Sung, D.; Oh, E.; Jang, S.; Farrand, L.; Hoang, V.-H.; Nguyen, C.-T.; Ann, J.; et al. C-Terminal HSP90 Inhibitor L80 Elicits Anti-Metastatic Effects in Triple-Negative Breast Cancer via STAT3 Inhibition. Cancer Lett. 2019, 447, 141–153. [Google Scholar] [CrossRef] [PubMed]
  105. Heinrich, J.C.; Donakonda, S.; Haupt, V.J.; Lennig, P.; Zhang, Y.; Schroeder, M. New HSP27 Inhibitors Efficiently Suppress Drug Resistance Development in Cancer Cells. Oncotarget 2016, 7, 68156–68169. [Google Scholar] [CrossRef] [PubMed]
  106. Rastogi, S.; Joshi, A.; Sato, N.; Lee, S.; Lee, M.-J.; Trepel, J.B.; Neckers, L. An Update on the Status of HSP90 Inhibitors in Cancer Clinical Trials. Cell Stress Chaperones 2024, 29, 519–539. [Google Scholar] [CrossRef] [PubMed]
  107. Li, J.; Tang, C.; Li, L.; Li, R.; Fan, Y. Quercetin Blocks T-AUCB-Induced Autophagy by Hsp27 and Atg7 Inhibition in Glioblastoma Cells In Vitro. J. Neurooncol. 2016, 129, 39–45. [Google Scholar] [CrossRef]
  108. Chen, X.; Dong, X.-S.; Gao, H.-Y.; Jiang, Y.-F.; Jin, Y.-L.; Chang, Y.-Y.; Chen, L.-Y.; Wang, J.-H. Suppression of HSP27 Increases the Anti-Tumor Effects of Quercetin in Human Leukemia U937 Cells. Mol. Med. Rep. 2016, 13, 689–696. [Google Scholar] [CrossRef]
  109. Elattar, T.M.; Virji, A.S. The Inhibitory Effect of Curcumin, Genistein, Quercetin and Cisplatin on the Growth of Oral Cancer Cells In Vitro. Anticancer Res. 2000, 20, 1733–1738. [Google Scholar]
  110. Huang, C.-F.; Liu, S.-H.; Ho, T.-J.; Lee, K.-I.; Fang, K.-M.; Lo, W.-C.; Liu, J.-M.; Wu, C.-C.; Su, C.-C. Quercetin Induces Tongue Squamous Cell Carcinoma Cell Apoptosis via the JNK Activation-Regulated ERK/GSK-3α/β-Mediated Mitochondria-Dependent Apoptotic Signaling Pathway. Oncol. Lett. 2022, 23, 78. [Google Scholar] [CrossRef]
  111. Son, H.-K.; Kim, D. Quercetin Induces Cell Cycle Arrest and Apoptosis in YD10B and YD38 Oral Squamous Cell Carcinoma Cells. Asian Pac. J. Cancer Prev. 2023, 24, 283–289. [Google Scholar] [CrossRef]
  112. Chan, C.-Y.; Lien, C.-H.; Lee, M.-F.; Huang, C.-Y. Quercetin Suppresses Cellular Migration and Invasion in Human Head and Neck Squamous Cell Carcinoma (HNSCC). Biomedicine 2016, 6, 15. [Google Scholar] [CrossRef]
  113. Chen, L.; Xia, J.-S.; Wu, J.-H.; Chen, Y.-G.; Qiu, C.-J. Quercetin Suppresses Cell Survival and Invasion in Oral Squamous Cell Carcinoma via the miR-1254/CD36 Cascade In Vitro. Hum. Exp. Toxicol. 2021, 40, 1413–1421. [Google Scholar] [CrossRef]
  114. Zhu, Y.-W.; Liu, C.-L.; Li, X.-M.; Shang, Y. Quercetin Induces Ferroptosis by Inactivating mTOR/S6KP70 Pathway in Oral Squamous Cell Carcinoma. Toxicol. Mech. Methods 2024, 34, 669–675. [Google Scholar] [CrossRef] [PubMed]
  115. Zhuang, Y.; Coppock, J.D.; Haugrud, A.B.; Lee, J.H.; Messerli, S.M.; Miskimins, W.K. Dichloroacetate and Quercetin Prevent Cell Proliferation, Induce Cell Death and Slow Tumor Growth in a Mouse Model of HPV-Positive Head and Neck Cancer. Cancers 2024, 16, 1525. [Google Scholar] [CrossRef] [PubMed]
  116. Chan, C.-Y.; Hong, S.-C.; Chang, C.-M.; Chen, Y.-H.; Liao, P.-C.; Huang, C.-Y. Oral Squamous Cell Carcinoma Cells with Acquired Resistance to Erlotinib Are Sensitive to Anti-Cancer Effect of Quercetin via Pyruvate Kinase M2 (PKM2). Cells 2023, 12, 179. [Google Scholar] [CrossRef] [PubMed]
  117. Liu, Q.; Jiang, D.; Zhang, S.; Ru, Y.; Li, J.; Guo, P.; Jiao, W.; Miao, J.; Sun, L.; Chen, M.; et al. Light-Activated Photosensitizer/Quercetin Co-Loaded Extracellular Vesicles for Precise Oral Squamous Cell Carcinoma Therapy. Int. J. Pharm. 2025, 671, 125224. [Google Scholar] [CrossRef]
  118. Heinrich, J.-C.; Tuukkanen, A.; Schroeder, M.; Fahrig, T.; Fahrig, R. RP101 (Brivudine) Binds to Heat Shock Protein HSP27 (HSPB1) and Enhances Survival in Animals and Pancreatic Cancer Patients. J. Cancer Res. Clin. Oncol. 2011, 137, 1349–1361. [Google Scholar] [CrossRef]
  119. McConnell, J.R.; McAlpine, S.R. Heat Shock Proteins 27, 40, and 70 as Combinational and Dual Therapeutic Cancer Targets. Bioorg. Med. Chem. Lett. 2013, 23, 1923–1928. [Google Scholar] [CrossRef]
  120. Gibert, B.; Hadchity, E.; Czekalla, A.; Aloy, M.-T.; Colas, P.; Rodriguez-Lafrasse, C.; Arrigo, A.-P.; Diaz-Latoud, C. Inhibition of Heat Shock Protein 27 (HspB1) Tumorigenic Functions by Peptide Aptamers. Oncogene 2011, 30, 3672–3681. [Google Scholar] [CrossRef]
  121. Ko, A.H.; Murphy, P.B.; Peyton, J.D.; Shipley, D.L.; Al-Hazzouri, A.; Rodriguez, F.A.; Womack, M.S.; Xiong, H.Q.; Waterhouse, D.M.; Tempero, M.A.; et al. A Randomized, Double-Blinded, Phase II Trial of Gemcitabine and Nab-Paclitaxel Plus Apatorsen or Placebo in Patients with Metastatic Pancreatic Cancer: The RAINIER Trial. Oncologist 2017, 22, 1427–e129. [Google Scholar] [CrossRef] [PubMed]
  122. Rosenberg, J.E.; Hahn, N.M.; Regan, M.M.; Werner, L.; Alva, A.; George, S.; Picus, J.; Alter, R.; Balar, A.; Hoffman-Censits, J.; et al. Apatorsen plus Docetaxel versus Docetaxel Alone in Platinum-Resistant Metastatic Urothelial Carcinoma (Borealis-2). Br. J. Cancer 2018, 118, 1434–1441. [Google Scholar] [CrossRef] [PubMed]
  123. Lelj-Garolla, B.; Kumano, M.; Beraldi, E.; Nappi, L.; Rocchi, P.; Ionescu, D.N.; Fazli, L.; Zoubeidi, A.; Gleave, M.E. Hsp27 Inhibition with OGX-427 Sensitizes Non-Small Cell Lung Cancer Cells to Erlotinib and Chemotherapy. Mol. Cancer Ther. 2015, 14, 1107–1116. [Google Scholar] [CrossRef]
  124. Li, J.; Zhang, X.; Shen, J.; Guo, J.; Wang, X.; Liu, J. Bortezomib Promotes Apoptosis of Multiple Myeloma Cells by Regulating HSP27. Mol. Med. Rep. 2019, 20, 2410–2418. [Google Scholar] [CrossRef]
  125. Hadchity, E.; Aloy, M.-T.; Paulin, C.; Armandy, E.; Watkin, E.; Rousson, R.; Gleave, M.; Chapet, O.; Rodriguez-Lafrasse, C. Heat Shock Protein 27 as a New Therapeutic Target for Radiation Sensitization of Head and Neck Squamous Cell Carcinoma. Mol. Ther. 2009, 17, 1387–1394. [Google Scholar] [CrossRef]
  126. Nappi, L.; Aguda, A.H.; Nakouzi, N.A.; Lelj-Garolla, B.; Beraldi, E.; Lallous, N.; Thi, M.; Moore, S.; Fazli, L.; Battsogt, D.; et al. Ivermectin Inhibits HSP27 and Potentiates Efficacy of Oncogene Targeting in Tumor Models. J. Clin. Investig. 2020, 130, 699–714. [Google Scholar] [CrossRef]
  127. Xu, N.; Lu, M.; Wang, J.; Li, Y.; Yang, X.; Wei, X.; Si, J.; Han, J.; Yao, X.; Zhang, J.; et al. Ivermectin Induces Apoptosis of Esophageal Squamous Cell Carcinoma via Mitochondrial Pathway. BMC Cancer 2021, 21, 1307. [Google Scholar] [CrossRef]
  128. Chen, L.; Bi, S.; Wei, Q.; Zhao, Z.; Wang, C.; Xie, S. Ivermectin Suppresses Tumour Growth and Metastasis through Degradation of PAK1 in Oesophageal Squamous Cell Carcinoma. J. Cell Mol. Med. 2020, 24, 5387–5401. [Google Scholar] [CrossRef]
  129. Chi, K.N.; Yu, E.Y.; Jacobs, C.; Bazov, J.; Kollmannsberger, C.; Higano, C.S.; Mukherjee, S.D.; Gleave, M.E.; Stewart, P.S.; Hotte, S.J. A Phase I Dose-Escalation Study of Apatorsen (OGX-427), an Antisense Inhibitor Targeting Heat Shock Protein 27 (Hsp27), in Patients with Castration-Resistant Prostate Cancer and Other Advanced Cancers. Ann. Oncol. 2016, 27, 1116–1122. [Google Scholar] [CrossRef]
  130. Shrestha, L.; Bolaender, A.; Patel, H.J.; Taldone, T. Heat Shock Protein (HSP) Drug Discovery and Development: Targeting Heat Shock Proteins in Disease. Curr. Top. Med. Chem. 2016, 16, 2753–2764. [Google Scholar] [CrossRef]
  131. Zhao, K.; Zhou, G.; Liu, Y.; Zhang, J.; Chen, Y.; Liu, L.; Zhang, G. HSP70 Family in Cancer: Signaling Mechanisms and Therapeutic Advances. Biomolecules 2023, 13, 601. [Google Scholar] [CrossRef]
  132. Ding, L.-X.; Zhang, J.; Yang, S.-S.; Wu, J.; Su, T.; Wang, W.-M. Heat Shock Proteins 70 Regulate Cell Motility and Invadopodia-Associated Proteins Expression in Oral Squamous Cell Carcinoma. Front. Endocrinol. 2022, 13, 890218. [Google Scholar] [CrossRef]
  133. Park, S.-H.; Baek, K.-H.; Shin, I.; Shin, I. Subcellular Hsp70 Inhibitors Promote Cancer Cell Death via Different Mechanisms. Cell Chem. Biol. 2018, 25, 1242–1254.e8. [Google Scholar] [CrossRef]
  134. Yang, C.-Y.; Lin, C.-K.; Hsieh, C.-C.; Tsao, C.-H.; Lin, C.-S.; Peng, B.; Chen, Y.-T.; Ting, C.-C.; Chang, W.-C.; Lin, G.-J.; et al. Anti-Oral Cancer Effects of Triptolide by Downregulation of DcR3 In Vitro, In Vivo, and in Preclinical Patient-Derived Tumor Xenograft Model. Head Neck 2019, 41, 1260–1269. [Google Scholar] [CrossRef] [PubMed]
  135. Li, C.; Zhang, B.; Lv, W.; Lai, C.; Chen, Z.; Wang, R.; Long, X.; Feng, X. Triptolide Inhibits Cell Growth and GRP78 Protein Expression but Induces Cell Apoptosis in Original and Radioresistant NPC Cells. Oncotarget 2016, 7, 49588–49596. [Google Scholar] [CrossRef] [PubMed]
  136. Kuo, C.-S.; Yang, C.-Y.; Lin, C.-K.; Lin, G.-J.; Sytwu, H.-K.; Chen, Y.-W. Triptolide Suppresses Oral Cancer Cell PD-L1 Expression in the Interferon-γ-Modulated Microenvironment In Vitro, In Vivo, and in Clinical Patients. Biomed. Pharmacother. 2021, 133, 111057. [Google Scholar] [CrossRef] [PubMed]
  137. Oliveira, A.; Beyer, G.; Chugh, R.; Skube, S.J.; Majumder, K.; Banerjee, S.; Sangwan, V.; Li, L.; Dawra, R.; Subramanian, S.; et al. Triptolide Abrogates Growth of Colon Cancer and Induces Cell Cycle Arrest by Inhibiting Transcriptional Activation of E2F. Lab. Investig. 2015, 95, 648–659. [Google Scholar] [CrossRef]
  138. Jacobson, B.A.; Chen, E.Z.; Tang, S.; Belgum, H.S.; McCauley, J.A.; Evenson, K.A.; Etchison, R.G.; Jay-Dixon, J.; Patel, M.R.; Raza, A.; et al. Triptolide and Its Prodrug Minnelide Suppress Hsp70 and Inhibit In Vivo Growth in a Xenograft Model of Mesothelioma. Genes Cancer 2015, 6, 144–152. [Google Scholar] [CrossRef]
  139. Caicedo-Granados, E.; Lin, R.; Clements-Green, C.; Yueh, B.; Sangwan, V.; Saluja, A. Wild-Type P53 Reactivation by Small-Molecule MinnelideTM in Human Papillomavirus (HPV)-Positive Head and Neck Squamous Cell Carcinoma. Oral Oncol. 2014, 50, 1149–1156. [Google Scholar] [CrossRef]
  140. Roh, J.-L.; Kim, E.H.; Jang, H.; Shin, D. Nrf2 Inhibition Reverses the Resistance of Cisplatin-Resistant Head and Neck Cancer Cells to Artesunate-Induced Ferroptosis. Redox Biol. 2017, 11, 254–262. [Google Scholar] [CrossRef]
  141. Okamoto, H.; Yoshikawa, K.; Shimada, A.; Sano, R.; Inukai, D.; Yamanaka, S.; Suzuki, S.; Ueda, R.; Ueda, H.; Fujimoto, Y.; et al. Artesunate and Cisplatin Synergistically Inhibit HNSCC Cell Growth and Promote Apoptosis with Artesunate-induced Decreases in Rb and Phosphorylated Rb Levels. Oncol. Rep. 2023, 50, 154. [Google Scholar] [CrossRef] [PubMed]
  142. Rousaki, A.; Miyata, Y.; Jinwal, U.K.; Dickey, C.A.; Gestwicki, J.E.; Zuiderweg, E.R.P. Allosteric Drugs: The Interaction of Antitumor Compound MKT-077 with Human Hsp70 Chaperones. J. Mol. Biol. 2011, 411, 614–632. [Google Scholar] [CrossRef] [PubMed]
  143. Miyata, Y.; Li, X.; Lee, H.-F.; Jinwal, U.K.; Srinivasan, S.R.; Seguin, S.P.; Young, Z.T.; Brodsky, J.L.; Dickey, C.A.; Sun, D.; et al. Synthesis and Initial Evaluation of YM-08, a Blood-Brain Barrier Permeable Derivative of the Heat Shock Protein 70 (Hsp70) Inhibitor MKT-077, Which Reduces Tau Levels. ACS Chem. Neurosci. 2013, 4, 930–939. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, J.; Liu, J.; Guo, S.-Y.; Liu, H.-L.; Li, S.-Z. HSP70 Inhibitor Combined with Cisplatin Suppresses the Cervical Cancer Proliferation In Vitro and Transplanted Tumor Growth: An Experimental Study. Asian Pac. J. Trop. Med. 2017, 10, 184–188. [Google Scholar] [CrossRef]
  145. Skorupan, N.; Ahmad, M.I.; Steinberg, S.M.; Trepel, J.B.; Cridebring, D.; Han, H.; Von Hoff, D.D.; Alewine, C. A Phase II Trial of the Super-Enhancer Inhibitor MinnelideTM in Advanced Refractory Adenosquamous Carcinoma of the Pancreas. Future Oncol. 2022, 18, 2475–2481. [Google Scholar] [CrossRef]
  146. McKeon, A.M.; Egan, A.; Chandanshive, J.; McMahon, H.; Griffith, D.M. Novel Improved Synthesis of HSP70 Inhibitor, Pifithrin-μ. In Vitro Synergy Quantification of Pifithrin-μ Combined with Pt Drugs in Prostate and Colorectal Cancer Cells. Molecules 2016, 21, 949. [Google Scholar] [CrossRef]
  147. Lim, S.H.; Saluja, A.; Vickers, S.; Hong, J.Y.; Kim, S.T.; Lavania, S.; Pandey, S.; Gupta, V.K.; Velagapudi, M.R.; Lee, J. The Safety and Efficacy Outcomes of Minnelide given Alone or in Combination with Paclitaxel in Advanced Gastric Cancer: A Phase I Trial. Cancer Lett. 2024, 597, 217041. [Google Scholar] [CrossRef]
  148. Borazanci, E.; Saluja, A.; Gockerman, J.; Velagapudi, M.; Korn, R.; Von Hoff, D.; Greeno, E. First-in-Human Phase I Study of Minnelide in Patients With Advanced Gastrointestinal Cancers: Safety, Pharmacokinetics, Pharmacodynamics, and Antitumor Activity. Oncologist 2024, 29, 132–141. [Google Scholar] [CrossRef]
  149. Magyar, C.T.J.; Vashist, Y.K.; Stroka, D.; Kim-Fuchs, C.; Berger, M.D.; Banz, V.M. Heat Shock Protein 90 (HSP90) Inhibitors in Gastrointestinal Cancer: Where Do We Currently Stand?-A Systematic Review. J. Cancer Res. Clin. Oncol. 2023, 149, 8039–8050. [Google Scholar] [CrossRef]
  150. Basset, C.A.; Conway de Macario, E.; Leone, L.G.; Macario, A.J.L.; Leone, A. The Chaperone System in Cancer Therapies: Hsp90. J. Mol. Histol. 2023, 54, 105–118. [Google Scholar] [CrossRef]
  151. Misso, G.; Giuberti, G.; Lombardi, A.; Grimaldi, A.; Ricciardiello, F.; Giordano, A.; Tagliaferri, P.; Abbruzzese, A.; Caraglia, M. Pharmacological Inhibition of HSP90 and Ras Activity as a New Strategy in the Treatment of HNSCC. J. Cell Physiol. 2013, 228, 130–141. [Google Scholar] [CrossRef] [PubMed]
  152. Cohen, S.M.; Mukerji, R.; Samadi, A.K.; Zhao, H.; Blagg, B.S.J.; Cohen, M.S. Novel C-Terminal Hsp90 Inhibitor for Head and Neck Squamous Cell Cancer (HNSCC) with In Vivo Efficacy and Improved Toxicity Profiles Compared with Standard Agents. Ann. Surg. Oncol. 2012, 19, S483–S490. [Google Scholar] [CrossRef] [PubMed]
  153. Shintani, S.; Zhang, T.; Aslam, A.; Sebastian, K.; Yoshimura, T.; Hamakawa, H. P53-Dependent Radiosensitizing Effects of Hsp90 Inhibitor 17-Allylamino-17-Demethoxygeldanamycin on Human Oral Squamous Cell Carcinoma Cell Lines. Int. J. Oncol. 2006, 29, 1111–1117. [Google Scholar] [CrossRef][Green Version]
  154. Ui, T.; Morishima, K.; Saito, S.; Sakuma, Y.; Fujii, H.; Hosoya, Y.; Ishikawa, S.; Aburatani, H.; Fukayama, M.; Niki, T.; et al. The HSP90 Inhibitor 17-N-Allylamino-17-Demethoxy Geldanamycin (17-AAG) Synergizes with Cisplatin and Induces Apoptosis in Cisplatin-Resistant Esophageal Squamous Cell Carcinoma Cell Lines via the Akt/XIAP Pathway. Oncol. Rep. 2014, 31, 619–624. [Google Scholar] [CrossRef]
  155. Alexiou, G.A.; Goussia, A.; Voulgaris, S.; Fotopoulos, A.D.; Fotakopoulos, G.; Ntoulia, A.; Zikou, A.; Tsekeris, P.; Argyropoulou, M.I.; Kyritsis, A.P. Prognostic Significance of MRP5 Immunohistochemical Expression in Glioblastoma. Cancer Chemother. Pharmacol. 2012, 69, 1387–1391. [Google Scholar] [CrossRef]
  156. Shin, M.K.; Jeong, K.-H.; Choi, H.; Ahn, H.-J.; Lee, M.-H. Heat Shock Protein 90 Inhibitor Enhances Apoptosis by Inhibiting the AKT Pathway in Thermal-Stimulated SK-MEL-2 Human Melanoma Cell Line. J. Dermatol. Sci. 2018, 90, 357–360. [Google Scholar] [CrossRef]
  157. Wu, X.; Marmarelis, M.E.; Hodi, F.S. Activity of the Heat Shock Protein 90 Inhibitor Ganetespib in Melanoma. PLoS ONE 2013, 8, e56134. [Google Scholar] [CrossRef][Green Version]
  158. Naz, S.; Leiker, A.J.; Choudhuri, R.; Preston, O.; Sowers, A.L.; Gohain, S.; Gamson, J.; Mathias, A.; Waes, C.V.; Cook, J.A.; et al. Pharmacological Inhibition of HSP90 Radiosensitizes Head and Neck Squamous Cell Carcinoma Xenograft by Inhibition of DNA Damage Repair, Nucleotide Metabolism, and Radiation-Induced Tumor Vasculogenesis. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 1295–1305. [Google Scholar] [CrossRef]
  159. Mehta, R.K.; Pal, S.; Kondapi, K.; Sitto, M.; Dewar, C.; Devasia, T.; Schipper, M.J.; Thomas, D.G.; Basrur, V.; Pai, M.P.; et al. Low-Dose Hsp90 Inhibitor Selectively Radiosensitizes HNSCC and Pancreatic Xenografts. Clin. Cancer Res. 2020, 26, 5246–5257. [Google Scholar] [CrossRef]
  160. Spiegelberg, D.; Dascalu, A.; Mortensen, A.C.; Abramenkovs, A.; Kuku, G.; Nestor, M.; Stenerlöw, B. The Novel HSP90 Inhibitor AT13387 Potentiates Radiation Effects in Squamous Cell Carcinoma and Adenocarcinoma Cells. Oncotarget 2015, 6, 35652–35666. [Google Scholar] [CrossRef]
  161. McLaughlin, M.; Barker, H.E.; Khan, A.A.; Pedersen, M.; Dillon, M.; Mansfield, D.C.; Patel, R.; Kyula, J.N.; Bhide, S.A.; Newbold, K.L.; et al. HSP90 Inhibition Sensitizes Head and Neck Cancer to Platin-Based Chemoradiotherapy by Modulation of the DNA Damage Response Resulting in Chromosomal Fragmentation. BMC Cancer 2017, 17, 86. [Google Scholar] [CrossRef]
  162. Yin, X.; Zhang, H.; Lundgren, K.; Wilson, L.; Burrows, F.; Shores, C.G. BIIB021, a Novel Hsp90 Inhibitor, Sensitizes Head and Neck Squamous Cell Carcinoma to Radiotherapy. Int. J. Cancer 2010, 126, 1216–1225. [Google Scholar] [CrossRef] [PubMed]
  163. Friedman, J.A.; Wise, S.C.; Hu, M.; Gouveia, C.; Broek, R.V.; Freudlsperger, C.; Kannabiran, V.R.; Arun, P.; Mitchell, J.B.; Chen, Z.; et al. HSP90 Inhibitor SNX5422/2112 Targets the Dysregulated Signal and Transcription Factor Network and Malignant Phenotype of Head and Neck Squamous Cell Carcinoma. Transl. Oncol. 2013, 6, 429-IN5. [Google Scholar] [CrossRef] [PubMed]
  164. Mielczarek-Lewandowska, A.; Hartman, M.L.; Czyz, M. Inhibitors of HSP90 in Melanoma. Apoptosis 2020, 25, 12–28. [Google Scholar] [CrossRef] [PubMed]
  165. Solárová, Z.; Mojžiš, J.; Solár, P. Hsp90 Inhibitor as a Sensitizer of Cancer Cells to Different Therapies (Review). Int. J. Oncol. 2015, 46, 907–926. [Google Scholar] [CrossRef][Green Version]
  166. Moon, H.-J.; Park, S.-Y.; Lee, S.-H.; Kang, C.-D.; Kim, S.-H. Nonsteroidal Anti-Inflammatory Drugs Sensitize CD44-Overexpressing Cancer Cells to Hsp90 Inhibitor Through Autophagy Activation. Oncol. Res. 2019, 27, 835–847. [Google Scholar] [CrossRef]
  167. Lee, Y.-C.; Chang, W.-W.; Chen, Y.-Y.; Tsai, Y.-H.; Chou, Y.-H.; Tseng, H.-C.; Chen, H.-L.; Wu, C.-C.; Chang-Chien, J.; Lee, H.-T.; et al. Hsp90α Mediates BMI1 Expression in Breast Cancer Stem/Progenitor Cells through Facilitating Nuclear Translocation of c-Myc and EZH2. Int. J. Mol. Sci. 2017, 18, 1986. [Google Scholar] [CrossRef]
  168. Pedersen, K.S.; Kim, G.P.; Foster, N.R.; Wang-Gillam, A.; Erlichman, C.; McWilliams, R.R. Phase II Trial of Gemcitabine and Tanespimycin (17AAG) in Metastatic Pancreatic Cancer: A Mayo Clinic Phase II Consortium Study. Investig. New Drugs 2015, 33, 963–968. [Google Scholar] [CrossRef]
  169. Wagner, A.J.; Chugh, R.; Rosen, L.S.; Morgan, J.A.; George, S.; Gordon, M.; Dunbar, J.; Normant, E.; Grayzel, D.; Demetri, G.D. A Phase I Study of the HSP90 Inhibitor Retaspimycin Hydrochloride (IPI-504) in Patients with Gastrointestinal Stromal Tumors or Soft-Tissue Sarcomas. Clin. Cancer Res. 2013, 19, 6020–6029. [Google Scholar] [CrossRef]
  170. Shah, S.; Luke, J.J.; Jacene, H.A.; Chen, T.; Giobbie-Hurder, A.; Ibrahim, N.; Buchbinder, E.L.; McDermott, D.F.; Flaherty, K.T.; Sullivan, R.J.; et al. Results from Phase II Trial of HSP90 Inhibitor, STA-9090 (Ganetespib), in Metastatic Uveal Melanoma. Melanoma Res. 2018, 28, 605–610. [Google Scholar] [CrossRef]
  171. Eroglu, Z.; Chen, Y.A.; Gibney, G.T.; Weber, J.S.; Kudchadkar, R.R.; Khushalani, N.I.; Markowitz, J.; Brohl, A.S.; Tetteh, L.F.; Ramadan, H.; et al. Combined BRAF and HSP90 Inhibition in Patients with Unresectable BRAF V600E-Mutant Melanoma. Clin. Cancer Res. 2018, 24, 5516–5524. [Google Scholar] [CrossRef]
  172. Hoy, S.M. Pimitespib: First Approval. Drugs 2022, 82, 1413–1418. [Google Scholar] [CrossRef]
Figure 1. The cellular processes mediated by HSP27, HSP70, and HSP90 proteins in carcinoma. (A) Inhibition of apoptosis: HSP27 inhibits apoptosis by interacting with AKT, activating NF-κβ, blocking Bax, cytochrome c, caspase-3, caspase-9, and suppressing FAS, TNF, and TRAIL receptors. HSP70 blocks apoptosis by the activation of the STAT3, BCL-2, and PI3K/AKT/mTOR pathways and suppression of the p38/MAPK pathway, Bax, AIF-1, ASK1, APAF-1 proteins, caspase-3, caspase-9, and Apo-2L/TRAIL. HSP90 inhibits apoptosis by the activation of the PI3K/AKT/mTOR pathway, NF-қβ, survivin, BCL-2 and the suppression of cytochrome c, Bax, XIAP, p53, caspase-3, caspase-8, FAS, and TNF receptors. (B) Epithelial–mesenchymal transition (EMT): HSP27 mediates EMT by the activation of EGF/β-catenin, β-catenin/MMP-3, β-catenin/Slug, NF-κβ, AKT, STAT3, and Twist and the decrease in E-cadherin expression. HSP70 regulates EMT by the activation of MAPK, MMP-2, pSTAT3, N-cadherin, vimentin and downregulation of E-cadherin. HSP90 controls EMT by the activation of the p38/MAPK and PI3K/AKT/mTOR signaling pathways, TNFα, NF-қβ, vimentin, and the inhibition of WNT/β-catenin pathways, and E-cadherin expression. (C) Tumor progression/metastasis: HSP27 promotes tumor progression and the formation of metastasis by the activation of NF-қβ, c-Myc, MMP-2, and MMP-9 and the downregulation of the p53 protein. HSP70 influences tumor progression and formation of metastasis by the activation of RHOA, WNT/β-catenin pathway, STAT3, and MMP-2 and the downregulation of p53 expression. HSP90 effects tumor progression and formation of metastasis by the activation of the RAS/RAF/ERK, PI3K/AKT/mTOR, and MAPK signaling pathways, MMP-2, MMP-9, and telomerase upregulation. (D) Angiogenesis: HSP27 increases angiogenesis by the upregulation of VEGFR2, IL-8, NF-қβ, and HIF-1α. HSP70 promotes angiogenesis by the upregulation of HIF-1α/VEGF activity and IL-5 secretion. HSP90 increases angiogenesis by the activation of HIF-1α, NF-қβ, AKT, VEGF/VEGFR signaling pathway and the upregulation of FLT3, IL-8, and EGFR2.
Figure 1. The cellular processes mediated by HSP27, HSP70, and HSP90 proteins in carcinoma. (A) Inhibition of apoptosis: HSP27 inhibits apoptosis by interacting with AKT, activating NF-κβ, blocking Bax, cytochrome c, caspase-3, caspase-9, and suppressing FAS, TNF, and TRAIL receptors. HSP70 blocks apoptosis by the activation of the STAT3, BCL-2, and PI3K/AKT/mTOR pathways and suppression of the p38/MAPK pathway, Bax, AIF-1, ASK1, APAF-1 proteins, caspase-3, caspase-9, and Apo-2L/TRAIL. HSP90 inhibits apoptosis by the activation of the PI3K/AKT/mTOR pathway, NF-қβ, survivin, BCL-2 and the suppression of cytochrome c, Bax, XIAP, p53, caspase-3, caspase-8, FAS, and TNF receptors. (B) Epithelial–mesenchymal transition (EMT): HSP27 mediates EMT by the activation of EGF/β-catenin, β-catenin/MMP-3, β-catenin/Slug, NF-κβ, AKT, STAT3, and Twist and the decrease in E-cadherin expression. HSP70 regulates EMT by the activation of MAPK, MMP-2, pSTAT3, N-cadherin, vimentin and downregulation of E-cadherin. HSP90 controls EMT by the activation of the p38/MAPK and PI3K/AKT/mTOR signaling pathways, TNFα, NF-қβ, vimentin, and the inhibition of WNT/β-catenin pathways, and E-cadherin expression. (C) Tumor progression/metastasis: HSP27 promotes tumor progression and the formation of metastasis by the activation of NF-қβ, c-Myc, MMP-2, and MMP-9 and the downregulation of the p53 protein. HSP70 influences tumor progression and formation of metastasis by the activation of RHOA, WNT/β-catenin pathway, STAT3, and MMP-2 and the downregulation of p53 expression. HSP90 effects tumor progression and formation of metastasis by the activation of the RAS/RAF/ERK, PI3K/AKT/mTOR, and MAPK signaling pathways, MMP-2, MMP-9, and telomerase upregulation. (D) Angiogenesis: HSP27 increases angiogenesis by the upregulation of VEGFR2, IL-8, NF-қβ, and HIF-1α. HSP70 promotes angiogenesis by the upregulation of HIF-1α/VEGF activity and IL-5 secretion. HSP90 increases angiogenesis by the activation of HIF-1α, NF-қβ, AKT, VEGF/VEGFR signaling pathway and the upregulation of FLT3, IL-8, and EGFR2.
Cells 14 01897 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cierpikowski, P.; Bar, J. Heat Shock Proteins in Head and Neck Squamous Cell Carcinoma. Cells 2025, 14, 1897. https://doi.org/10.3390/cells14231897

AMA Style

Cierpikowski P, Bar J. Heat Shock Proteins in Head and Neck Squamous Cell Carcinoma. Cells. 2025; 14(23):1897. https://doi.org/10.3390/cells14231897

Chicago/Turabian Style

Cierpikowski, Piotr, and Julia Bar. 2025. "Heat Shock Proteins in Head and Neck Squamous Cell Carcinoma" Cells 14, no. 23: 1897. https://doi.org/10.3390/cells14231897

APA Style

Cierpikowski, P., & Bar, J. (2025). Heat Shock Proteins in Head and Neck Squamous Cell Carcinoma. Cells, 14(23), 1897. https://doi.org/10.3390/cells14231897

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop