IMR Press / FBL / Volume 28 / Issue 3 / DOI: 10.31083/j.fbl2803056
Open Access Review
Heat Shock Proteins in Non-Small-Cell Lung Cancer—Functional Mechanism
Guanyu Zhou1,2,†Yi Pu1,3,†Kejia Zhao1,2Yaohui Chen1,2,*Gao Zhang4,*
Show Less
1 Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
2 Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, 610041 Chengdu, Sichuan, China
3 Department of Burn and Reconstructive Surgery, Laboratory of Mitochondria and Metabolism, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan, China
4 Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, 999077 Hong Kong, China
*Correspondence: mcb.chenyaohui@gmail.com (Yaohui Chen); gzhang6@me.com (Gao Zhang)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(3), 56; https://doi.org/10.31083/j.fbl2803056
Submitted: 31 December 2022 | Revised: 3 February 2023 | Accepted: 10 February 2023 | Published: 20 March 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Non-small cell lung cancer (NSCLC) accounts for 80–85% of all lung cancers, which has the highest cancer-related mortality worldwide. Regardless of the therapeutic effects of chemotherapy or targeted therapy, drug resistance will occur after 1 year. Heat shock proteins (HSPs) are a class of molecular chaperones participated in protein stability and multiple intracellular signaling pathways. It has been widely reported that HSPs family is over expressed in non-small cell lung cancer, and these molecules are also associated with protein stability and multiple intracellular signaling pathways. The effect of chemotherapy drugs or targeted drugs on cancer cells is usually to induce apoptosis. It is necessary to explore the interaction between heat shock protein family and apoptosis pathway in NSCLC. Here we provide a brief review of how HSPs affect the apoptotic pathway in NSCLC.

Keywords
Heat shock proteins
non-small cell lung cancer
apoptotic
1. Introduction

Lung cancer is currently the most cancer-related mortality cause worldwide. In China, the incidence of lung cancer increased yearly of which, non-small cell lung cancer (NSCLC) accounted for 80%–85% of all lung cancers [1, 2, 3]. Classical treatments for advanced NSCLC are chemotherapy and targeted therapy (for tumors harboring activating mutations in cancer driving genes). NSCLC responds well at the beginning, but develops drug resistance after 1–2 years [4, 5, 6, 7]. Therefore, studying the mechanism of the occurrence and development of NSCLC is very important for the development of new adjuvant therapy.

Heat shock proteins (HSPs) are a class of highly conserved molecular chaperones [8]. The presence of HSPs can prevent protein misfolding and over-aggregation by regulating protein assembly, folding and translocation in cells [9, 10]. The expression of most HSPs maintains at low levels under normal conditions but will be significantly upregulated in response to stress or in cancer cells [11, 12, 13, 14]. HSPs are classified as HSP100, HSP90, HSP70, HSP60, and small HSPs (those whose molecular weight under 60 kDa) according to their molecular weights [15]. Several studies showed increased expression of HSPs in a variety of solid tumors, including NSCLC [16, 17, 18, 19, 20]. Multiple articles suggested that HSPs can participate in the apoptotic pathway in vitro and HSP inhibitors enhanced therapeutic efficacy in NSCLC preclinical mouse models treated with chemotherapy or targeted therapy [21, 22, 23, 24, 25], indicating that HSPs may play an important role in the occurrence, development and treatment of NSCLC.

Here, we review current researches of HSPs in NSCLC, emphasizing HSP expression in NSCLC tissues and possible mechanisms for drug resistance.

2. HSP90

HSP90 is a class of HSPs with a molecular weight of 90 kDa [26]. Human HSP90 includes four isoforms: HSP90α and β, Trap1, and Grp94. The first twos localize in the cytosol, Trap1 localizes in mitochondria, and Grp94 localizes in the endoplasmic reticulum [27]. HSP90 has three structural domains: N-terminal domain that binds to ATP, C-terminal domain for dimerization, and middle domain that recognizes and binds to target proteins. HSP90 transforms as a dimer in the cell and hydrolyzes ATP to function [28, 29].

2.1 Expression

Recently, it has been found that HSP90 is highly expressed in NSCLC, positively correlated with age and smoking status, negatively correlated with the 5-year survival rate [30]. In a recent study, serum HSP90α level was found significantly higher in NSCLC patients (n = 312) than in healthy controls (n = 160) (106.9 ± 66.73 ng/mL vs. 45.9 ± 12.1 ng/mL, respectively). Also, serum HSP90α increased before clinical recurrence in some patients who received both chemoradiotherapy and surgery in the same study [31]. Another study showed that high HSP90β expression often associated with shorter overall survival, indicating HSP90β alone could be used as a prognostic factor in NSCLC [32, 33].

2.2 HSP90 for Apoptosis

Multiple studies showed HSP90 participates in apoptotic pathways, especially in caspase pathways. For TRAP1 knockdown NSCLC cells, reactive oxygen species (ROS) production is significantly increased when treated with cisplatin, activating mitochondrial apoptotic pathway. Besides, the pro-apoptotic effect of cisplatin is significantly enhanced in TRAP1 knockdown NSCLC cells [34]. The Grp94 inhibitor PU-H71 induces NSCLC cells apoptosis through the mitochondrial apoptosis pathway by down regulating Bcl-2 and up-regulating Bax, resulting in cytochrome C release [35]. In NSCLC cell lines, HSP90 knockdown causes more cells to enter G2/M phase, inhibiting cell growth, increasing sensitivity to gefitinib, inducing apoptosis [36]. In both mouse NSCLC syngeneic and xenograft models, knockdown of HSP90 followed by treatment with epidermal growth factor receptor (EGFR) inhibitor resulted in enhanced antitumor activity. Meanwhile, immunohistochemistry showed that the combination of HSP90 inhibitor and EGFR inhibitor induced more tumor cell apoptosis in both on-treatment and resistant specimens [37]. In conclusion, HSP90 inhibition activates caspase pathway, leading to NSCLC apoptosis both in vitro and in vivo (Fig. 1).

Fig. 1.

The role of different heat shock protein family members in the apoptotic pathway. HSPs participate in anti-apoptosis mainly in two ways: (1) HSPs can block signal transmissions by competitively binding to apoptotic signaling molecules. (2) HSPs can directly bind to the apoptotic effector molecules to prevent apoptosis.

2.3 Other Mechanisms

NSCLC cells often obtain overexpression or activating mutation of proto-oncogenes, such as human epidermal growth factor receptor (HER2), serine/threonine kinase (Raf-1), anaplastic lymphoma kinase (ALK), protein kinase B (PKB or AKT). HSP90 is thought to stabilize above protooncogenes in NSCLC cells to support cancer initiation and progression [38, 39]. Using HSP90 inhibitor 17-AAG can downregulate EGFR, AKT through decreasing cellular thymidine phosphorylase via ubiquitin-26S proteasome pathway to inhibit tumor cells growth. Furthermore, combination of 17-AAG can increase the killing effect of cisplatin on NSCLC tumors [21, 22, 23]. Another HSP90 inhibitor, AUY922 can inhibit KRAS signaling and slow down NSCLC cell growth [40]. In KRAS mutant NSCLC cell lines, HSP90 confers resistance to chemotherapeutic agents and targeted therapy agents by stabilizing AXL/Elf4e [41]. HSP90 can also activate NF-κB pathway by stabilizing receptor-Interacting protein kinase (RIP) and IkappaB kinase beta (IKKβ) to promote tumor cells proliferation [42].

Abnormal up-regulation of HSP90 was found in chemotherapy-resistant NSCLC cells. In these chemotherapy-resistant cells, by inhibiting HSP90, the expression of Anti-Müllerian hormone (AMH) and Anti-Müllerian hormone type II receptor (AMHR2) increased, the epithelial–mesenchymal transition (EMT) of NSCLC cells was inhibited, and tumor cell sensitivity to cisplatin restored [43] (Fig. 2).

Fig. 2.

The role of Hsp90 in regulating NSCLC oncogenesis. (A) HSP90 can promote NSCLC oncogenesis by stabilizing mutated HER2, RAF-1, ALK and PKB products, or inhibiting the ubiquitin-proteasome pathway of EGFR and AKT. (B) HSP90 can stabilize RIPK and IKKβ to promote NF-κB signaling.

3. HSP70

The HSP70 family has numerous members, encoded by a multigene family, and is widely distributed in cells, for example, Grp75 is found in mitochondria and Grp78 is in the endoplasmic reticulum [44, 45]. Typically, HSPs1L, HSPs2, HSPs5, HSPs8, HSPs9, HSPs12A, HSPs12B and HSPs13 encode HSP70 proteins that are constitutively expressed in different types of cells, while other HSP70 proteins are expressed in response to cellular stress, such as those encoded by HSPs1A, HSPs1B, HSPs6, HSPs7, and HSPs14 [46]. Although there are many family members and the gene loci and amino acid sequences are different, HSP70s share a similar structure. They have a conserved ATP-binding region at the N-terminus that binds and hydrolyzes ATP [47]. The C-terminus is usually the site where HSP70s bind to target proteins, acting as chaperones. These two functional domains are linked by a highly conserved leucine-rich amino acid sequence [48, 49]. HSP70 itself has weak ATPase activity and usually needs to cochaperone with J-protein (HSP40) to enhance its ATPase activity for function [50].

3.1 Expression

HSP70 is overexpressed in NSCLC, and sufficient evidence suggests that the overexpression of HSP70 is associated with poor prognosis. Pfister et al. [51] found that HSP70 was expressed on the cell membrane in about 40% of 150 NSCLC tumor samples, revealing that HSP70 may be overexpressed and localized on the membrane in NSCLC patients. The result of a large cohort study in Japan showed that the mean plasma HSP70 level of 189 NSCLC patients was 2.41 ng/mL, which was higher than the mean level of 2.01 ng/mL in healthy cohort [52]. Vostakolaei et al. [53] also detected positive HSP70 in tumor cells from NSCLC patients, and high levels of HSP70 was accompanied with poor prognosis.

3.2 HSP70 for Apoptosis

HSP70 can also, like other chaperones, enable cancer cells to escape stress for further progression by inhibiting cell apoptosis. HSP70 can regulate both intracellular and extracellular apoptotic pathways at the same time. In the intracellular apoptotic pathway, HSP70 can prevent the combination of Apoptotic Peptidase Activating Factor 1 (APAF-1) to pro-caspase9 by directly binding to the caspase recruitment domain of APAF-1, thereby affecting the formation of apoptotic complexes in the mitochondrial [54]. HSP70 can regulate the level of Apoptosis signal-regulating kinase 1 (ASK1) protein by binding to CHIP protein in TNF-α-induced apoptosis model, inhibiting c-Jun N-terminal kinase (JNK) and p38. Alterations in JNK protein levels resulted in the release of cytochrome C from mitochondria and ultimately inhibited the apoptotic pathway [55, 56]. In addition, HSP70 can bind to TNF-mediated apoptosis-inducing ligand receptor 1/2 (TRAIL-R1/R2) in the extracellular apoptosis pathway, blocking the formation of the Death-inducing signaling complex (DISC), affecting the apoptotic function of caspase-8, a downstream of this pathway [57].

Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) can induce phosphorylation of 41 Y residues of HSP70, ubiquitinate and degrade HSP70, increasing the probability of gene mutation, and eventually induce EGFR T790M mutation, leading to drug resistance [58]. Continuous cisplatin stimulation in A549 cells induced cisplatin resistance, and increased expression of HSP70 was detected in the drug-resistant A549 cells after cisplatin stimulation. When stimulating the cisplatin-resistant cell line again with cisplatin, the level of HSP70 was down-regulated, and the apoptotic signal staining was increased by flow cytometry [59]. Studies on NSCLC A549 and H460 cell lines showed that A549 cell lines with high GRP78 expression were less sensitive to cisplatin [60] (Fig. 1).

3.3 Other Mechanisms

AIMP2-DX2 is a protein mutant that causes lung cancer. Semi Lim et al. [61] found that AIMP2-DX2 mutant can avoid Siah1-mediated ubiquitination by binding to HSP70 C-terminus, AIMP2-DX2 will eventually be stabilized in cells and cause NSCLC [62].

Recent studies have shown that OTUD3, a deubiquitinating enzyme, promotes NSCLC cell growth and metastasis by stabilizing GRP78 through binding it and co-localizating to the endoplasmic reticulum. It is evident that GRP78 may play an important role in the metastasis of cancer cells [63].

It has been reported that HSP70 modified by Glyoxalase I can activate TGF-β signaling pathway, up-regulate Smad7 and down-regulate Smad4, and finally induce EMT of NSCLC epithelial cells [64]. Under hypoxic conditions, GRP78 in A549 cell line was significantly increased, and GRP78 silence was found to inhibit the expression of Smad2/3 along, and the cell EMT transformation markers were also reduced [65] (Fig. 3).

Fig. 3.

HSP70 can stabilize AIMP2-DX2 to promote tumorigenesis, and participate in the regulation of SMAD2/3/4/7 to induce tumor EMT and further progression and metastasis.

4. HSP27

The expression of HSP27, also known as HSPB1, remains at low level under physiological condition and is induced by stress. The transcriptional regulation of HSP27 is affected by cell type and cellular environmental factors, such as heat shock factor 1 (HSF-1) and HIF1α [66]. The structure of HSP27 only contains a N-terminus and a C-terminus. There is no ATP-binding domain in HSP27 structure for its function does not depend on energy from ATP hydrolysis. Activation of HSP27 is through its own phosphorylation or methylglyoxal (MG) modification. HSP27 has four common phosphorylation sites, namely threonine at position 15, 78, 82, and 86. After phosphorylation, HSP27 will dimerize and function [67, 68]. The MG modification of HSP27 is associated with its oligomerization, and it occurs at arginine at position 188 in the C-terminus of HSP27 [69]. The MG modification of HSP27 is an important epigenetic modification for its physiological function and is essential for its inhibition of caspase-9 activity [70, 71].

4.1 Expression

A study using Immunohistochemistry (IHC) stain tested 76 NSCLC tumor samples, the expression of HSP27 was significantly higher in lung cancer than that in normal tissues, proportional to malignancy. Meanwhile, several studies have shown that patients with higher HSP27 expression have a significantly shorter 5-year survival than those with lower HSP27 expression [72, 73, 74, 75]. What’s more, in a multi-institution study, serum phosphorylated HSP27 (S78/S82) was detected in 109 NSCLC patients, and it was found that the level of serum phosphorylated HSP27 (S78/S82) was higher in advanced NSCLC patients than in early-stage NSCLC patients [76].

4.2 HSP70 for Apoptosis

A report in 2016 showed that HSP27 and caspase-9 colocalize in focal adhesions of lung cancer tissue, and the traditional Chinese medicine YangZheng XiaoJi can block this co-localization by inhibiting the phosphorylation of HSP27 protein S86, making insensitive lung cancer cell lines sensitive to chemotherapeutic agents [24]. Jeroen W J van Heijst and his colleagues [25] showed that in lung squamous cell carcinoma cells, the expression of HSP27 in tumor cells increased after incubation with cisplatin, and the increased HSP27 binded to caspase-3 to inhibit the activation of apoptotic pathway and increased tumor cells’ resistance to cisplatin. The apoptosis induced by TNF-related apoptosis-inducing ligand (TRAIL) was initiated by TRAIL binding to TRAIL-R1/R2. This interaction leaded to cleavage and activation of pro-caspase-8 by the adaptor protein FADD, however this effect is inhibited in A549 cells overexpressing HSP27 [77, 78, 79]. In NSCLC cells with high expression of HSP27 after radiotherapy, inhibition of HSP27 increased the expression of intracellular cytochrome C. After downregulate HSP27 using short interference RNA, it was found that the expression of Bax was up-regulated, the expression of Bcl2 was down-regulated, and pro-caspase-8 was activated to induce cell apoptosis, increasing the sensitivity of NSCLC to radiotherapy [80]. In addition, HSP27 can prevent the formation of apoptotic bodies in NSCLC cells by directly binding to cytochrome C and preventing the release of cytochrome C [81].

HSP27 can reduce the oxidative stress of cells and prevent apoptosis by reducing ROS and increasing reduced glutathione [82]. HSP27 is also able to resist apoptosis by participating in the regulation of the threonine kinase PKB, mimicking a positive survival signal [83]. Phosphorylated HSP27 may directly bind to other pro-apoptotic proteins such as death domain-associated protein (DAXX) to inhibit the apoptotic pathway. For example, it interacted with DAXX to affect the membrane Fas-mediated DAXX-ASK1-JNK apoptotic pathway [84, 85] (Fig. 1).

4.3 Other Mechanisms

Zhicheng Huang’s study [86] found that HSP27 expression was increased in A549 cells with TGF-β signaling pathway activation mutation, and down-regulation of SMAD3 could reverse this phenomenon, resulting in significant inhibition of tumor cell growth. In gefitinib-resistant NSCLC cells (harboring EGFR T790M mutation), the expression of phosphorylated-p38 (p-p38) and phosphorylated-MK2 (p-MK2) was increased after gefitinib stimulation, and p-MK2 has been proved to phosphorylate HSP27 (p-HSP27). p-HSP27 (Ser15, Ser78 and Ser82) can directly interact with phosphorylated-AKT (p-AKT), and inhibition of HSP27 phosphorylation or expression decreased the stability of p-AKT and the stability of EGFR. Therefore, gefitinib-resistant NSCLC with AKT activation will regain sensitivity to gefitinib [87, 88]. The study by Guang-Yin Zhao [89] showed that knockdown of HSP27 in NSCLC cell lines expressing 14-3-3ζ protein attenuated NSCLC cell migration. In a NSCLC mouse model, HSP27 has been shown to promote lung cancer development through the activator protein-1 (AP-1) pathway, however the mechanism remains to be elucidated [90] (Fig. 4A).

Fig. 4.

The role of Hsp27 and Hsp60 in regulating NSCLC metastasis and migration. (A) Phosphorylated HSP27 can bind to phosphorylated Akt to prevent its degradation through ubiquitin-proteasome pathway. (B) HSP60 can be sialylated on cell membrane after being glycosylated. Then HSP60 can bind to MAL-1 to promote tumor metastasis.

5. HSP60

HSP60 is an important member in heat shock family with a molecular weight of 60 kDa and its structure is highly conserved in a variety of organisms [91]. HSP60 is located in mitochondria under physiological conditions and ensures proper folding, assembly and repairment of mitochondria-associated proteins [92]. However, in tumor cells, HSP60 can be glycosylated, translocated to the cytoplasm or even on the cell membrane, affecting tumor cell migration [93, 94].

5.1 Expression

Several studies have shown that HSP60 significantly increased in tumor tissues of lung cancer patients compared with healthy control tissues, and is negatively correlated with prognosis, with no significant relationship with Tumor-Node-Metastasis (TNM) classification [95, 96]. It has also been shown that cancer cells located at the center of tumor tissue expressed higher levels of HSP60 in the cytoplasm than those located at the edge of tumor tissue at different invasive sites of tumor cells [97].

5.2 HSP60 for Apoptosis

HSP60 can bind to and stablize the inactive form of caspase-3, thereby exerting its anti-apoptotic effect in lung cancer. In fact, inhibition of the combination did significantly increase the apoptosis of lung cancer cells [98]. It has also been shown that HSP60 on mitochondria can combine with Bax-Bak-Bcl-XL to form macromolecular complexes and inhibit apoptosis intracellularly [99]. To manipulate the combining of HSP60 and Bax, cells were treated with antisense RNA. With the decrease of HSP60, an increase of unbound Bax and Bak in the cytosol was observed. The presence of HSP60 may be necessary for Bax translocation from the cytosol to the mitochondria, upon which subsequent cytochrome c release and caspase activation can occur in an orderly manner [99, 100]. 6-Shogaol (6-SH) is an anti-cancer active substance found in dried ginger. One study has shown that 6-SH can promote the proteasome-mediated degradation of HSP60, and can induce the reduction of Bcl-2 and survivin expression and the increase of pro-apoptotic protein Bax expression in A549 cells. Then, these 6-SH-treated A549 cells were arrested at G0/G1 phase and were induced to cell apoptosis [101] (Fig. 1).

5.3 Other Mechanisms

After N-glycosylation, HSP60 in tumor cells can translocate to tumor cell membrane [102]. HSP60 on the cell membrane can be further α2,3-sialylated, interacting with Maackia amurensis leukoagglutinin I (MAL-I) to promote the migration of NSCLC cells [103].

HSP60 in tumor cells can form a complex with P53, resulting in the reduction of the binding of P53 to molecules related to cell cycle arrest signals, thus making tumor cells to enter a highly active replication state. It has been shown that doxorubicin can attenuate cancer proliferation through acetylation of HSP60, reducing its interaction with P53. Some studies have also shown that P53 is closely related to cell apoptosis, so whether the combination of HSP60 and P53 has a role in this aspect remains to be elucidated by further studies [104, 105, 106] (Fig. 4B). Cancer cells with HSP60 knockdown by HSP60 shRNA showed reduced basal respiration and impaired ATP synthesizing capacity through oxidative phosphorylation (OXPHOS), and accompanied by reduced cancer cell proliferation [107].

6. Summary and Perspectives

HSP is a huge family with lots of family members and complicated functions. In general, HSPs are upregulated in NSCLC, regulating cancer development and drug resistance through apoptotic pathway, p53 pathway. Researches on HSP mechanism may help to seek future therapeutic targets.

In light of the existing research and biological application of HSP family proteins, there have been a lot of researches in the development of inhibitors for lung cancer and some have entered clinical trials. For example, phase I clinical trials of Debio 0932 and preclinical trials of 17-DMAG have been successfully conducted. In addition, inhibitors for HSP90 like NVP-AUY922, STA-9090, AT13387 and IPI504 have entered phase II clinical trials. OGX-427, an inhibitor of HSP27, has also entered phase II clinical trials. However, HSP70 inhibitors, such as Pifithrin-μ and VER-155008, and HSP60 inhibitor KHS101 have just entered preclinical experiments [107, 108, 109, 110, 111, 112, 113, 114]. Therefore, there is a large research potential in the development of inhibitors targeting HSP family in lung cancer.

Author Contributions

GuaZ and YP prepared the manuscript and figures. KZ helped with references collection and figures designation. YC and GaoZ designed manuscript outline and revised manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This research was funded by the National Natural Science Foundation of China (No. 31771549 to Y Chen).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Hirsch FR, Scagliotti GV, Mulshine JL, Kwon R, Curran WJ, Jr, Wu YL, et al. Lung cancer: current therapies and new targeted treatments. Lancet (London, England). 2017; 389: 299–311.
[2]
Rock CL, Thomson CA, Sullivan KR, Howe CL, Kushi LH, Caan BJ, et al. American Cancer Society nutrition and physical activity guideline for cancer survivors. CA: a Cancer Journal for Clinicians. 2022; 72: 230–262.
[3]
Chen P, Liu Y, Wen Y, Zhou C. Non-small cell lung cancer in China. Cancer Communications (London, England). 2022; 42: 937–970.
[4]
Paz-Ares L, Mezger J, Ciuleanu TE, Fischer JR, von Pawel J, Provencio M, et al. Necitumumab plus pemetrexed and cisplatin as first-line therapy in patients with stage IV non-squamous non-small-cell lung cancer (INSPIRE): an open-label, randomised, controlled phase 3 study. The Lancet. Oncology. 2015; 16: 328–337.
[5]
Field JK, Duffy SW, Baldwin DR, Whynes DK, Devaraj A, Brain KE, et al. UK Lung Cancer RCT Pilot Screening Trial: baseline findings from the screening arm provide evidence for the potential implementation of lung cancer screening. Thorax. 2016; 71: 161–170.
[6]
Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature. 2018; 553: 446–454.
[7]
da Cunha Santos G, Shepherd FA, Tsao MS. EGFR mutations and lung cancer. Annual Review of Pathology. 2011; 6: 49–69.
[8]
Vahid S, Thaper D, Gibson KF, Bishop JL, Zoubeidi A. Molecular chaperone Hsp27 regulates the Hippo tumor suppressor pathway in cancer. Scientific Reports. 2016; 6: 31842.
[9]
Bagatell R, Whitesell L. Altered Hsp90 function in cancer: a unique therapeutic opportunity. Molecular Cancer Therapeutics. 2004; 3: 1021–1030.
[10]
Dokladny K, Myers OB, Moseley PL. Heat shock response and autophagy–cooperation and control. Autophagy. 2015; 11: 200–213.
[11]
Jäättelä M. Heat shock proteins as cellular lifeguards. Annals of Medicine. 1999; 31: 261–271.
[12]
Lindquist S, Craig EA. The heat-shock proteins. Annual Review of Genetics. 1988; 22: 631–677.
[13]
Burdon RH. The heat shock proteins. Endeavour. 1988; 12: 133–138.
[14]
Xu Q. Role of heat shock proteins in atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2002; 22: 1547–1559.
[15]
Seigneuric R, Mjahed H, Gobbo J, Joly AL, Berthenet K, Shirley S, et al. Heat shock proteins as danger signals for cancer detection. Frontiers in Oncology. 2011; 1: 37.
[16]
Huang Q, Zu Y, Fu X, Wu T. Expression of heat shock protein 70 and 27 in non-small cell lung cancer and its clinical significance. Journal of Huazhong University of Science and Technology. Medical Sciences. Yi Xue Ying De Wen Ban. Yixue Yingdewen Ban. 2005; 25: 693–695.
[17]
Jiao W, Hong W, Li P, Sun S, Ma J, Qian M, et al. The dramatically increased chaperone activity of small heat-shock protein IbpB is retained for an extended period of time after the stress condition is removed. The Biochemical Journal. 2008; 410: 63–70.
[18]
Wang H, Xing J, Wang F, Han W, Ren H, Wu T, et al. Expression of Hsp27 and Hsp70 in lymphocytes and plasma in healthy workers and coal miners with lung cancer. Journal of Huazhong University of Science and Technology. Medical Sciences. Yi Xue Ying De Wen Ban. Yixue Yingdewen Ban. 2010; 30: 415–420.
[19]
Calderwood SK, Gong J. Heat Shock Proteins Promote Cancer: It’s a Protection Racket. Trends in Biochemical Sciences. 2016; 41: 311–323.
[20]
Zhong L, Peng X, Hidalgo GE, Doherty DE, Stromberg AJ, Hirschowitz EA. Antibodies to HSP70 and HSP90 in serum in non-small cell lung cancer patients. Cancer Detection and Prevention. 2003; 27: 285–290.
[21]
Sauvageot CME, Weatherbee JL, Kesari S, Winters SE, Barnes J, Dellagatta J, et al. Efficacy of the HSP90 inhibitor 17-AAG in human glioma cell lines and tumorigenic glioma stem cells. Neuro-oncology. 2009; 11: 109–121.
[22]
Shen Y, Ji C, Jian X, Zhou J, Zhang Q, Qiao N, et al. Regulation of the EGFR Pathway by HSP90 Is Involved in the Pathogenesis of Cushing’s Disease. Frontiers in Endocrinology. 2021; 11: 601984.
[23]
Lee SC, Min HY, Choi H, Kim HS, Kim KC, Park SJ, et al. Synthesis and Evaluation of a Novel Deguelin Derivative, L80, which Disrupts ATP Binding to the C-terminal Domain of Heat Shock Protein 90. Molecular Pharmacology. 2015; 88: 245–255.
[24]
Owen S, Zhao H, Dart A, Wang Y, Ruge F, Gao Y, et al. Heat shock protein 27 is a potential indicator for response to YangZheng XiaoJi and chemotherapy agents in cancer cells. International Journal of Oncology. 2016; 49: 1839–1847.
[25]
van Heijst JWJ, Niessen HWM, Musters RJ, van Hinsbergh VWM, Hoekman K, Schalkwijk CG. Argpyrimidine-modified Heat shock protein 27 in human non-small cell lung cancer: a possible mechanism for evasion of apoptosis. Cancer Letters. 2006; 241: 309–319.
[26]
Hall JA, Forsberg LK, Blagg BSJ. Alternative approaches to Hsp90 modulation for the treatment of cancer. Future Medicinal Chemistry. 2014; 6: 1587–1605.
[27]
Prodromou C. Mechanisms of Hsp90 regulation. The Biochemical Journal. 2016; 473: 2439–2452.
[28]
Silverstein AM, Galigniana MD, Chen MS, Owens-Grillo JK, Chinkers M, Pratt WB. Protein phosphatase 5 is a major component of glucocorticoid receptor.hsp90 complexes with properties of an FK506-binding immunophilin. The Journal of Biological Chemistry. 1997; 272: 16224–16230.
[29]
Verma S, Goyal S, Jamal S, Singh A, Grover A. Hsp90: Friends, clients and natural foes. Biochimie. 2016; 127: 227–240.
[30]
Duan XF, Xin YW. Overexpression of molecule GRP94 favors tumor progression in lung adenocarcinoma by interaction with regulatory T cells. Thoracic Cancer. 2020; 11: 704–712.
[31]
Wang Y, Seyed Barghi SM, Yang Y, Akhavan-Sigari R. Value of HSP90α in Lung Cancer Diagnosis and Recurrence Prediction: A Cohort Study. Oncology Research and Treatment. 2021; 44: 583–589.
[32]
Gallegos Ruiz MI, Floor K, Roepman P, Rodriguez JA, Meijer GA, Mooi WJ, et al. Integration of gene dosage and gene expression in non-small cell lung cancer, identification of HSP90 as potential target. PLoS ONE. 2008; 3: e0001722.
[33]
Kim SH, Ji JH, Park KT, Lee JH, Kang KW, Park JH, et al. High-level expression of Hsp90β is associated with poor survival in resectable non-small-cell lung cancer patients. Histopathology. 2015; 67: 509–519.
[34]
Zhang X, Dong Y, Gao M, Hao M, Ren H, Guo L, et al. Knockdown of TRAP1 promotes cisplatin-induced apoptosis by promoting the ROS-dependent mitochondrial dysfunction in lung cancer cells. Molecular and Cellular Biochemistry. 2021; 476: 1075–1082.
[35]
Gallerne C, Prola A, Lemaire C. Hsp90 inhibition by PU-H71 induces apoptosis through endoplasmic reticulum stress and mitochondrial pathway in cancer cells and overcomes the resistance conferred by Bcl-2. Biochimica et Biophysica Acta. 2013; 1833: 1356–1366.
[36]
Wang YQ, Shen AJ, Sun JY, Wang X, Liu HC, Zhang MM, et al. Targeting Hsp90 with FS-108 circumvents gefitinib resistance in EGFR mutant non-small cell lung cancer cells. Acta Pharmacologica Sinica. 2016; 37: 1587–1596.
[37]
Chen Z, Akbay E, Mikse O, Tupper T, Cheng K, Wang Y, et al. Co-clinical trials demonstrate superiority of crizotinib to chemotherapy in ALK-rearranged non-small cell lung cancer and predict strategies to overcome resistance. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2014; 20: 1204–1211.
[38]
Xu Y, Singer MA, Lindquist S. Maturation of the tyrosine kinase c-src as a kinase and as a substrate depends on the molecular chaperone Hsp90. Proceedings of the National Academy of Sciences of the United States of America. 1999; 96: 109–114.
[39]
Grbovic OM, Basso AD, Sawai A, Ye Q, Friedlander P, Solit D, et al. V600E B-Raf requires the Hsp90 chaperone for stability and is degraded in response to Hsp90 inhibitors. Proceedings of the National Academy of Sciences of the United States of America. 2006; 103: 57–62.
[40]
Park KS, Oh B, Lee MH, Nam KY, Jin HR, Yang H, et al. The HSP90 inhibitor, NVP-AUY922, sensitizes KRAS-mutant non-small cell lung cancer with intrinsic resistance to MEK inhibitor, trametinib. Cancer Letters. 2016; 372: 75–81.
[41]
Yang H, Liang SQ, Xu D, Yang Z, Marti TM, Gao Y, et al. HSP90/AXL/eIF4E-regulated unfolded protein response as an acquired vulnerability in drug-resistant KRAS-mutant lung cancer. Oncogenesis. 2019; 8: 45.
[42]
Wang X, Ju W, Renouard J, Aden J, Belinsky SA, Lin Y. 17-allylamino-17-demethoxygeldanamycin synergistically potentiates tumor necrosis factor-induced lung cancer cell death by blocking the nuclear factor-kappaB pathway. Cancer Research. 2006; 66: 1089–1095.
[43]
Beck TN, Korobeynikov VA, Kudinov AE, Georgopoulos R, Solanki NR, Andrews-Hoke M, et al. Anti-Müllerian Hormone Signaling Regulates Epithelial Plasticity and Chemoresistance in Lung Cancer. Cell Reports. 2016; 16: 657–671.
[44]
Brocchieri L, Conway de Macario E, Macario AJL. hsp70 genes in the human genome: Conservation and differentiation patterns predict a wide array of overlapping and specialized functions. BMC Evolutionary Biology. 2008; 8: 19.
[45]
Munro S, Pelham HR. A C-terminal signal prevents secretion of luminal ER proteins. Cell. 1987; 48: 899–907.
[46]
Daugaard M, Kirkegaard-Sørensen T, Ostenfeld MS, Aaboe M, Høyer-Hansen M, Orntoft TF, et al. Lens epithelium-derived growth factor is an Hsp70-2 regulated guardian of lysosomal stability in human cancer. Cancer Research. 2007; 67: 2559–2567.
[47]
Saibil H. Chaperone machines for protein folding, unfolding and disaggregation. Nature Reviews. Molecular Cell Biology. 2013; 14: 630–642.
[48]
Kumar DP, Vorvis C, Sarbeng EB, Cabra Ledesma VC, Willis JE, Liu Q. The four hydrophobic residues on the Hsp70 inter-domain linker have two distinct roles. Journal of Molecular Biology. 2011; 411: 1099–1113.
[49]
Swain JF, Dinler G, Sivendran R, Montgomery DL, Stotz M, Gierasch LM. Hsp70 chaperone ligands control domain association via an allosteric mechanism mediated by the interdomain linker. Molecular Cell. 2007; 26: 27–39.
[50]
Kampinga HH, Craig EA. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nature Reviews. Molecular Cell Biology. 2010; 11: 579–592.
[51]
Pfister K, Radons J, Busch R, Tidball JG, Pfeifer M, Freitag L, et al. Patient survival by Hsp70 membrane phenotype: association with different routes of metastasis. Cancer. 2007; 110: 926–935.
[52]
Suzuki K, Ito Y, Wakai K, Kawado M, Hashimoto S, Seki N, et al. Serum heat shock protein 70 levels and lung cancer risk: a case-control study nested in a large cohort study. Cancer Epidemiology, Biomarkers & Prevention: a Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology. 2006; 15: 1733–1737.
[53]
Vostakolaei MA, Abdolalizadeh J, Hejazi MS, Kordi S, Molavi O. Hsp70 in Cancer: Partner or Traitor to Immune System. Iranian Journal of Allergy, Asthma, and Immunology. 2019; 18: 589–604.
[54]
Steel R, Doherty JP, Buzzard K, Clemons N, Hawkins CJ, Anderson RL. Hsp72 inhibits apoptosis upstream of the mitochondria and not through interactions with Apaf-1. The Journal of Biological Chemistry. 2004; 279: 51490–51499.
[55]
Gao Y, Han C, Huang H, Xin Y, Xu Y, Luo L, et al. Heat shock protein 70 together with its co-chaperone CHIP inhibits TNF-alpha induced apoptosis by promoting proteasomal degradation of apoptosis signal-regulating kinase1. Apoptosis: an International Journal on Programmed Cell Death. 2010; 15: 822–833.
[56]
Tournier C, Hess P, Yang DD, Xu J, Turner TK, Nimnual A, et al. Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway. Science (New York, N.Y.). 2000; 288: 870–874.
[57]
Guo F, Sigua C, Bali P, George P, Fiskus W, Scuto A, et al. Mechanistic role of heat shock protein 70 in Bcr-Abl-mediated resistance to apoptosis in human acute leukemia cells. Blood. 2005; 105: 1246–1255.
[58]
Cao X, Zhou Y, Sun H, Xu M, Bi X, Zhao Z, et al. EGFR-TKI-induced HSP70 degradation and BER suppression facilitate the occurrence of the EGFR T790 M resistant mutation in lung cancer cells. Cancer Letters. 2018; 424: 84–96.
[59]
Wang Z, Liu G, Jiang J. Profiling of apoptosis- and autophagy-associated molecules in human lung cancer A549 cells in response to cisplatin treatment using stable isotope labeling with amino acids in cell culture. International Journal of Oncology. 2019; 54: 1071–1085.
[60]
Ei ZZ, Choochuay K, Tubsuwan A, Pinkaew D, Suksomtip M, Vinayanuwattikun C, et al. GRP78/BiP determines senescence evasion cell fate after cisplatin-based chemotherapy. Scientific Reports. 2021; 11: 22448.
[61]
Lim S, Cho HY, Kim DG, Roh Y, Son SY, Mushtaq AU, et al. Targeting the interaction of AIMP2-DX2 with HSP70 suppresses cancer development. Nature Chemical Biology. 2020; 16: 31–41.
[62]
Choi JW, Kim DG, Lee AE, Kim HR, Lee JY, Kwon NH, et al. Cancer-associated splicing variant of tumor suppressor AIMP2/p38: pathological implication in tumorigenesis. PLoS Genetics. 2011; 7: e1001351.
[63]
Du T, Li H, Fan Y, Yuan L, Guo X, Zhu Q, et al. The deubiquitylase OTUD3 stabilizes GRP78 and promotes lung tumorigenesis. Nature Communications. 2019; 10: 2914.
[64]
Antognelli C, Gambelunghe A, Muzi G, Talesa VN. Glyoxalase I drives epithelial-to-mesenchymal transition via argpyrimidine-modified Hsp70, miR-21 and SMAD signalling in human bronchial cells BEAS-2B chronically exposed to crystalline silica Min-U-Sil 5: Transformation into a neoplastic-like phenotype. Free Radical Biology & Medicine. 2016; 92: 110–125.
[65]
Sun LL, Chen CM, Zhang J, Wang J, Yang CZ, Lin LZ. Glucose-Regulated Protein 78 Signaling Regulates Hypoxia-Induced Epithelial-Mesenchymal Transition in A549 Cells. Frontiers in Oncology. 2019; 9: 137.
[66]
Dai C, Whitesell L, Rogers AB, Lindquist S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell. 2007; 130: 1005–1018.
[67]
Nakashima M, Adachi S, Yasuda I, Yamauchi T, Kawaguchi J, Itani M, et al. Phosphorylation status of heat shock protein 27 plays a key role in gemcitabine-induced apoptosis of pancreatic cancer cells. Cancer Letters. 2011; 313: 218–225.
[68]
Wu R, Kausar H, Johnson P, Montoya-Durango DE, Merchant M, Rane MJ. Hsp27 regulates Akt activation and polymorphonuclear leukocyte apoptosis by scaffolding MK2 to Akt signal complex. The Journal of Biological Chemistry. 2007; 282: 21598–21608.
[69]
Sakamoto H, Mashima T, Yamamoto K, Tsuruo T. Modulation of heat-shock protein 27 (Hsp27) anti-apoptotic activity by methylglyoxal modification. The Journal of Biological Chemistry. 2002; 277: 45770–45775.
[70]
Rogalla T, Ehrnsperger M, Preville X, Kotlyarov A, Lutsch G, Ducasse C, et al. Regulation of Hsp27 oligomerization, chaperone function, and protective activity against oxidative stress/tumor necrosis factor alpha by phosphorylation. The Journal of Biological Chemistry. 1999; 274: 18947–18956.
[71]
Bruey JM, Paul C, Fromentin A, Hilpert S, Arrigo AP, Solary E, et al. Differential regulation of HSP27 oligomerization in tumor cells grown in vitro and in vivo. Oncogene. 2000; 19: 4855–4863.
[72]
Sheng B, Qi C, Liu B, Lin Y, Fu T, Zeng Q. Increased HSP27 correlates with malignant biological behavior of non-small cell lung cancer and predicts patient’s survival. Scientific Reports. 2017; 7: 13807.
[73]
Li S, Zhang W, Fan J, Lai Y, Che G. Clinicopathological and prognostic significance of heat shock protein 27 (HSP27) expression in non-small cell lung cancer: a systematic review and meta-analysis. SpringerPlus. 2016; 5: 1165.
[74]
Yu Z, Zhi J, Peng X, Zhong X, Xu A. Clinical significance of HSP27 expression in colorectal cancer. Molecular Medicine Reports. 2010; 3: 953–958.
[75]
Liang C, Xu Y, Ge H, Li G, Wu J. The clinicopathological and prognostic value of HSP27 in hepatocellular carcinoma: a systematic review and meta-analysis. OncoTargets and Therapy. 2018; 11: 1293–1303.
[76]
Zimmermann M, Nickl S, Lambers C, Hacker S, Mitterbauer A, Hoetzenecker K, et al. Discrimination of clinical stages in non-small cell lung cancer patients by serum HSP27 and HSP70: a multi-institutional case-control study. Clinica Chimica Acta; International Journal of Clinical Chemistry. 2012; 413: 1115–1120.
[77]
Aydin C, Sanlioglu AD, Karacay B, Ozbilim G, Dertsiz L, Ozbudak O, et al. Decoy receptor-2 small interfering RNA (siRNA) strategy employing three different siRNA constructs in combination defeats adenovirus-transferred tumor necrosis factor-related apoptosis-inducing ligand resistance in lung cancer cells. Human Gene Therapy. 2007; 18: 39–50.
[78]
Sanlioglu AD, Karacay B, Koksal IT, Griffith TS, Sanlioglu S. DcR2 (TRAIL-R4) siRNA and adenovirus delivery of TRAIL (Ad5hTRAIL) break down in vitro tumorigenic potential of prostate carcinoma cells. Cancer Gene Therapy. 2007; 14: 976–984.
[79]
Zhuang H, Jiang W, Cheng W, Qian K, Dong W, Cao L, et al. Down-regulation of HSP27 sensitizes TRAIL-resistant tumor cell to TRAIL-induced apoptosis. Lung Cancer (Amsterdam, Netherlands). 2010; 68: 27–38.
[80]
Xu L, Lin X, Zheng Y, Zhou H. Silencing of heat shock protein 27 increases the radiosensitivity of non small cell lung carcinoma cells. Molecular Medicine Reports. 2019; 20: 613–621.
[81]
Bruey JM, Ducasse C, Bonniaud P, Ravagnan L, Susin SA, Diaz-Latoud C, et al. Hsp27 negatively regulates cell death by interacting with cytochrome c. Nature Cell Biology. 2000; 2: 645–652.
[82]
Wyttenbach A, Sauvageot O, Carmichael J, Diaz-Latoud C, Arrigo AP, Rubinsztein DC. Heat shock protein 27 prevents cellular polyglutamine toxicity and suppresses the increase of reactive oxygen species caused by huntingtin. Human Molecular Genetics. 2002; 11: 1137–1151.
[83]
Rane MJ, Pan Y, Singh S, Powell DW, Wu R, Cummins T, et al. Heat shock protein 27 controls apoptosis by regulating Akt activation. The Journal of Biological Chemistry. 2003; 278: 27828–27835.
[84]
Charette SJ, Landry J. The interaction of HSP27 with Daxx identifies a potential regulatory role of HSP27 in Fas-induced apoptosis. Annals of the New York Academy of Sciences. 2000; 926: 126–131.
[85]
Charette SJ, Lavoie JN, Lambert H, Landry J. Inhibition of Daxx-mediated apoptosis by heat shock protein 27. Molecular and Cellular Biology. 2000; 20: 7602–7612.
[86]
Huang Z, Yang C, Sun S, Nan Y, Lang Z, Wang X, et al. Heat Shock Protein 27, a Novel Regulator of Transforming Growth Factor β Induced Resistance to Cisplatin in A549 Cell. Pharmacology. 2017; 100: 283–291.
[87]
Choi SK, Kim M, Lee H, Kwon Y, Cha HJ, Jang SJ, et al. Activation of the HSP27-AKT axis contributes to gefitinib resistance in non-small cell lung cancer cells independent of EGFR mutations. Cellular Oncology (Dordrecht). 2022; 45: 913–930.
[88]
Martelli AM, Tabellini G, Bressanin D, Ognibene A, Goto K, Cocco L, et al. The emerging multiple roles of nuclear Akt. Biochimica et Biophysica Acta. 2012; 1823: 2168–2178.
[89]
Clark GJ, Drugan JK, Rossman KL, Carpenter JW, Rogers-Graham K, Fu H, et al. 14-3-3 zeta negatively regulates raf-1 activity by interactions with the Raf-1 cysteine-rich domain. The Journal of Biological Chemistry. 1997; 272: 20990–20993.
[90]
Zhang S, Hu Y, Huang Y, Xu H, Wu G, Dai H. Heat shock protein 27 promotes cell proliferation through activator protein-1 in lung cancer. Oncology Letters. 2015; 9: 2572–2576.
[91]
Shi J, Fu M, Zhao C, Zhou F, Yang Q, Qiu L. Characterization and function analysis of Hsp60 and Hsp10 under different acute stresses in black tiger shrimp, Penaeus monodon. Cell Stress & Chaperones. 2016; 21: 295–312.
[92]
Clare DK, Saibil HR. ATP-driven molecular chaperone machines. Biopolymers. 2013; 99: 846–859.
[93]
Chun JN, Choi B, Lee KW, Lee DJ, Kang DH, Lee JY, et al. Cytosolic Hsp60 is involved in the NF-kappaB-dependent survival of cancer cells via IKK regulation. PLoS ONE. 2010; 5: e9422.
[94]
Campanella C, Rappa F, Sciumè C, Marino Gammazza A, Barone R, Bucchieri F, et al. Heat shock protein 60 levels in tissue and circulating exosomes in human large bowel cancer before and after ablative surgery. Cancer. 2015; 121: 3230–3239.
[95]
Michils A, Redivo M, Zegers de Beyl V, de Maertelaer V, Jacobovitz D, Rocmans P, et al. Increased expression of high but not low molecular weight heat shock proteins in resectable lung carcinoma. Lung Cancer (Amsterdam, Netherlands). 2001; 33: 59–67.
[96]
Xu X, Wang W, Shao W, Yin W, Chen H, Qiu Y, et al. Heat shock protein-60 expression was significantly correlated with the prognosis of lung adenocarcinoma. Journal of Surgical Oncology. 2011; 104: 598–603.
[97]
Mori D, Nakafusa Y, Miyazaki K, Tokunaga O. Differential expression of Janus kinase 3 (JAK3), matrix metalloproteinase 13 (MMP13), heat shock protein 60 (HSP60), and mouse double minute 2 (MDM2) in human colorectal cancer progression using human cancer cDNA microarrays. Pathology, Research and Practice. 2005; 201: 777–789.
[98]
Campanella C, Bucchieri F, Ardizzone NM, Marino Gammazza A, Montalbano A, Ribbene A, et al. Upon oxidative stress, the antiapoptotic Hsp60/procaspase-3 complex persists in mucoepidermoid carcinoma cells. European Journal of Histochemistry: EJH. 2008; 52: 221–228.
[99]
Ağababaoğlu İ, Önen A, Demir AB, Aktaş S, Altun Z, Ersöz H, et al. Chaperonin (HSP60) and annexin-2 are candidate biomarkers for non-small cell lung carcinoma. Medicine. 2017; 96: e5903.
[100]
Gupta S, Knowlton AA. HSP60, Bax, apoptosis and the heart. Journal of Cellular and Molecular Medicine. 2005; 9: 51–58.
[101]
Mulati S, Jiang R, Wang J, Tao Y, Zhang W. 6-Shogaol Exhibits a Promoting Effect with Tax via Binding HSP60 in Non-Small-Cell Lung Cancer. Cells. 2022; 11: 3678.
[102]
Hayoun D, Kapp T, Edri-Brami M, Ventura T, Cohen M, Avidan A, et al. HSP60 is transported through the secretory pathway of 3-MCA-induced fibrosarcoma tumour cells and undergoes N-glycosylation. The FEBS Journal. 2012; 279: 2083–2095.
[103]
Singh P, Kumari M, Bal A, Srinivasan R, Ghosh S. Heat shock protein 60 is a disease-associated sialoglycoprotein in human non-small cell lung cancer. Biological Chemistry. 2020; 401: 969–983.
[104]
Marino Gammazza A, Campanella C, Barone R, Caruso Bavisotto C, Gorska M, Wozniak M, et al. Doxorubicin anti-tumor mechanisms include Hsp60 post-translational modifications leading to the Hsp60/p53 complex dissociation and instauration of replicative senescence. Cancer Letters. 2017; 385: 75–86.
[105]
Litwiniec A, Grzanka A, Helmin-Basa A, Gackowska L, Grzanka D. Features of senescence and cell death induced by doxorubicin in A549 cells: organization and level of selected cytoskeletal proteins. Journal of Cancer Research and Clinical Oncology. 2010; 136: 717–736.
[106]
Childs BG, Baker DJ, Kirkland JL, Campisi J, van Deursen JM. Senescence and apoptosis: dueling or complementary cell fates? EMBO Reports. 2014; 15: 1139–1153.
[107]
Parma B, Ramesh V, Gollavilli PN, Siddiqui A, Pinna L, Schwab A, et al. Metabolic impairment of non-small cell lung cancers by mitochondrial HSPD1 targeting. Journal of Experimental & Clinical Cancer Research: CR. 2021; 40: 248.
[108]
Spigel DR, Shipley DL, Waterhouse DM, Jones SF, Ward PJ, Shih KC, et al. A Randomized, Double-Blinded, Phase II Trial of Carboplatin and Pemetrexed with or without Apatorsen (OGX-427) in Patients with Previously Untreated Stage IV Non-Squamous-Non-Small-Cell Lung Cancer: The SPRUCE Trial. The Oncologist. 2019; 24: e1409–e1416.
[109]
Krause SW, Gastpar R, Andreesen R, Gross C, Ullrich H, Thonigs G, et al. Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: a clinical phase i trial. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2004; 10: 3699–3707.
[110]
Zhou Y, Ma J, Zhang J, He L, Gong J, Long C. Pifithrin-μ is efficacious against non-small cell lung cancer via inhibition of heat shock protein 70. Oncology Reports. 2017; 37: 313–322.
[111]
Wen W, Liu W, Shao Y, Chen L. VER-155008, a small molecule inhibitor of HSP70 with potent anti-cancer activity on lung cancer cell lines. Experimental Biology and Medicine (Maywood, N.J.). 2014; 239: 638–645.
[112]
Segawa T, Fujii Y, Tanaka A, Bando SI, Okayasu R, Ohnishi K, et al. Radiosensitization of human lung cancer cells by the novel purine-scaffold Hsp90 inhibitor, PU-H71. International Journal of Molecular Medicine. 2014; 33: 559–564.
[113]
Mellatyar H, Talaei S, Pilehvar-Soltanahmadi Y, Dadashpour M, Barzegar A, Akbarzadeh A, et al. 17-DMAG-loaded nanofibrous scaffold for effective growth inhibition of lung cancer cells through targeting HSP90 gene expression. Biomedicine & Pharmacotherapy. 2018; 105: 1026–1032.
[114]
Parma B, Wurdak H, Ceppi P. Harnessing mitochondrial metabolism and drug resistance in non-small cell lung cancer and beyond by blocking heat-shock proteins. Drug Resistance Updates: Reviews and Commentaries in Antimicrobial and Anticancer Chemotherapy. 2022; 65: 100888.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top