Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors
Abstract
:1. Overview on Combinational Anti-Cancer Therapies
2. Molecular Chaperones as Drug Targets in Combinational Therapy
2.1. Inhibitors of HSF in Anti-Cancer Therapy Formulations
2.2. HSP90-Targeting Molecules in Anti-Cancer Therapeutic Schedules
2.3. Combinations of HSP70 Inhibitors with Anti-Tumor Drugs
2.4. Combinations of HSP27 Inhibitors with Anti-Tumor Drugs
3. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
17-AAG | 17-(allylamino)-17-demethoxygeldanamycin |
17-DMAG | 17-desmethoxy-17-N,N-dimethylaminoethylaminogeldanamycin |
ADP | adenosine diphosphate |
Akt | serine-threonine protein kinase |
AMPK | AMP-activated protein kinase |
ATP | adenosine triphosphate |
ATPase | adenosine triphosphatase |
Bcl-2 | B-cell lymphoma 2 protein |
BRAF | B-Raf proto-oncogene |
CTD | C-terminal domain |
CTLA-4 | cytotoxic T-lymphocyte-associated protein 4 |
EGCG | epigallocatechin gallate |
EGFR | epidermal growth factor receptor |
EMT | epithelial mesenchymal transition |
EphA2 | ephrin type-A receptor 2 |
GDA | geldanamycin |
GRP94 | glucose related protein |
HER | human epidermal growth factor receptor |
HIF-1α | hypoxia-inducible factor 1-alpha |
HSE | heat shock element |
HSF1 | heat shock factor 1 |
HSP | heat shock proteins |
JAK2 | Janus kinase 2 |
KRAS | K-Ras proto-oncogene |
MAPK | mitogen-activated protein kinase |
MCL-1 | induced myeloid leukemia cell differentiation protein |
MDM2 | mouse double minute 2 homolog |
MEK | mitogen-activated protein kinase |
mTOR | mammalian target of rapamycin |
NBD | nucleotide-binding domain |
NSCLC | non-small cell lung cancer |
PAC-1 | procaspase activating compound 1 |
PES | 2-phenylethynesulfonamide, pifithrin-μ |
PD-1 | programmed cell death protein 1 |
PD-L1 | programmed death-ligand 1 |
pTEFb | positive transcription elongation factor b |
RDC | radicicol |
TFIIH | transcription factor II human |
XPB | xeroderma pigmentosum type B |
References
- Jia, J.; Zhu, F.; Ma, X.; Cao, Z.; Cao, Z.W.; Li, Y.; Li, Y.X.; Chen, Y.Z. Mechanisms of drug combinations: Interaction and network perspectives. Nat. Rev. Drug Discov. 2009, 8, 111–128. [Google Scholar] [CrossRef] [PubMed]
- O’Neil, J.; Benita, Y.; Feldman, I.; Chenard, M.; Roberts, B.; Liu, Y.; Li, J.; Kral, A.; Lejnine, S.; Loboda, A.; et al. An Unbiased Oncology Compound Screen to Identify Novel Combination Strategies. Mol. Cancer Ther. 2016, 15, 1155–1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Friedman, A.A.; Amzallag, A.; Pruteanu-Malinici, I.; Baniya, S.; Cooper, Z.A.; Piris, A.; Hargreaves, L.; Igras, V.; Frederick, D.T.; Lawrence, D.P.; et al. Landscape of targeted anti-cancer drug synergies in melanoma identifies a novel BRAF-VEGFR/PDGFR combination treatment. PLoS ONE 2015, 10, e0140310. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Xu, Y.; Cui, H.; Huang, T.; Wang, D.; Lian, B.; Li, W.; Qin, G.; Chen, L.; Xie, L. Prediction of synergistic anti-cancer drug combinations based on drug target network and drug induced gene expression profiles. Artif. Intell. Med. 2017, 83, 35–43. [Google Scholar] [CrossRef] [PubMed]
- Sorokin, M.; Kholodenko, R.; Suntsova, M.; Malakhova, G.; Garazha, A.; Kholodenko, I.; Poddubskaya, E.; Lantsov, D.; Stilidi, I.; Arhiri, P.; et al. Oncobox bioinformatical platform for selecting potentially effective combinations of target cancer drugs using high-throughput gene expression data. Cancers 2018, 10, 365. [Google Scholar] [CrossRef] [PubMed]
- Pan, J. RNA polymerase—An important molecular target of triptolide in cancer cells. Cancer Lett. 2010, 292, 149–152. [Google Scholar] [CrossRef] [PubMed]
- Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef]
- Csermely, P.; Hódsági, J.; Korcsmáros, T.; Módos, D.; Perez-Lopez, Á.R.; Szalay, K.; Veres, D.V.; Lenti, K.; Wu, L.Y.; Zhang, X.S. Cancer stem cells display extremely large evolvability: Alternating plastic and rigid networks as a potential Mechanism: Network models, novel therapeutic target strategies, and the contributions of hypoxia, inflammation and cellular senescence. Semin. Cancer Biol. 2015, 30, 42–51. [Google Scholar] [CrossRef]
- Lamb, J.; Crawford, E.D.; Peck, D.; Modell, J.W.; Blat, I.C.; Wrobel, M.J.; Lerner, J.; Brunet, J.P.; Subramanian, A.; Ross, K.N.; et al. The Connectivity Map: Using gene-expression signatures to connect small molecules, genes, and disease. Science 2006, 29, 1929–1935. [Google Scholar] [CrossRef]
- Wedel, S.; Hudak, L.; Seibel, J.M.; Makarević, J.; Juengel, E.; Tsaur, I.; Waaga-Gasser, A.; Haferkamp, A.; Blaheta, R.A. Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis. BMC Cancer. 2011, 11, 375. [Google Scholar] [CrossRef]
- Ouchida, A.T.; Li, Y.; Geng, J.; Najafov, A.; Ofengeim, D.; Sun, X.; Yu, Q.; Yuan, J. Synergistic effect of a novel autophagy inhibitor and Quizartinib enhances cancer cell death. Cell Death Dis. 2018, 9, 138. [Google Scholar] [CrossRef] [PubMed]
- Peh, J.; Fan, T.M.; Wycislo, K.L.; Roth, H.S.; Hergenrother, P.J. The Combination of Vemurafenib and Procaspase-3 Activation Is Synergistic in Mutant BRAF Melanomas. Mol. Cancer Ther. 2016, 15, 1859–1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Y.; Zhu, J.; Zhang, H.; Liu, Y.; Sun, H. Metformin inhibits ovarian cancer growth and migration in vitro and in vivo by enhancing cisplatin cytotoxicity. Am. J. Transl. Res. 2018, 10, 3086–3098. [Google Scholar] [PubMed]
- Song, W.; Tang, Z.; Zhang, D.; Zhang, Y.; Yu, H.; Li, M.; Lv, S.; Sun, H.; Deng, M.; Chen, X. Anti-tumor efficacy of c(RGDfK)-decorated polypeptide-based micelles co-loaded with docetaxel and cisplatin. Biomaterials 2014, 35, 3005–3014. [Google Scholar] [CrossRef] [PubMed]
- Cavazzoni, A.; Alfieri, R.R.; Cretella, D.; Saccani, F.; Ampollini, L.; Galetti, M.; Quaini, F.; Graiani, G.; Madeddu, D.; Mozzoni, P.; et al. Combined use of anti-ErbB monoclonal antibodies and erlotinib enhances antibody-dependent cellular cytotoxicity of wild-type erlotinib-sensitive NSCLC cell lines. Mol. Cancer. 2012, 11, 91. [Google Scholar] [CrossRef]
- Kang, Y.W.; Lee, J.E.; Jung, K.H.; Son, M.K.; Shin, S.M.; Kim, S.J.; Fang, Z.; Yan, H.H.; Park, J.H.; Han, B.; et al. KRAS targeting antibody synergizes anti-cancer activity of gemcitabine against pancreatic cancer. Cancer Lett. 2018, 438, 174–186. [Google Scholar] [CrossRef]
- Hodi, F.S.; Lawrence, D.; Lezcano, C.; Wu, X.; Zhou, J.; Sasada, T.; Zeng, W.; Giobbie-Hurder, A.; Atkins, M.B.; Ibrahim, N.; et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014, 2, 632–642. [Google Scholar] [CrossRef]
- Hu-Lieskovan, S.; Mok, S.; Homet Moreno, B.; Tsoi, J.; Robert, L.; Goedert, L.; Pinheiro, E.M.; Koya, R.C.; Graeber, T.G.; Comin-Anduix, B.; et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci. Transl. Med. 2015, 7, 279ra41. [Google Scholar] [CrossRef]
- Liu, L.; Mayes, P.A.; Eastman, S.; Shi, H.; Yadavilli, S.; Zhang, T.; Yang, J.; Seestaller-Wehr, L.; Zhang, S.Y.; Hopson, C.; et al. The BRAF and MEK Inhibitors Dabrafenib and Trametinib: Effects on Immune Function and in Combination with Immunomodulatory Antibodies Targeting PD-1, PD-L1, and CTLA-4. Clin. Cancer Res. 2015, 21, 1639–1651. [Google Scholar] [CrossRef] [Green Version]
- Lemke, D.; Pledl, H.W.; Zorn, M.; Jugold, M.; Green, E.; Blaes, J.; Löw, S.; Hertenstein, A.; Ott, M.; Sahm, F.; et al. Slowing down glioblastoma progression in mice by running or the anti-malarial drug dihydroartemisinin? Induction of oxidative stress in murine glioblastoma therapy. Oncotarget 2016, 7, 56713–56725. [Google Scholar] [CrossRef]
- Garofalo, M.; Saari, H.; Somersalo, P.; Crescenti, D.; Kuryk, L.; Aksela, L.; Capasso, C.; Madetoja, M.; Koskinen, K.; Oksanen, T.; et al. Antitumor effect of oncolytic virus and paclitaxel encapsulated in extracellular vesicles for lung cancer treatment. J. Control Release 2018, 283, 223–234. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Liu, Y.; Cheng, Y.; Wei, Y.; Wei, X. Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim. Biophys. Acta. Rev. Cancer. 2019, 1871, 199–224. [Google Scholar] [CrossRef] [PubMed]
- Kaur, K.; Rath, G.; Chandra, S.; Singh, R.; Goyal, A.K. Chemotherapy with si-RNA and Anti-Cancer Drugs. Curr. Drug Deliv. 2018, 15, 300–311. [Google Scholar] [CrossRef]
- Akerfelt, M.; Morimoto, R.I.; Sistonen, L. Heat shock factors: Integrators of cell stress, development and lifespan. Nat. Rev. Mol. Cell Biol. 2010, 11, 545–555. [Google Scholar] [CrossRef] [PubMed]
- Anckar, J.; Sistonen, L. Regulation of HSF1 function in the heat stress response: Implications in aging and disease. Annu. Rev. Biochem. 2011, 80, 1089–1115. [Google Scholar] [CrossRef]
- Gomez-Pastor, R.; Burchfiel, E.T.; Thiele, D.J. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat. Rev. Mol. Cell Biol. 2018, 19, 4–19. [Google Scholar] [CrossRef]
- Dai, W.; Ye, J.; Zhang, Z.; Yang, L.; Ren, H.; Wu, H.; Chen, J.; Ma, J.; Zhai, E.; Cai, S.; et al. Increased expression of heat shock factor 1 (HSF1) is associated with poor survival in gastric cancer patients. Diagn. Pathol. 2018, 13, 80. [Google Scholar] [CrossRef]
- Mendillo, M.L.; Santagata, S.; Koeva, M.; Bell, G.W.; Hu, R.; Tamimi, R.M.; Fraenkel, E.; Ince, T.A.; Whitesell, L.; Lindquist, S. HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers. Cell 2012, 150, 549–562. [Google Scholar] [CrossRef]
- Sharma, C.; Seo, Y.H. Small Molecule Inhibitors of HSF1-Activated Pathways as Potential Next-Generation Anticancer Therapeutics. Molecules 2018, 23, 2757. [Google Scholar] [CrossRef]
- Kijima, T.; Prince, T.; Neckers, L.; Koga, F.; Fujii, Y. Heat shock factor 1 (HSF1)-targeted anticancer therapeutics: Overview of current preclinical progress. Expert Opin. Ther. Targets 2019, 23, 369–377. [Google Scholar] [CrossRef]
- Xiong, J.; Su, T.; Qu, Z.; Yang, Q.; Wang, Y.; Li, J.; Zhou, S. Triptolide has anticancer and chemosensitization effects by down-regulating Akt activation through the MDM2/REST pathway in human breast cancer. Oncotarget 2016, 7, 23933–23946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, L.; Xiong, X.; Shen, M.; Ru, D.; Gao, P.; Zhang, X.; Huang, C.; Sun, Y.; Li, H.; Duan, Y. Co-delivery of triptolide and curcumin for ovarian cancer targeting therapy via mPEG-DPPE/CaP nanoparticle. J. Biomed. Nanotechnol. 2018, 14, 1761–1772. [Google Scholar] [CrossRef] [PubMed]
- Carpenter, R.L.; Sirkisoon, S.; Zhu, D.; Rimkus, T.; Harrison, A.; Anderson, A.; Paw, I.; Qasem, S.; Xing, F.; Liu, Y.; et al. Combined inhibition of AKT and HSF1 suppresses breast cancer stem cells and tumor growth. Oncotarget 2017, 8, 73947–73963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nikotina, A.D.; Koludarova, L.; Komarova, E.Y.; Mikhaylova, E.R.; Aksenov, N.D.; Suezov, R.; Kartzev, V.G.; Margulis, B.A.; Guzhova, I.V. Discovery and optimization of cardenolides inhibiting HSF1 activation in human colon HCT-116 cancer cells. Oncotarget 2018, 9, 27268–27279. [Google Scholar] [CrossRef]
- Modi, S.; Stopeck, A.T.; Gordon, M.S.; Mendelson, D.; Solit, D.B.; Bagatell, R.; Ma, W.; Wheler, J.; Rosen, N.; Norton, L.; et al. Combination of trastuzumab and tanespimycin (17-AAG, KOS-953) is safe and active in trastuzumab-refractory HER-2 overexpressing breast cancer: A phase I dose-escalation study. J. Clin. Oncol. Am. Soc. Clin. Oncol. 2007, 25, 5410–5417. [Google Scholar] [CrossRef]
- Acquaviva, J.; Smith, D.L.; Jimenez, J.P.; Zhang, C.; Sequeira, M.; He, S.; Sang, J.; Bates, R.C.; Proia, D.A. Overcoming acquired BRAF inhibitor resistance in melanoma via targeted inhibition of Hsp90 with ganetespib. Mol. Cancer Ther. 2014, 13, 353–363. [Google Scholar] [CrossRef]
- Proia, D.A.; Kaufmann, G.F. Targeting Heat-Shock Protein 90 (HSP90) as a Complementary Strategy to Immune Checkpoint Blockade for Cancer Therapy. Cancer Immunol. Res. 2015, 3, 583–589. [Google Scholar] [CrossRef] [Green Version]
- 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. Exp. Biol. Med. (Maywood) 2014, 239, 638–645. [Google Scholar] [CrossRef]
- Prince, T.; Ackerman, A.; Cavanaugh, A.; Schreiter, B.; Juengst, B.; Andolino, C.; Danella, J.; Chernin, M.; Williams, H. Dual targeting of HSP70 does not induce the heat shock response and synergistically reduces cell viability in muscle invasive bladder cancer. Oncotarget 2018, 9, 32702–32717. [Google Scholar] [CrossRef]
- Kim, S.H.; Kang, J.G.; Kim, C.S.; Ihm, S.H.; Choi, M.G.; Yoo, H.J.; Lee, S.J. Hsp70 inhibition potentiates radicicol-induced cell death in anaplastic thyroid carcinoma cells. Anticancer Res. 2014, 34, 4829–4837. [Google Scholar]
- 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] [PubMed]
- 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]
- Kumano, M.; Furukawa, J.; Shiota, M.; Zardan, A.; Zhang, F.; Beraldi, E.; Wiedmann, R.M.; Fazli, L.; Zoubeidi, A.; Gleave, M.E. Cotargeting stress-activated Hsp27 and autophagy as a combinatorial strategy to amplify endoplasmic reticular stress in prostate cancer. Mol. Cancer Ther. 2012, 11, 1661–1671. [Google Scholar] [CrossRef] [PubMed]
- Lamoureux, F.; Thomas, C.; Yin, M.J.; Fazli, L.; Zoubeidi, A.; Gleave, M.E. Suppression of heat shock protein 27 using OGX-427 induces endoplasmic reticulum stress and potentiates heat shock protein 90 inhibitors to delay castrate-resistant prostate cancer. Eur. Urol. 2014, 66, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Westerheide, S.D.; Kawahara, T.L.; Orton, K.; Morimoto, R.I. Triptolide, an inhibitor of the human heat shock response that enhances stress-induced cell death. J. Biol. Chem. 2006, 281, 9616–9622. [Google Scholar] [CrossRef]
- Titov, D.V.; Gilman, B.; He, Q.L.; Bhat, S.; Low, W.K.; Dang, Y.; Smeaton, M.; Demain, A.L.; Miller, P.S.; Kugel, J.F.; et al. XPB, a subunit of TFIIH, is a target of the natural product triptolide. Nat. Chem. Biol. 2011, 7, 182–188. [Google Scholar] [CrossRef] [Green Version]
- Phillips, P.A.; Dudeja, V.; McCarroll, J.A.; Borja-Cacho, D.; Dawra, R.K.; Grizzle, W.E.; Vickers, S.M.; Saluja, A.K. Triptolide induces pancreatic cancer cell death via inhibition of heat shock protein 70. Cancer Res. 2007, 67, 9407–9416. [Google Scholar] [CrossRef]
- Antonoff, M.B.; Chugh, R.; Borja-Cacho, D.; Dudeja, V.; Clawson, K.A.; Skube, S.J.; Sorenson, B.S.; Saltzman, D.A.; Vickers, S.M.; Saluja, A.K. Triptolide therapy for neuroblastoma decreases cell viability in vitro and inhibits tumor growth in vivo. Surgery 2009, 146, 282–290. [Google Scholar] [CrossRef]
- Banerjee, S.; Thayanithy, V.; Sangwan, V.; Mackenzie, T.N.; Saluja, A.K.; Subramanian, S. Minnelide reduces tumor burden in preclinical models of osteosarcoma. Cancer Lett. 2013, 335, 412–420. [Google Scholar] [CrossRef] [Green Version]
- Wan, C.K.; Wang, C.; Cheung, H.Y.; Yang, M.; Fong, W.F. Triptolide induces Bcl-2 cleavage and mitochondria dependent apoptosis in p53-deficient HL-60 cells. Cancer Lett. 2006, 241, 31–41. [Google Scholar] [CrossRef]
- Xi, C.; Peng, S.; Wu, Z.; Zhou, Q.; Zhou, J. Toxicity of triptolide and the molecular mechanisms involved. Biomed. Pharm. 2017, 90, 531–541. [Google Scholar] [CrossRef] [PubMed]
- Yokota, S.; Kitahara, M.; Nagata, K. Benzylidene lactam compound, KNK437, a novel inhibitor of acquisition of thermotolerance and heat shock protein induction in human colon carcinoma cells. Cancer Res. 2000, 60, 2942–2948. [Google Scholar] [PubMed]
- Qi, W.; White, M.C.; Choi, W.; Guo, C.; Dinney, C.; McConkey, D.J.; Siefker-Radtke, A. Inhibition of inducible heat shock protein-70 (hsp72) enhances bortezomib-induced cell death in human bladder cancer cells. PLoS ONE 2013, 8, e69509. [Google Scholar] [CrossRef] [PubMed]
- Bustany, S.; Cahu, J.; Descamps, G.; Pellat-Deceunynck, C.; Sola, B. Heat shock factor 1 is a potent therapeutic target for enhancing the efficacy of treatments for multiple myeloma with adverse prognosis. J. Hematol. Oncol. 2015, 8, 40. [Google Scholar] [CrossRef]
- Yoon, Y.J.; Kim, J.A.; Shin, K.D.; Shin, D.S.; Han, Y.M.; Lee, Y.J.; Lee, J.S.; Kwon, B.M.; Han, D.C. KRIBB11 inhibits HSP70 synthesis through inhibition of heat shock factor 1 function by impairing the recruitment of positive transcription elongation factor b to the hsp70 promoter. J. Biol. Chem. 2011, 286, 1737–1747. [Google Scholar] [CrossRef]
- Wales, C.T.; Taylor, F.R.; Higa, A.T.; McAllister, H.A.; Jacobs, A.T. ERK-dependent phosphorylation of HSF1 mediates chemotherapeutic resistance to benzimidazole carbamates in colorectal cancer cells. Anticancer Drugs 2015, 26, 657–666. [Google Scholar] [CrossRef]
- Antonietti, P.; Linder, B.; Hehlgans, S.; Mildenberger, I.C.; Burger, M.C.; Fulda, S.; Steinbach, J.P.; Gessler, F.; Rödel, F.; Mittelbronn, M.; et al. Interference with the HSF1/HSP70/BAG3 Pathway Primes Glioma Cells to Matrix Detachment and BH3 Mimetic-Induced Apoptosis. Mol. Cancer Ther. 2017, 16, 156–168. [Google Scholar] [CrossRef]
- Kang, M.J.; Yun, H.H.; Lee, J.H. KRIBB11 accelerates Mcl-1 degradation through an HSF1-independent, Mule-dependent pathway in A549 non-small cell lung cancer cells. Biochem. Biophys. Res. Commun. 2017, 492, 304–309. [Google Scholar] [CrossRef]
- Chen, K.; Qian, W.; Li, J.; Jiang, Z.; Cheng, L.; Yan, B.; Cao, J.; Sun, L.; Zhou, C.; Lei, M.; et al. Loss of AMPK activation promotes the invasion and metastasis of pancreatic cancer through an HSF1-dependent pathway. Mol. Oncol. 2017, 11, 1475–1492. [Google Scholar] [CrossRef]
- Dai, S.; Tang, Z.; Cao, J.; Zhou, W.; Li, H.; Sampson, S.; Dai, C. Suppression of the HSF1-mediated proteotoxic stress response by the metabolic stress sensor AMPK. EMBO J. 2015, 34, 275–293. [Google Scholar] [CrossRef]
- Schaefer, S.; Svenstrup, T.H.; Guerra, B. The small-molecule kinase inhibitor D11 counteracts 17-AAG-mediated up-regulation of HSP70 in brain cancer cells. PLoS ONE 2017, 12, e0177706. [Google Scholar] [CrossRef] [PubMed]
- Kühnel, A.; Schilling, D.; Combs, S.E.; Haller, B.; Schwab, M.; Multhoff, G. Radiosensitization of HSF-1 Knockdown Lung Cancer Cells by Low Concentrations of Hsp90 Inhibitor NVP-AUY922. Cells 2019, 8, 1166. [Google Scholar] [CrossRef] [PubMed]
- Mijatovic, T.; Mathieu, V.; Gaussin, J.F.; De Nève, N.; Ribaucour, F.; Van Quaquebeke, E.; Dumont, P.; Darro, F.; Kiss, R. Cardenolide-induced lysosomal membrane permeabilization demonstrates therapeutic benefits in experimental human non-small cell lung cancers. Neoplasia 2006, 8, 402–412. [Google Scholar] [CrossRef] [PubMed]
- Chiosis, G.; Vilenchik, M.; Kim, J.; Solit, D. Hsp90: The vulnerable chaperone. Drug Disc. Today 2004, 9, 881–888. [Google Scholar] [CrossRef]
- Sreedhar, A.S.; Soti, C.; Csermely, P. Inhibition of Hsp90: A new strategy for inhibiting protein kinases. Biochim. Biophys. Acta 2004, 1697, 233–242. [Google Scholar] [CrossRef]
- Zhang, H.; Burrows, F. Targeting multiple signal transduction pathways through inhibition of Hsp90. J. Mol. Med. 2004, 82, 488–499. [Google Scholar] [CrossRef]
- Neckers, L. Hsp90 inhibitors as novel cancer chemotherapeutic agents. Trends Mol. Med. 2002, 8, S55–S61. [Google Scholar] [CrossRef]
- Picard, D. Heat-shock protein 90, a chaperone for folding and regulation. Cell Mol. Life Sci. 2002, 59, 1640–1648. [Google Scholar] [CrossRef]
- Richter, K.; Buchner, J. Hsp90: Chaperoning signal transduction. J. Cell Physiol. 2001, 188, 281–290. [Google Scholar] [CrossRef]
- Tatokoro, M.; Koga, F.; Yoshida, S.; Kihara, K. Heat shock protein 90 targeting therapy: State of the art and future perspective. EXCLI J. 2015, 14, 48–58. [Google Scholar]
- Whitesell, L.; Lindquist, S.L. HSP90 and the chaperoning of cancer. Nat. Rev. Cancer 2005, 5, 761–772. [Google Scholar] [CrossRef] [PubMed]
- Whitesell, L.; Santagata, S.; Lin, N.U. Inhibiting HSP90 to treat cancer: A strategy in evolution. Curr. Mol. Med. 2012, 12, 1108–1124. [Google Scholar] [CrossRef] [PubMed]
- Marcu, M.G.; Chadli, A.; Bouhouche, I.; Catelli, M.; Neckers, L.M. The heat shock protein 90 antagonist novobiocin interacts with a previously unrecognized ATP-binding domain in the carboxyl terminus of the chaperone. J. Biol. Chem. 2000, 275, 37181–37186. [Google Scholar] [CrossRef] [PubMed]
- McConkey, D.J.; Zhu, K. Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist. Updates 2008, 11, 164–179. [Google Scholar] [CrossRef]
- Burlison, J.A.; Blagg, B.S. Synthesis and evaluation of coumermycin A1 analogues that inhibit the Hsp90 protein folding machinery. Org. Lett. 2006, 8, 4855–4858. [Google Scholar] [CrossRef]
- Yin, Z.; Henry, E.C.; Gasiewicz, T.A. (-)-Epigallocatechin-3-gallate is a novel Hsp90 inhibitor. Biochemistry 2009, 48, 336–345. [Google Scholar] [CrossRef]
- Roe, S.M.; Prodromou, C.; O’Brien, R.; Ladbury, J.E.; Piper, P.W.; Pearl, L.H. Structural basis for inhibition of the Hsp90 molecular chaperone by the antitumor antibiotics radicicol and geldanamycin. J. Med. Chem. 1999, 42, 260–266. [Google Scholar] [CrossRef]
- Schulte, T.W.; Akinaga, S.; Soga, S.; Sullivan, W.; Stensgard, B.; Toft, D.; Neckers, L.M. Antibiotic radicicol binds to the N-terminal domain of Hsp90 and shares important biological activities with geldanamycin. Cell Stress Chaperones 1998, 3, 100–108. [Google Scholar] [CrossRef]
- Supko, J.G.; Hickman, R.L.; Grever, M.R.; Malspeis, L. Preclinical pharmacologic evaluation of geldanamycin as an antitumor agent. Cancer Chemother. Pharmacol. 1995, 36, 305–315. [Google Scholar] [CrossRef]
- Dikalov, S.; Landmesser, U.; Harrison, D.G. Geldanamycin leads to superoxide formation by enzymatic and non-enzymatic cycling. J. Biol. Chem. 2002, 277, 25480–25485. [Google Scholar] [CrossRef]
- Dikalov, S.; Rumyantseva, G.V.; Piskunov, A.V.; Weiner, L.M. Role of Quinone-iron (III) interaction in NADPH-dependent enzymatic generation of hydroxyl radicals. Biochemistry 1992, 31, 8947–8953. [Google Scholar] [CrossRef] [PubMed]
- Kamal, A.; Thao, L.; Sensintaffar, J.; Zhang, L.; Boehm, M.F.; Fritz, L.C.; Burrows, F.J. A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors. Nature 2003, 425, 407–410. [Google Scholar] [CrossRef] [PubMed]
- Ge, J.; Normant, E.; Porter, J.R.; Ali, J.A.; Dembski, M.S.; Gao, Y.; Georges, A.T.; Grenier, L.; Pak, R.H.; Patterson, J.; et al. Design, synthesis, and biological evaluation of hydroquinone derivatives of 17-amino-17-demethoxygeldanamycin as potent, water-soluble inhibitors of Hsp90. J. Med. Chem. 2006, 49, 4606–4615. [Google Scholar] [CrossRef] [PubMed]
- Taldone, T.; Gozman, A.; Maharaj, R.; Chiosis, G. Targeting Hsp90: Small-molecule inhibitors and their clinical development. Curr. Opin. Pharmacol. 2008, 8, 370–374. [Google Scholar] [CrossRef] [PubMed]
- Gartner, E.M.; Silverman, P.; Simon, M.; Flaherty, L.; Abrams, J.; Ivy, P.; Lorusso, P.M. A phase II study of 17-allylamino-17-demethoxygeldanamycin in metastatic or locally advanced, unresectable breast cancer. Breast Cancer Res. Treat. 2012, 131, 933–937. [Google Scholar] [CrossRef]
- 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]
- Richardson, P.G.; Badros, A.Z.; Jagannath, S.; Tarantolo, S.; Wolf, J.L.; Albitar, M.; Berman, D.; Messina, M.; Anderson, K.C. Tanespimycin with bortezomib: Activity in relapsed/refractory patients with multiple myeloma. Br. J. Haematol. 2010, 150, 428–437. [Google Scholar] [CrossRef]
- Ronnen, E.A.; Kondagunta, G.V.; Ishill, N.; Sweeney, S.M.; Deluca, J.K.; Schwartz, L.; Bacik, J.; Motzer, R.J. A phase II trial of 17-(Allylamino)-17-demethoxygeldanamycin in patients with papillary and clear cell renal cell carcinoma. Investig. New Drugs 2006, 24, 543–546. [Google Scholar] [CrossRef]
- Heath, E.I.; Hillman, D.W.; Vaishampayan, U.; Sheng, S.; Sarkar, F.; Harper, F.; Gaskins, M.; Pitot, H.C.; Tan, W.; Ivy, S.P. A phase II trial of 17-allylamino-17-demethoxygeldanamycin in patients with hormone-refractory metastatic prostate cancer. Clin. Cancer Res. 2008, 14, 7940–7946. [Google Scholar] [CrossRef]
- Pacey, S.; Gore, M.; Chao, D.; Banerji, U.; Larkin, J.; Sarker, S.; Owen, K.; Asad, Y.; Raynaud, F.; Walton, M.; et al. A Phase II trial of 17-allylamino, 17-demethoxygeldanamycin (17-AAG, tanespimycin) in patients with metastatic melanoma. Investig. New Drugs 2012, 30, 341–349. [Google Scholar] [CrossRef]
- Hong, D.S.; Banerji, U.; Tavana, B.; George, G.C.; Aaron, J.; Kurzrock, R. Targeting the molecular chaperone heat shock protein 90 (HSP90): Lessons learned and future directions. Cancer Treat. Rev. 2013, 39, 375–387. [Google Scholar] [CrossRef]
- Castro, J.E.; Prada, C.E.; Loria, O.; Kamal, A.; Chen, L.; Burrows, F.J.; Kipps, T.J. ZAP-70 is a novel conditional heat shock protein 90 (Hsp90) client: Inhibition of Hsp90 leads to ZAP-70 degradation, apoptosis, and impaired signaling in chronic lymphocytic leukemia. Blood 2005, 106, 2506–2512. [Google Scholar] [CrossRef] [PubMed]
- Modi, S.; Stopeck, A.; Linden, H.; Solit, D.; Chandarlapaty, S.; Rosen, N.; D’Andrea, G.; Dickler, M.; Moynahan, M.E.; Sugarman, S.; et al. HSP90 inhibition is effective in breast cancer: A phase II trial of tanespimycin (17-AAG) plus trastuzumab in patients with HER2-positive metastatic breast cancer progressing on trastuzumab. Clin. Cancer Res. 2011, 17, 5132–5139. [Google Scholar] [CrossRef] [PubMed]
- Mbofung, R.M.; McKenzie, J.A.; Malu, S.; Zhang, M.; Peng, W.; Liu, C.; Kuiatse, I.; Tieu, T.; Williams, L.; Devi, S.; et al. HSP90 inhibition enhances cancer immunotherapy by upregulating interferon response genes. Nat. Commun. 2017, 8, 451. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Smith, D.L.; Sequeira, M.; Sang, J.; Bates, R.C.; Proia, D.A. The HSP90 inhibitor ganetespib has chemosensitizer and radiosensitizer activity in colorectal cancer. Investig. New Drugs 2014, 32, 577–586. [Google Scholar] [CrossRef] [Green Version]
- Nagaraju, G.P.; Alese, O.B.; Landry, J.; Diaz, R.; El-Rayes, B.F. HSP90 inhibition downregulates thymidylate synthase and sensitizes colorectal cancer cell lines to the effect of 5FU-based chemotherapy. Oncotarget 2014, 5, 9980–9991. [Google Scholar] [CrossRef]
- Bendell, J.C.; Jones, S.F.; Hart, L.; Pant, S.; Moyhuddin, A.; Lane, C.M.; Earwood, C.; Murphy, P.; Patton, J.; Penley, W.C.; et al. A Phase I Study of the Hsp90 Inhibitor AUY922 plus Capecitabine for the Treatment of Patients with Advanced Solid Tumors. Cancer Investig. 2015, 33, 477–482. [Google Scholar] [CrossRef]
- Camphausen, K.; Tofilon, P.J. Inhibition of Hsp90: A multitarget approach to radiosensitization. Clin. Cancer Res. 2007, 13, 4326–4330. [Google Scholar] [CrossRef]
- Kinzel, L.; Ernst, A.; Orth, M.; Albrecht, V.; Hennel, R.; Brix, N.; Frey, B.; Gaipl, U.S.; Zuchtriegel, G.; Reichel, C.A.; et al. A novel HSP90 inhibitor with reduced hepatotoxicity synergizes with radiotherapy to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer. Oncotarget 2016, 7, 43199–43219. [Google Scholar] [CrossRef]
- Vaishampayan, U.N.; Burger, A.M.; Sausville, E.A.; Heilbrun, L.K.; Li, J.; Horiba, M.N.; Egorin, M.J.; Ivy, P.; Pacey, S.; Lorusso, P.M. Safety, efficacy, pharmacokinetics, and pharmacodynamics of the combination of sorafenib and tanespimycin. Clin. Cancer Res. 2010, 16, 3795–3804. [Google Scholar] [CrossRef]
- Meehan, R.; Kummar, S.; Do, K.; O’Sullivan Coyne, G.; Juwara, L.; Zlott, J.; Rubinstein, L.; Doroshow, J.H.; Chen, A.P. A phase I study of Ganetespib and Ziv-Aflibercept in patients with advanced carcinomas and sarcomas. Oncologist 2018, 23, 1269-e125. [Google Scholar] [CrossRef] [PubMed]
- Kawabe, M.; Mandic, M.; Taylor, J.L.; Vasquez, C.A.; Wesa, A.K.; Neckers, L.M.; Storkus, W.J. Heat shock protein 90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin enhances EphA2+ tumor cell recognition by specific CD8+ T cells. Cancer Res. 2009, 69, 6995–7003. [Google Scholar] [CrossRef] [PubMed]
- Maloney, A.; Workman, P. Hsp90 as a new therapeutic target for cancer therapy: The Story Unfolds. Expert Opin. Biol. Ther. 2002, 2, 3–24. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Street, T.O. 5’-N-ethylcarboxamidoadenosine is not a paralog-specific Hsp90 inhibitor. Protein Sci. 2016, 25, 2209–2215. [Google Scholar] [CrossRef]
- Mogk, A.; Bukau, B.; Kampinga, H.H. Cellular handling of protein aggregates by disaggregation machines. Mol. Cell. 2018, 69, 214–226. [Google Scholar] [CrossRef]
- Finka, A.; Sharma, S.K.; Goloubinoff, P. Multi-layered molecular mechanisms of polypeptide holding, unfolding and disaggregation by HSP70/HSP110 chaperones. Front. Mol. Biosci. 2015, 2, 29. [Google Scholar] [CrossRef] [Green Version]
- Calderwood, S.K.; Gong, J. Heat shock proteins promote cancer: It’s a protection racket. Trends Biochem. Sci. 2016, 41, 311–323. [Google Scholar] [CrossRef]
- Kabbage, M.; Dickman, M.B. The BAG proteins: A ubiquitous family of chaperone regulators. Cell Mol. Life Sci. 2008, 65, 1390–1402. [Google Scholar] [CrossRef]
- Gamerdinger, M.; Hajieva, P.; Kaya, A.M.; Wolfrum, U.; Hartl, F.U.; Behl, C. Protein quality control during aging involves recruitment of the macroautophagy pathway by BAG3. EMBO J. 2009, 28, 889–901. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Colvin, T.; Rauch, J.N.; Acosta-Alvear, D.; Kampmann, M.; Dunyak, B.; Hann, B.; Aftab, B.T.; Murnane, M.; Cho, M.; et al. Validation of the Hsp70-Bag3 protein-protein interaction as a potential therapeutic target in cancer. Mol. Cancer Ther. 2015, 14, 642–648. [Google Scholar] [CrossRef]
- Komarova, E.Y.; Afanasyeva, E.A.; Bulatova, M.M.; Cheetham, M.E.; Margulis, B.A.; Guzhova, I.V. Downstream caspases are novel targets for the antiapoptotic activity of the molecular chaperone hsp70. Cell Stress Chaperones 2004, 9, 265–275. [Google Scholar] [CrossRef] [PubMed]
- Budina-Kolomets, A.; Webster, M.R.; Leu, J.I.; Jennis, M.; Krepler, C.; Guerrini, A.; Kossenkov, A.V.; Xu, W.; Karakousis, G.; Schuchter, L.; et al. HSP70 inhibition limits FAK-dependent invasion and enhances the response to melanoma treatment with BRAF inhibitors. Cancer Res. 2016, 76, 2720–2730. [Google Scholar] [CrossRef] [PubMed]
- Joshi, S.; Wang, T.; Araujo, T.L.S.; Sharma, S.; Brodsky, J.L.; Chiosis, G. Adapting to stress—Chaperome networks in cancer. Nat. Rev. Cancer. 2018, 18, 562–575. [Google Scholar] [CrossRef] [PubMed]
- Gestwicki, J.E.; Shao, H. Inhibitors and chemical probes for molecular chaperone networks. J. Biol. Chem. 2019, 294, 2151–2161. [Google Scholar] [CrossRef] [Green Version]
- Budina-Kolomets, A.; Balaburski, G.M.; Bondar, A.; Beeharry, N.; Yen, T.; Murphy, M.E. Comparison of the activity of three different HSP70 inhibitors on apoptosis, cell cycle arrest, autophagy inhibition, and HSP90 inhibition. Cancer Biol. Ther. 2014, 15, 194–199. [Google Scholar] [CrossRef]
- Rodina, A.; Taldone, T.; Kang, Y.; Patel, P.D.; Koren, J., 3rd; Yan, P.; DaGama Gomes, E.M.; Yang, C.; Patel, M.R.; Shrestha, L.; et al. Affinity purification probes of potential use to investigate the endogenous Hsp70 interactome in cancer. ACS Chem. Biol. 2014, 9, 1698–1705. [Google Scholar] [CrossRef]
- Zeng, Y.; Cao, R.; Zhang, T.; Li, S.; Zhong, W. Design and synthesis of piperidine derivatives as novel human heat shock protein 70 inhibitors for the treatment of drug-resistant tumors. Eur. J. Med. Chem. 2015, 97, 19–31. [Google Scholar] [CrossRef] [Green Version]
- Cheeseman, M.D.; Westwood, I.M.; Barbeau, O.; Rowlands, M.; Dobson, S.; Jones, A.M.; Jeganathan, F.; Burke, R.; Kadi, N.; Workman, P.; et al. Exploiting protein conformational change to optimize adenosine-derived inhibitors of HSP70. J. Med. Chem. 2016, 59, 4625–4636. [Google Scholar] [CrossRef]
- Lazarev, V.F.; Sverchinsky, D.V.; Mikhaylova, E.R.; Semenyuk, P.I.; Komarova, E.Y.; Niskanen, S.A.; Nikotina, A.D.; Burakov, A.V.; Kartsev, V.G.; Guzhova, I.V.; et al. Sensitizing tumor cells to conventional drugs: HSP70 chaperone inhibitors, their selection and application in cancer models. Cell Death Dis. 2018, 9, 41. [Google Scholar] [CrossRef]
- Chang, L.; Miyata, Y.; Ung, P.M.; Bertelsen, E.B.; McQuade, T.J.; Carlson, H.A.; Zuiderweg, E.R.; Gestwicki, J.E. Chemical screens against a reconstituted multiprotein complex: Myricetin blocks DnaJ regulation of DnaK through an allosteric mechanism. Chem. Biol. 2011, 18, 210–221. [Google Scholar] [CrossRef]
- Adam, C.; Baeurle, A.; Brodsky, J.L.; Wipf, P.; Schrama, D.; Becker, J.C.; Houben, R. The HSP70 modulator MAL3-101 inhibits Merkel cell carcinoma. PLoS ONE 2014, 9, e92041. [Google Scholar] [CrossRef] [PubMed]
- Enthammer, M.; Papadakis, E.S.; Salomé Gachet, M.; Deutsch, M.; Schwaiger, S.; Koziel, K.; Ashraf, M.I.; Khalid, S.; Wolber, G.; Packham, G.; et al. Isolation of a novel thioflavin S-derived compound that inhibits BAG-1-mediated protein interactions and targets BRAF inhibitor-resistant cell lines. Mol. Cancer Ther. 2013, 12, 2400–2414. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Srinivasan, S.R.; Connarn, J.; Ahmad, A.; Young, Z.T.; Kabza, A.M.; Zuiderweg, E.R.; Sun, D.; Gestwicki, J.E. Analogs of the allosteric heat shock protein 70 (Hsp70) inhibitor, MKT-077, as anti-cancer agents. ACS Med. Chem. Lett. 2013, 4. [Google Scholar] [CrossRef]
- Sverchinsky, D.V.; Lazarev, V.F.; Semenyuk, P.I.; Mitkevich, V.A.; Guzhova, I.V.; Margulis, B.A. Peptide fragments of Hsp70 modulate its chaperone activity and sensitize tumor cells to anti-cancer drugs. FEBS Lett. 2017, 591, 4074–4082. [Google Scholar] [CrossRef] [Green Version]
- Cao, X.; Zhou, Y.; Sun, H.; Xu, M.; Bi, X.; Zhao, Z.; Shen, B.; Wan, F.; Hong, Z.; Lan, L.; 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 Lett. 2018, 424, 84–96. [Google Scholar] [CrossRef]
- Sekihara, K.; Harashima, N.; Tongu, M.; Tamaki, Y.; Uchida, N.; Inomata, T.; Harada, M. Pifithrin-μ, an inhibitor of heat-shock protein 70, can increase the antitumor effects of hyperthermia against human prostate cancer cells. PLoS ONE 2013, 8, e78772. [Google Scholar] [CrossRef]
- Court, K.A.; Hatakeyama, H.; Wu, S.Y.; Lingegowda, M.S.; Rodríguez-Aguayo, C.; López-Berestein, G.; Ju-Seog, L.; Rinaldi, C.; Juan, E.J.; Sood, A.K.; et al. HSP70 inhibition synergistically enhances the effects of magnetic fluid hyperthermia in ovarian cancer. Mol. Cancer Ther. 2017, 16, 966–976. [Google Scholar] [CrossRef]
- Sannino, S.; Guerriero, C.J.; Sabnis, A.J.; Stolz, D.B.; Wallace, C.T.; Wipf, P.; Watkins, S.C.; Bivona, T.G.; Brodsky, J.L. Compensatory increases of select proteostasis networks after Hsp70 inhibition in cancer cells. J. Cell Sci. 2018, 131, jcs217760. [Google Scholar] [CrossRef]
- Yaglom, J.A.; Wang, Y.; Li, A.; Li, Z.; Monti, S.; Alexandrov, I.; Lu, X.; Sherman, M.Y. Cancer cell responses to Hsp70 inhibitor JG-98: Comparison with Hsp90 inhibitors and finding synergistic drug combinations. Sci. Rep. 2018, 8, 3010. [Google Scholar] [CrossRef]
- Ishaq, M.; Ojha, R.; Sharma, K.; Sharma, G.; Singh, S.K.; Majumdar, S. Functional inhibition of Hsp70 by Pifithrin-μ switches Gambogic acid induced caspase dependent cell death to caspase independent cell death in human bladder cancer cells. Biochim. Biophys. Acta 2016, 1863, 2560–2573. [Google Scholar] [CrossRef]
- Li, X.; Shao, H.; Taylor, I.R.; Gestwicki, J.E. Targeting allosteric control mechanisms in heat shock protein 70 (Hsp70). Curr. Top Med. Chem. 2016, 16, 2729–2740. [Google Scholar] [CrossRef] [PubMed]
- Schilling, D.; Garrido, C.; Combs, S.E.; Multhoff, G. The Hsp70 inhibiting peptide aptamer A17 potentiates radiosensitization of tumor cells by Hsp90 inhibition. Cancer Lett. 2017, 390, 146–152. [Google Scholar] [CrossRef] [PubMed]
- Multhoff, G.; Botzler, C.; Jennen, L.; Schmidt, J.; Ellwart, J.; Issels, R. Heat shock protein 72 on tumor cells: A recognition structure for natural killer cells. J. Immunol. 1997, 158, 4341–4350. [Google Scholar]
- Shevtsov, M.; Multhoff, G. Heat Shock Protein-Peptide and HSP-Based Immunotherapies for the Treatment of Cancer. Front. Immunol. 2016, 7, e171. [Google Scholar] [CrossRef] [PubMed]
- Shevtsov, M.; Huile, G.; Multhoff, G. Membrane heat shock protein 70: A theranostic target for cancer therapy. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018, 373, 20160526. [Google Scholar] [CrossRef]
- Shevtsov, M.; Stangl, S.; Nikolaev, B.; Yakovleva, L.; Marchenko, Y.; Tagaeva, R.; Sievert, W.; Pitkin, E.; Mazur, A.; Tolstoy, P.; et al. Granzyme B Functionalized Nanoparticles Targeting Membrane Hsp70-Positive Tumors for Multimodal Cancer Theranostics. Small 2019, 15, e1900205. [Google Scholar] [CrossRef]
- Multhoff, G.; Pfister, K.; Gehrmann, M.; Hantschel, M.; Gross, C.; Hafner, M.; Hiddemann, W. A 14-mer Hsp70 peptide stimulates natural killer (NK) cell activity. Cell Stress Chaperones 2001, 6, 337–344. [Google Scholar] [CrossRef]
- Krause, S.W.; Gastpar, R.; Andreesen, R.; Gross, C.; Ullrich, H.; Thonigs, G.; Pfister, K.; Multhoff, G. 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. Clin. Cancer Res. 2004, 10, 3699–3707. [Google Scholar] [CrossRef]
- Specht, H.M.; Ahrens, N.; Blankenstein, C.; Duell, T.; Fietkau, R.; Gaipl, U.S.; Günther, C.; Gunther, S.; Habl, G.; Hautmann, H.; et al. Heat Shock Protein 70 (Hsp70) Peptide Activated Natural Killer (NK) Cells for the Treatment of Patients with Non-Small Cell Lung Cancer (NSCLC) after Radiochemotherapy (RCTx)—From Preclinical Studies to a Clinical Phase II Trial. Front. Immunol. 2015, 6, e162. [Google Scholar] [CrossRef]
- Shevtsov, M.; Pitkin, E.; Ischenko, A.; Stangl, S.; Khachatryan, W.; Galibin, O.; Edmond, S.; Lobinger, D.; Multhoff, G. Ex vivo Hsp70-Activated NK Cells in Combination With PD-1 Inhibition Significantly Increase Overall Survival in Preclinical Models of Glioblastoma and Lung Cancer. Front. Immunol. 2019, 10, e454. [Google Scholar] [CrossRef]
- Kokowski, K.; Stangl, S.; Seier, S.; Hildebrandt, M.; Vaupel, P.; Multhoff, G. Radiochemotherapy combined with NK cell transfer followed by second-line PD-1 inhibition in a patient with NSCLC stage IIIb inducing long-term tumor control: A case study. Strahlenther. Onkol. 2019, 195, 352–361. [Google Scholar] [CrossRef] [PubMed]
- Zoubeidi, A.; Gleave, M. Small heat shock proteins in cancer therapy and prognosis. Int. J. Biochem. Cell Biol. 2012, 44, 1646–1656. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Yu, E.Y.; Ellard, S.L.; Hotte, S.J.; Gingerich, J.R.; Joshua, A.M.; Gleave, M.E.; Chi, K.N. A randomized phase 2 study of a HSP27 targeting antisense, apatorsen with prednisone versus prednisone alone, in patients with metastatic castration resistant prostate cancer. Investig. New Drugs 2018, 36, 278–287. [Google Scholar] [CrossRef] [PubMed]
- Ko, A.H.; Murphy, P.B.; Peyton, J.D.; Shipley, D.L.; Al-Hazzouri, A.; Rodriguez, F.A.; Womack, M.S., 4th; 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]
Therapeutic Factors 1 | Factor 2 | Cancer Type | Outcome | Reference |
---|---|---|---|---|
AC220 quizartinib (FLT3 TK inhibitor) | TAK-165 HER2 inhibitor | Variety of human tumors | Cytotoxicity, autophagy | Ouchida et al., 2018 [11] |
Vemurafenib, inhibitor of mutant BRAF | PAC-1 pro-caspase activator | Human melanoma in vitro & in vivo | Effect in caspase-3 activation, inhibition of tumor regrowth | Peh et al., 2016 [12] |
Metformin AMPK activator | Cisplatin | Skov-3 and HEY ovarian cancer in vitro & in vivo | Inhibition of T and Smad-Smad3 phosphorylation GFβ1 expression | Zheng et al., 2018 [13] |
Cisplatin packed together in amphiphilic PLG-g | Do Ve/PEG graft copolymer cetaxel | B16F1 cells & graft | Anti-metastasis effect and prolonged circulation | Song et al., 2014 [14] |
Cetuximab anti-ErbB/HER monoclonal antibody | Erlotinib tyrosine-kinase inhibitor | NSCLC non-small-cell lung cancer | Antibody-dependent, NK mediated cytotoxicity | Cavazzoni et al., 2012 [15] |
Anti-KRAS antibody | Gemcitabine | Pancreatic cancer in vitro & in vivo | Inhibition RAS signaling | Kang et al., 2018 [16] |
Bevacizumab CTLA4 blockade | Ipilimumab VEGF inhibition | Patients with metastatic melanoma | Survival up to 25 months, immune response | Hodi et al., 2014 [17] |
Trametinib and dabrafenib | Anti-PD1 antibody | BRAF(V600E) melanoma | Anti-tumor effect in vivo, reduction of metastasis | Hu-Lieskovan et al., 2015 [18] |
Dabrafenib or trametinib BRAF & MEK inhibitors | Anti-PD-1, PD-L1, and CTLA-4 (checkpoints) antibodies | Carcinoma in vitro & in vivo | Anti-tumor immune response | Liu et al., 2015 [19] |
Physical exercise or dihydroartemisinin (inducer of oxidative stress) | Temozolomide | Glioblastoma in vitro & in vivo | Reduced clonogenicity/migration, lowered metastasis | Lemke et al., 2016 [20] |
Oncolytic virus both encapsulated in extracellular vesicles | Paclitaxel | Lung cancer | Anti-tumor effect in vivo | Garofalo et al., 2018 [21] |
Inhibitor | Concurrent Therapy | Cancer Type | Outcome | Reference |
---|---|---|---|---|
HSF1 inhibitors | ||||
Triptolide | Doxorubicin | MCF-7 and MDA-MB-468 human breast cancer | Inhibition of tumor growth and enhancement of anti-tumor effects of doxorubicin | Xiong et al., 2016 [31] |
Triptolide | Curcumin | Ovarian cancer | Tumor inhibition rate of 68.78% | Liu et al., 2018 [32] |
KRIBB11 | Akt small molecule inhibitor MK-2206 | Breast cancer | Synergistic killing of breast cancer cells and breast cancer stem cells; inhibition of tumor growth | Carpenter et al., 2017 [33] |
Cardenolide CL-43 | Cisplatin/etoposide/doxorubicin | HCT-116 human colon carcinoma | Additive anti-tumor effect | Nikotina et al., 2018 [34] |
HSP90 inhibitors | ||||
Tanespimycin (17-AAG) | Trastuzumab | HER2-positive metastatic breast cancer progressing on trastuzumab | Significant anticancer activity | Modi et al., 2011 [35] |
Ganetespib (STA-9090) | BRAF(V600E) inhibitor vemurafenib/MEK inhibitor TAK-733 | Melanoma | Tumor regression in vemurafenib-resistant xenografts | Acquaviva et al., 2014 [36] |
Ganetespib | Anti-PD-L1 antibody STI-A1015 | MC38 colon carcinoma and B16 melanoma | Enhanced anti-tumor efficacy of the combinatorial regimen | Proia et al., 2015 [37] |
HSP70 inhibitors | ||||
VER-155008 | 17-AAD inhibitor of HSP90 | NSCLC cells | Synergistic effect on NSCLC cells proliferation | Wen et al., 2014 [38] |
VER-155008/MAL3-101 | STA-9090 inhibitor of HSP90 | Muscle invasive bladder cancer (MIBC) cells | Synergistic anti-tumor effect | Prince et al., 2018 [39] |
VER-155008 | Radicicol inhibitor of HSP90 | Anaplastic thyroid carcinoma cells | Enhanced anti-tumor activity of combinatorial therapy | Kim et al., 2014 [40] |
Pifithrin-μ | Cisplatin/Oxaliplatin | HT29 colorectal and PC-3 prostate cancer cells | Synergistic anti-tumor effect | McKeon et al., 2016 [41] |
HSP27 inhibitors | ||||
Apartorsen | Docetaxel | Platinum-resistant metastatic urothelial carcinoma | Improved OS compared to docetaxel alone | Rosenberg et al., 2018 [42] |
OGX-427 | Autophagy inhibitor chloroquine | PC-3 prostate cancer | Inhibition of tumor progression in vivo | Kumano et al., 2012 [43] |
OGX-427 | HSP90 inhibitor PF-04929113 | Castrate-resistant prostate cancer | OGX-427 synergistically enhanced anti-tumor effect of HSP90 inhibitor | Lamoureux et al., 2014 [44] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Shevtsov, M.; Multhoff, G.; Mikhaylova, E.; Shibata, A.; Guzhova, I.; Margulis, B. Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors. Int. J. Mol. Sci. 2019, 20, 5284. https://doi.org/10.3390/ijms20215284
Shevtsov M, Multhoff G, Mikhaylova E, Shibata A, Guzhova I, Margulis B. Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors. International Journal of Molecular Sciences. 2019; 20(21):5284. https://doi.org/10.3390/ijms20215284
Chicago/Turabian StyleShevtsov, Maxim, Gabriele Multhoff, Elena Mikhaylova, Atsushi Shibata, Irina Guzhova, and Boris Margulis. 2019. "Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors" International Journal of Molecular Sciences 20, no. 21: 5284. https://doi.org/10.3390/ijms20215284
APA StyleShevtsov, M., Multhoff, G., Mikhaylova, E., Shibata, A., Guzhova, I., & Margulis, B. (2019). Combination of Anti-Cancer Drugs with Molecular Chaperone Inhibitors. International Journal of Molecular Sciences, 20(21), 5284. https://doi.org/10.3390/ijms20215284