Skip to main content
Neoplasia (New York, N.Y.) logoLink to Neoplasia (New York, N.Y.)
. 2021 Nov 12;23(12):1261–1274. doi: 10.1016/j.neo.2021.11.006

Overcoming prostate cancer drug resistance with a novel organosilicon small molecule

Rui Zhao a,b, Xiaowei Ma b,c, Lijuan Bai b,d, Xin Li b,e, Kenza Mamouni b, Yang Yang b,f, HongYan Liu g, Alira Danaher e, Nicholas Cook e, Omer Kucuk h,i, Robert S Hodges j,k, Lajos Gera j,k, Daqing Wu b,e,i,l,
PMCID: PMC8604682  PMID: 34781084

Abstract

A major challenge to the treatment of advanced prostate cancer (PCa) is the development of resistance to androgen-deprivation therapy (ADT) and chemotherapy. It is imperative to discover effective therapies to overcome drug resistance and improve clinical outcomes. We have developed a novel class of silicon-containing compounds and evaluated the anticancer activities and mechanism of action using cellular and animal models of drug-resistant PCa. Five organosilicon compounds were evaluated for their anticancer activities in the NCI-60 panel and established drug-resistant PCa cell lines. GH1504 exhibited potent in vitro cytotoxicity in a broad spectrum of human cancer cells, including PCa cells refractory to ADT and chemotherapy. Molecular studies identified several potential targets of GH1504, most notably androgen receptor (AR), AR variant 7 (AR-v7) and survivin. Mechanistically, GH1504 may promote the protein turnover of AR, AR-v7 and survivin, thereby inducing apoptosis in ADT-resistant and chemoresistant PCa cells. Animal studies demonstrated that GH1504 effectively inhibited the in vivo growth of ADT-resistant CWR22Rv1 and chemoresistant C4-2B-TaxR xenografts in subcutaneous and intraosseous models. These preclinical results indicated that GH1504 is a promising lead that can be further developed as a novel therapy for drug-resistant PCa.

Keywords: Prostate cancer, Castration-resistance, Chemoresistance, Small-molecule therapy, Silicon-containing compounds, Preclinical studies

Abbreviations: ADT, androgen deprivation therapy; AMDP(OEt)4, 1-aminomethylenedisphosphonic acid tetraethyl ester amide residue; AR, androgen receptor; AR-V7, AR variant 7; Atmp, 4-amino-2,2,6,6-tetramethylpiperidine amide residue; BmSimob, 4-[(butyldimethylsilyl)methoxy]-benzoyl; Bip, β-(4-biphenylyl)alanine residue; BOP, benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate; COX-2, cyclooxygenase-2; CRPC, castration-resistant prostate cancer; DIEA, N,N-diisopropylethylamine; GRP78, glucose-regulated protein 78kD; HSP27, heat shock protein 27; HSP90, heat shock protein 90; IGF-1R, insulin-like growth factor-1 receptor; IHC, immunohistochemistry; KIF15, Kinesin family member 15; LBD, ligand-binding domain; MSipob, 4-[3-(trimethylsilyl)propoxy]-benzoyl; NTD, N-terminal domain; OC2Y, O-2,6-dichlorobenzyl-tyrosine residue; PCa, prostate cancer; PROTAC, proteolysis-targeting chimaera; PSA, prostate-specific antigen; PyBOP, benzotriazol-1-yloxytripyrrolidinophophonium hexafluorophosphate; SIAH2, siah E3 ubiquitin protein ligase 2; TFA, trifluoroacetic acid

Introduction

Prostate cancer (PCa) is the most commonly diagnosed cancer and the second leading cause of cancer deaths in American men [1]. In 2021, an estimated 248,530 new cases are expected, and 34,130 patients will die [2]. Although most patients with localized PCa respond to standard treatments, including surgery, radiation therapy and first-generation androgen-deprivation therapy (ADT), in many cases PCa continues to progress towards castration resistance (CRPC) [3]. Docetaxel was approved in 2004 as a first-line chemotherapy for metastatic CRPC, although it only demonstrated a limited survival advantage [4]. Since 2010, the FDA has approved several new medications for the treatment of CRPC, mainly the new-generation chemotherapy (cabazitaxel) and ADT (abiraterone acetate, enzalutamide, apalutamide, and darolutamide) [5]. In recent Phase III clinical trials, such as STAMPEDE and CHAARTED, the combination of docetaxel and standard ADT (“chemohormonal” therapy) demonstrated prolonged overall survival compared to single chemotherapy or ADT in patients with advanced PCa [6,7]. Unfortunately, CRPC remains a dreadful evolution of PCa and eventually develops extreme resistance to ADT, chemotherapy, or both. Clearly, it is an urgent and unmet medical need to develop effective treatments to overcome therapeutic resistance and improve the survival and quality of life of CRPC patients.

At the molecular levels, CRPC is characterized by a significant rewiring of various oncogenic signaling pathways that allow cancer cells to evade their apoptotic fate and survive, despite drug treatments [8], [9], [10]. Research over the last decade has confirmed that the reactivation of the androgen receptor (AR) signaling is likely the most important molecular event during CRPC progression; this can be achieved via multiple mechanisms, including the amplification, mutation and alternative splicing of the AR gene [3,[11], [12], [13], [14]]. AR overexpression could be identified in up to 80% of CRPC specimens, whereas hormone-naïve tumors rarely exhibit AR amplification [13]. AR variant 7 (AR-v7), the best-characterized splicing variant so far, lacks the C-terminal ligand-binding domain (LBD) of full-length AR but retains an N-terminal domain (NTD); therefore it is capable of inducing androgen-independent AR transactivation [15]. Several liquid biopsy methods for AR-v7 detection have been established, and the occurrence and upregulation of AR-v7 in circulating tumor cells have been correlated to acquired resistance to abiraterone and enzalutamide in CRPC patients [16], [17], [18], [19], [20]. In preclinical models, both AR- and AR-v7-dependent transcriptional complementation are required for resistance to the second-generation ADT [21,22], and small-molecule inhibition of AR-v7 could sensitize CRPC cells to ADT treatment [23], [24], [25], [26], [27]. These observations indicated that the activation of AR-v7 expression and signaling might play an important role in ADT resistance, and AR-v7 represents a promising target to overcome therapeutic resistance [22].

Several mechanisms of chemoresistance have been proposed [8]. Rare subpopulations of cancer cells with stemness or neuroendocrine characteristics may be responsible for intrinsic chemoresistance [28], and overexpression of drug efflux pumps (such as the ATP-binding cassette transporter p-glycoprotein) may be pivotal in acquiring chemoresistance [29,30]. Deregulation of apoptotic pathways has been implicated in both chemoresistance and ADT resistance. Previous studies from us and others demonstrated that overexpression of survivin, a classic oncofetal gene that is differentially expressed in normal and cancer cells, promotes PCa progression and confers therapeutic resistance [31], [32], [33], [34].

The complex biology and high heterogeneity of advanced cancers indicated that a targeted therapy aimed at a single molecule or pathway would be less effective, and novel agents that can simultaneous inhibit/block multiple oncogenic signals (such as AR, AR-v7 and survivin) should be developed to overcome therapeutic resistance [35]. Molecular hybridization, a medicinal chemistry strategy that combines two or more pharmacophores into a single chemical entity, has been recognized as a promising approach to develop novel drug candidates with higher efficacy through multiple-target interaction [36,37]. During the past decade, we have designed several generations of small molecules that have a common three-component (“A-B-C”) structure consisting of three distinct pharmacophores, with the expectation of co-targeting multiple oncogenic factors and achieving better anticancer activities. Several compounds with biphenylalanine (Bip) or O-(2,6-dichlorobenzyl)-L-tyrosine (OC2Y) residues at the “B” position, including BKM570, BKM1644, BKM1740 and BKM1972, demonstrated promising efficacy in preclinical models of PCa and other cancer types [38], [39], [40], [41], [42], [43], [44], [45], [46], [47]. Recently, we reported that LG1836, a molecule containing an α-phenylcinnamic acid derivative as the “A” component and OC2Y as the “B” component, effectively inhibited the in vitro and in vivo growth of CRPC via the suppression of AR, AR-v7 and survivin [48]. These results warrant further efforts to develop more efficacious compounds that can be used to treat drug-resistant PCa.

Replacing a carbon atom with silicon, or the Sila-substitution (C/Si-exchange) of existing drugs, has been actively explored as an effective approach in the search for new biologically active small molecules [49], [50], [51], [52]. Carbon and silicon present some key differences in term of atomic size, bond length, electronegativity and lipophilicity. These differences can lead to changes in how the carbon and silicon analogues interact with their targets, thus resulting in different physico-chemistry and pharmacology between the two types of compounds [52]. For example, a silicon analogue is generally more lipophilic than its carbon equivalent, which may markedly increase tissue penetration and cellular uptake of the compound, thereby having improved in vivo stability and anticancer activity [53]. Bikzhanova et al. reported that the addition of a silicon to indomethacin, an anti-inflammatory drug, resulted in sila-indomethacin, or “silamethacin”. Compared with the parent compound, the silicon derivative retained its function as a selective cyclooxygenase-2 (COX-2) inhibitor while demonstrating enhanced potency in human cancer cells [54,55]. Similarly, the introduction of a (trimethylsilyl)ethyl group into camptothecin, a DNA topoisomerase inhibitor, significantly improved its anticancer activity in a broad spectrum of human cancer cells, including multi-drug resistant cells. Compared to camptothecins, the lipophilic silicon-containing Karenitecin (BNP1350) exhibited enhanced tissue penetration and bioavailability [56], and has entered clinical trials in melanoma, glioma and lung cancer. Inspired by the “C/Si-exchange” concept for rational drug design and built upon on our earlier generations of anticancer compounds, we developed a novel class of silicon-containing small molecules and determined their potential for the treatment of CRPC. In this report, we described one of such compounds, namely GH1504, that exhibited promising efficacy against both ADT-resistant and chemoresistant PCa in preclinical models.

Materials and methods

Chemical synthesis

Silicon-containing compounds with a three-component (“A-B-C”) structure were synthesized following the methods that we described previously [46,57]. The “A” component 4-[3-(trimethylsilyl)propoxy]benzoic acid (CAS: 948301-32-4) or 4-[butyl-dimethylsilyl)methoxy]benzoic acid (CAS: 948301-30-2) were synthesized separately using a modified Hegyes method [58] from sodium 4-(methoxycarbonyl)phenolate (Oxchem Corporation, Wood Dale, IL) and (3-chloropropyl)trimethylsilane (CAS: 2344-83-4; Gelest, Inc.) and from (chloromethyl)dimethyl-n-butylsilane (CAS: 3121-75-3; Jiwan Pharmaceutical Technology Co., Ltd., Jinan City, Shandong, China) separately (Fig. 1). The N-protected amino acid “B” components were coupled separately to the “C” component amine derivatives 4-amino-2,2,6,6-tetramethylpiperidine or aminomethylenediphosphonate tetraethyl ester in solution using a peptide coupling reagent benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate (BOP) or benzotriazol-1-yloxytripyrrolidinophophonium hexafluorophosphate (PyBOP) in the presence of N,N-Diisopropylethylamine (DIEA). The trifluoroacetic acid (TFA) deprotected “B-C” amino acid amide derivatives were acylated with the organosilicon “A” components in solution using a BOP coupling method to provide the desired organosilicon acyl-amino acid amide small molecules with an “A-B-C” structure (Fig. 1). The compounds were purified by preparative high-performance liquid chromatography (HPLC) or crystallization and characterized by analytical HPLC, thin-layer chromatography (TLC) and liquid chromatography-mass spectrometry (LC-MS).

Fig. 1.

Fig. 1

Synthesis and chemical structures of organosilicon compounds tested in this study. R1 group contains the silicon atom.

Cell culture and reagents

Human PCa cell lines LNCaP (American Type Culture Collection, Manassas, VA), C4-2, and C4-2B (provided by Dr. Leland WK Chung, Cedars-Sinai Medical Center, Los Angeles, CA) were routinely maintained in T-medium (ThermoFisher Scientific, Waltham, MA) with 5% fetal bovine serum (FBS; Atlanta Biologicals, Atlanta, GA). Docetaxel-resistant C4-2B-TaxR [31] and abiraterone-resistant C4-2B-AbiR [59] cells were provided by Dr. Allen C. Gao (University of California Davis, Davis, CA). C4-2B-TaxR cells were routinely maintained in RPMI1640 medium (Corning Inc., Corning, NY) supplemented with 10% FBS and 100 nM docetaxel (LC Laboratories, Woburn, MA). The final concentration of docetaxel in the culture medium was reduced to 5 nM before experimental assays. C4-2B-AbiR cells were routinely maintained in the presence of 10 μM abiraterone acetate (Selleck Chemicals, Houston, TX). CWR22Rv1 cells (provided by Dr. Jin-Tang Dong, Emory University, Atlanta, GA) were cultured in RPMI1640 medium supplemented with 10 % FBS, 1.5 g/L sodium bicarbonate (Corning), 4.5 g/L glucose, 10 mM Hepes and 1 mM sodium pyruvate (Hyclone, Logan, UT). PC-3 (ATCC) and DU145 (provided by Dr. Geou-Yarh Liou, Clark Atlanta University) were cultured in Dulbecco's Modified Eagle's Medium (DMEM, Corning) supplemented with 10% FBS. Cycloheximide (CHX) and dimethyl sulfoxide (DMSO) were purchased from Sigma-Aldrich (St. Louis, MO). DMSO was used as the vehicle control in in vitro and in vivo studies.

In vitro cytotoxicity assay

In vitro anticancer activity of silicon-containing compounds in the NCI-60 panel of human cancer cell lines was evaluated at the National Cancer Institute Developmental Therapeutics Program (Rockville, MD). Cell viability after 48 h of incubation was determined by sulforhodamine B staining. Growth inhibition of 50% (GI50) was calculated as the drug concentration resulting in a 50% reduction in the net protein increase in control cells during the drug incubation [60]. To determine the half minimal inhibitory concentration (IC50), defined as the concentration of drug required for 50% inhibition of cell proliferation after 72 h of incubation, CellTiter 96 AQueous non-radioactive cell proliferation assay kit (Promega, Madison, WI) or cell counting kit-8 (CCK-8, Dojindo Molecular Technologies, Inc., Rockville, MD) were used following the manufacturers’ instructions. IC50 values were calculated with SigmaPlot program (Systat Software Inc., San Jose, CA).

Polymerase Chain Reaction (PCR) analysis

Total RNA was prepared with Qiagen RNeasy Kit (Valencia, CA). The first-strand cDNA was synthesized using SuperScript® III First-Strand Synthesis System (ThermoFisher Scientific). Quantitative PCR (qPCR) was performed by the Stratagene Mx3005P system (Agilent Technologies, Santa Clara, CA) using a Brilliant® SYBR® Green QPCR Master Mix (Stratagene, San Diego, CA) according to the manufacturer's instructions. PCR primers used in this study are listed in Table S1.

Western blot analysis

Total cell lysates were prepared using Radioimmunoprecipitation (RIPA) Lysis and Extraction Buffer (ThermoFisher Scientific). Immunoblotting analyses were performed following standard procedures. Primary antibodies used in this study are listed in Table S2.

Protein half-life determination

PCa cells were incubated with CHX (50 μg/ml) for 2 h to inhibit further protein synthesis and then treated with DMSO or GH1504 for varying times. Total cell lysates were analyzed by Western blotting. The desired protein bands were quantitated and normalized by the intensity of the corresponding loading controls using the ImageJ program, and the data were graphed using the SigmaPlot program. The protein degradation rate was expressed as half-life (T½), the time for degradation of 50% of the protein, which was determined by an exponential decay fitting algorithm.

In vivo xenograft tumor growth

All animal procedures performed in this study were approved by Augusta University Institutional Animal Care and Use Committee (IACUC) and followed the National Institutes of Health guidelines. For the subcutaneous model, 2.0 × 106 CWR22Rv1 cells were mixed with Matrigel™ Membrane Matrix HC (Corning) and inoculated into both flanks (1.0 × 106 cells per site) of male athymic nude mice (Hsd: athymic nude-nu; 5 weeks old; Envigo RMS, Inc, Indianapolis, IN). Tumor-bearing mice were randomly divided into 2 groups and treated with vehicle control (DMSO; 5 mice) or GH1504 at 7.5 mg/kg (8 mice), 3 times per week, via intraperitoneal injection. Mice were weighed twice per week, and tumor sizes were measured and calculated using the formula of (length × width2 x ½).

For the intratibial xenograft model, a total of 2.0 × 106 C4-2B-TaxR cells were inoculated into the bilateral tibiae of male athymic nude mice (5 weeks old). Following the confirmation of tumor formation by rising prostate-specific antigen (PSA) levels in mouse sera (≥ 1.0 ng/ml), mice were randomly divided into 2 groups and treated with vehicle control (6 mice) or GH1504 at 7.5 mg/kg (5 mice), 3 times per week, via intraperitoneal injection. Mice were weighed twice per week, and tumor growth in bilateral tibiae was followed by serum PSA using an enzyme-linked immunosorbent assay (ELISA) kit from United Biotech, Inc (Mountain View, CA). At the end point, X-ray radiography was performed using MX-20 System (Faxitron, Tucson, Arizona). Major organs were examined for any abnormality.

Histopathology

Haemotoxylin and eosin (H&E) and immunohistochemistry (IHC) in xenograft tissues were performed following standard procedures. The antibodies used in IHC are listed in Table S2.

Statistical analysis

Each in vitro experiment was repeated independently at least three times. All results were presented as means ± standard error of the mean (SEM). Unpaired, two-tailed, Student t-test was used in statistical analyses. p < 0.05 was taken as a significant difference between means. To assess the longitudinal effect of treatment on tumor growth, a two-way analysis of variance (ANOVA) was performed to test the overall difference across different treatment groups during the entire study period. GraphPad Prism version 8.0 (GraphPad Software Inc., La Jolla, CA) was used to perform the statistical analyses.

Results

Anticancer activity screen of organosilicon compounds in the NCI-60 human cancer cell panel

To obtained an unbiased assessment of newly-developed organosilicon compounds as potential anticancer agents, we submitted five compounds (GH1501, GH1502, GH1503, GH1504, and GH1505; Table 1) to the National Cancer Institute Developmental Therapeutics Program for evaluation of their cytotoxic activity in the NCI-60 cell line panel, which contains nine types of diverse cancer cells representing leukemia, melanoma, non-small-cell lung, colon, kidney, ovarian, breast, prostate, and central nervous system cancers [60]. As summarized in Table 2, the five organosilicon compounds effectively suppressed the in vitro proliferation of multiple cancer cell lines. Among the tested compounds, GH1503 and GH1504 demonstrated high potency against all NCI-60 cancer cell lines, with an average GI50 value of 0.968 μM and 0.973 μM (Fig. 2A, 2B), respectively. Based on the NCI-60 screen results, GH1503 and GH1504 were selected as potential leads for further evaluation.

Table 1.

Several novel silicon-containing small-molecule compounds.

Name Chemical Formula Liner Formula Molecular Weight CAS Number
GH1501 MSipob-OC2Y-Atmp C38H51Cl2N3O4Si 712.83 1648613-37-9
GH1502 MSipob-Bip-Atmp C37H51N3O3Si 613.92 1648613-38-0
GH1503 BmSimob-OC2Y-Atmp C39H53Cl2N3O4Si 726.86 1648613-40-4
GH1504 BmSimob-Bip-Atmp C38H53N3O3Si 627.94 1648613-41-5
GH1505 MSipob-Bip-AMDP(OEt)4 C37H54N2O9P2Si 760.88 1648613-42-6

Table 2.

GI50 (nM) of selected anticancer small-molecule compounds in NCI-60 panel.

Panel/Cell Line GH1501 GH1502 GH1503 GH1504 GH1505
Breast Cancer
BT-549 2010 1990 2110 1960 2090
HS 578T 1920 1670 2010 1850 2610
MCF-7 335 506 356 332 1880
MDA-MB-231/ATCC 881 1360 494 880 1620
MDA-MB-468 1700 1640 1540 198 1720
T-47D 1900 1920 1840 1820 3100
Average 1458 1514 1392 1173 2170
Colon Cancer
COLO 205 216 392 342 215 1840
HCC-2998 1050 1560 1070 1110 2020
HCT-15 357 437 323 309 2250
HCT-116 206 297 319 218 1930
HT29 263 430 342 315 2020
KM12 363 413 382 376 2610
SW-620 313 392 318 316 2190
Average 395 560 442 408 2123
CNS Cancer
SF-268 903 1200 611 679 2070
SF-295 —— —— —— ——- ——
SF-539 1770 1810 1730 1770 1820
SNB-19 1720 1690 1600 1550 2060
SNB-75 1340 1320 1350 1630 ——
U-251 230 443 234 238 2090
Average 1193 1347 1105 1173 2010
Leukemia
CCRF-CEM 1200 1380 531 1500 2780
HL-60(TB) 813 1150 790 1220 2940
K-562 322 416 333 386 3460
MOLT-4 353 744 315 775 2860
RPMI-8226 1320 1800 893 1950 1970
SR ——- —— —— ——- ——-
Average 802 1098 572 1166 2802
Melanoma
LOX IMVI 197 212 191 207 1850
M14 341 466 334 286 2100
MALME-3M 1460 1610 1200 1750 1930
MDA-MB-435 269 305 250 292 1900
SK-MEL-2 1770 2050 1560 944 2110
SK-MEL-5 1720 1740 1680 234 1580
SK-MEL-28 1500 1690 1580 1330 1760
UACC-62 1490 1720 1500 1150 1620
UACC-257 1880 1840 1870 1780 2420
Average 1181 10047 1129 886 1919
Non-Small Cell Lung Cancer
A549/ATCC 407 1140 332 326 2840
EKVX —— —— —— —— ——
HOP-62 1860 1820 1510 1360 2000
HOP-92 1170 1280 1110 1370 2090
NCI-H23 1650 1830 1560 1760 3050
NCI-H226 1770 1770 1340 183 1840
NCI-H322M 1740 1840 1570 1710 5110
NCI-H460 253 370 259 348 1970
NCI-H522 1820 1940 1970 1060 2020
Average 1334 1499 1206 1015 2615
Ovarian Cancer
IGROV1 1200 1540 704 1640 3222
NCI/ADR-RES 1480 1710 1250 1540 3310
OVCAR-3 1250 1780 761 202 1870
OVCAR-4 1620 1720 711 521 2540
OVCAR-5 1680 1870 1620 1810 1710
OVCAR-8 268 662 263 216 2960
SK-OV-3 2080 2110 2030 2120 2590
Average 1368 1627 1048 1150 2600
Prostate Cancer
DU145 347 541 274 368 1930
PC-3 1320 1530 490 1300 2100
Average 834 1036 382 834 2015
Renal Cancer
786-0 369 1490 468 780 2760
A-498 1750 1770 1950 1950 3640
ACHN 1320 1440 478 402 2270
CAKI-1 1360 1620 1080 1580 2900
RXF 393 1510 1640 392 179 1870
SN12C 564 1490 613 526 1710
TK-10 1850 1920 1890 1110 2500
UO-31 1520 1610 1500 1550 2770
Average 1280 1623 1046 1010 2553
60 Cell Lines Average 1128 1316 968 973 2335

Fig. 2.

Fig. 2

In vitro effects of GH1504 in human cell lines. (A) Growth inhibition of 50% (GI50) of GH1504 in 9 types of human cancer cells in the NCI-60 panel. GI50 was calculated as the drug concentration resulting in a 50% reduction in the net protein increase in control cells after 48 h of incubation. (B) GI50 of GH1504 in all human cell lines included in the NCI-60 panel. (C) Half minimal inhibitory concentration (IC50) of GH1504 in established PCa cells. IC50 was determined as the concentration of drug required for 50% inhibition of cell proliferation after 72 h of incubation.

GH1504 exhibits potent in vitro cytotoxicity against ADT-resistant and chemoresistant PCa cells

Firstly, we determined and compared the in vitro cytotoxicity of GH1503 and GH1504 in CWR22Rv1 and C4-2B-TaxR, two representative drug-resistant PCa cell lines. CWR22Rv1 cells express both full-length AR (H875Y) and AR-v7, and are resistant to treatment with abiraterone and enzalutamide [61,62]; C4-2B-TaxR is a highly docetaxel-resistant C4-2B derivative that represents the phenotypes of acquired chemoresistance [31]. The IC50 values of GH1504 in CWR22Rv1 (0.89 μM) and C4-2B-TaxR (2.38 μM) were found to be lower than those of GH1503 (0.90 μM and 3.29 μM), respectively. Therefore, GH1504 was selected as a potential lead compound for targeting both ADT-resistant and chemoresistant PCa cells. The in vitro cytotoxicity of GH1504 was further evaluated in a panel of established PCa cell lines with different molecular and cellular characteristics, including androgen-dependent LNCaP, androgen-independent C4-2 and C4-2B [63], abiraterone-resistant C4-2B-AbiR [59], and AR-negative PC-3 and DU145 cells. The average IC50 value of GH1504 in these cell lines was determined as 1.50 μM (Fig. 2C). Taken together, these results indicated that GH1504 had low micromolar cytotoxicity in multiple PCa cells with diverse phenotypes, including those refractory to ADT and docetaxel chemotherapy.

GH1504 promotes protein degradation of AR, AR-v7 and survivin in CWR22Rv1 cells

We identified several downstream targets of GH1504 that may mediate its effects in ADT-resistant CWR22Rv1 cells. As shown in Fig. 3A, GH1504 treatment reduced the expression of AR, AR-v7 and survivin in a time-dependent manner. Heat-shock protein 27 (HSP27), an oncogenic protein that may play an important role in CRPC progression and therapeutic resistance [64,65], was also downregulated at 72 h following GH1504 incubation. On the other hand, expression of heat-shock protein 90 (HSP90), a molecular chaperon of AR and survivin [66], [67], [68], was not affected. We further investigated the mechanism by which GH1504 inhibited the expression of AR, AR-v7 and survivin. qPCR analyses found that GH1504 only slightly increased AR-v7 expression at 24 h but did not significantly change the RNA levels of AR and survivin during the time course between 0 h and 24 h (Fig. 3B). To determine whether GH1504 affected the stability of these proteins, a CHX pulse‐chase assay was performed. In the presence of CHX, an inhibitor of de novo protein synthesis, GH1504 treatment caused a significant reduction in the protein levels of AR, AR-v7 and survivin and shortened their predicted T1/2 from 21.8 h to 2.6 h (AR), 13.0 h to 12.2 h (AR-v7) and > 24.0 h to 23.7 h (survivin), respectively (Fig. 3C). These results indicated that GH1504 may promote protein degradation of AR, AR-v7 and survivin in CWR22Rv1 cells.

Fig. 3.

Fig. 3

In vitro Effects of GH1504 on the expression of AR, AR-v7 and survivin in CWR22Rv1 cells. (A) Protein expression of AR, AR-v7 and survivin in CWR22Rv1 cells treated with 0.9 µM GH1504 at the indicated time points. (B) qPCR analyses of RNA expression of AR, AR-v7 and survivin in CWR22Rv1 cells treated with 0.9 µM GH1504 at the indicated time points. p > 0.05 for all pairwise comparisons between 0 h and the indicated time points for all genes, except for that between 0 h and 24 h of AR-v7 (p = 0.035). (C) CWR22Rv1 cells were pre-incubated with CHX (50 µg/ml, 2 h) prior to treatment with DMSO or GH1504 (0.9 µM) at the indicated time points. Protein expression of AR, AR-v7 and survivin was analyzed by Western blotting and quantitated using the ImageJ program.

GH1504 inhibits the in vivo growth of CWR22Rv1 tumors in mice

We determined the efficacy of GH1504 against the in vivo growth of ADT-resistant PCa. CWR22Rv1 tumors were inoculated subcutaneously into male athymic nude mice and treated with vehicle or GH1504 (7.5 mg/kg), respectively, three times per week, via the intraperitoneal route. At the endpoint, the average tumor size of each treatment group was determined as 1096.11 ± 182.45 mm3 (vehicle) and 866.55 ± 221.72 mm3 (GH1504), respectively. Statistical analyses showed that compared with the vehicle control, GH1504 effectively suppressed the subcutaneous growth of CWR22Rv1 tumors (p = 0.014) (Fig. 4A). The treatments with both vehicle and GH1504 were associated with gains in the body weight of animals, specifically 26.69% in the control group and 11.21% in the GH1504 group (Fig. 4B). No significant adverse effects on animal behavior were observed. IHC analyses were performed to determine the in vivo effects of GH1504 on the expression of AR, AR-v7, and survivin in CWR22Rv1 tumor specimens (Fig. 4D). Compared with the control group, GH1504-treated tumors had reduced expression of AR, AR-v7, and survivin at tissue levels. These results demonstrated that as a monotherapy, GH1504 inhibited the in vivo growth of ADT-resistant PCa cells in immunocompromised mice.

Fig. 4.

Fig. 4

GH1504 inhibits the subcutaneous growth of CWR22Rv1 tumors in male athymic nude mice. (A) CWR22Rv1 tumor-bearing mice were treated with vehicle (n = 5) or GH1504 (7.5 mg/kg, intraperitoneally, 3 times per week; n = 8). Two-way ANOVA analysis was used to evaluate the statistical difference of the tumor sizes between different treatment groups. p = 0.014. (B) Average body weight of athymic nude mice treated with vehicle or GH1504. (C) H&E staining and IHC expression of AR, AR-v7 and survivin in CWR22Rv1 tumors treated with vehicle or GH1504. Scale bar: 50 µm.

GH1504 promotes protein degradation of AR and survivin in C4-2B-TaxR cells

GH1504 exhibited a low micromolar IC50 (2.38 μM) in the highly docetaxel-resistant C4-2B-TaxR cells [31] (Fig. 2C). Similar to its effect in CWR22Rv1 cells, GH1504 markedly reduced the protein expression of AR and survivin in a time-dependent manner and temporarily reduced HSP27 protein at 48 h (Fig. 5A). qPCR analyses found that following GH1504 treatment, AR mRNA level began to reduce at 6 h and survivin mRNA expression was lower at 12 h and 24 h (Fig. 5B). We further examined whether GH1504 affected the protein stability of AR and survivin in C4-2B-TaxR cells. In the presence of CHX, GH1504 shortened the predicted T1/2 of full-length AR (from 30.2 h to 22.0 h) and survivin (from 39.2 h to 29.0 h) (Fig. 5C). These results indicated that in C4-2B-TaxR cells, GH1504 may affect the expression of AR and survivin at both transcriptional and post-translational levels.

Fig. 5.

Fig. 5

In vitro Effects of GH1504 on the expression of AR and survivin in C4-2B-TaxR cells. (A) Protein expression of AR and survivin in C4-2B-TaxR cells treated with 2.3 µM GH1504 at the indicated time points. (B) qPCR analyses of RNA expression of AR and survivin in C4-2B-TaxR cells treated with 2.3 µM GH1504 at the indicated time points. * p < 0.05; ** p < 0.01; *** p < 0.001. (C) C4-2B-TaxR cells were pre-incubated with CHX (50 µg/ml, 2 h) prior to treatment with DMSO or GH1504 (2.3 µM) at the indicated time points. Protein expression of AR and survivin was analyzed by Western blotting and quantitated using the ImageJ program.

GH1504 retards the skeletal growth of C4-2B-TaxR xenografts in athymic nude mice

We evaluated the in vivo efficacy of GH1504 in the intratibial model of C4-2B-TaxR cells, which mimicked the clinicopathology of bone metastatic, chemoresistant PCa [31,63]. Following the confirmation of successful tumor inoculation, athymic nude mice were randomized into two groups and treated with the vehicle control or GH1504 (7.5 mg/kg), respectively, three times per week, via the intraperitoneal route. Previous studies from other groups and us have shown that serum PSA level is an excellent indicator of PCa growth in mouse skeletons [44,69]. Following an 11-week treatment, the average PSA level of each treatment group was 150.36 ± 61.29 ng/ml (control) and 85.51 ± 57.94 ng/ml (GH1504), respectively (Fig. 6A). Statistical analysis showed there were significant differences between the vehicle control and GH1504 groups (p = 0.02). The average body weight of mice in the control group was increased by 4.20% at the endpoint, and GH1504 treatment resulted in a slight decrease in the average body weight (-0.16%; Fig. 6B). X-ray radiography demonstrated that GH1504 treatment improved the skeletal architecture and reduced both osteolytic and osteoblastic lesions in tumor-bearing bones (Fig. 6C). No significant adverse effects on animal behavior were observed. Tissue expression of AR and survivin was further evaluated by IHC staining in bone xenografts, which showed that GH1504 could reduce the in vivo expression of AR and survivin in C4-2B-TaxR tumors (Fig. 6D). Taken together, these results indicated that GH1504 is effective in retarding the in vivo growth of chemoresistant PCa in mouse skeletons.

Fig. 6.

Fig. 6

GH1504 inhibits the intratibial growth of chemoresistant C4-2B-TaxR tumors in male athymic nude mice. (A) Serum PSA values of C4-2B-TaxR tumor-bearing mice treated with vehicle (n = 6) or GH1504 (7.5 mg/kg, intraperitoneally, 3 times per week; n = 5). Two-way ANOVA analysis was used to evaluate the statistical difference of the PSA values between different treatment groups. p = 0.02. (B) Average body weight of athymic nude mice treated with vehicle or GH1504. (C) Representative X-ray radiography of tumor-bearing tibiae in different treatment groups. Yellow arrow: osteolytic lesions; red arrow: osteoblastic lesions. (D) H&E staining and IHC expression of AR and survivin in C4-2B-TaxR bone tumors treated with vehicle or GH1504. Scale bar: 50 µm (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.).

Discussion

The treatment landscape for PCa has been extensively expanded over the last decade [5]. Several recent trials further demonstrated that the combination of docetaxel with standard ADT, or “chemohormonal therapy,” significantly improved overall survival in patients with hormone-sensitive PCa [6,7]. Unfortunately, many patients develop extreme drug resistance and have a poor prognosis. There is a great need for novel agents that can overcome resistance to both ADT and docetaxel chemotherapy. Given the central role of AR in CRPC progression, recent efforts have heavily been focused on developing more specific and potent inhibitors of AR signaling, and several new antiandrogens have entered clinical phases [14]. Darolutamide, a novel AR antagonist targeting both wild-type and mutated LBD variants, received FDA approval in 2019 for the treatment of CRPC [70]. In comparison, no significant progress has been made in developing a treatment to overcome chemoresistance. Since 2010, there has been only one registered trial in docetaxel-resistant CRPC, and that trial failed to demonstrate any survival benefits of figitumumab, a monoclonal antibody against insulin-like growth factor-1 receptor (IGF-1R) [71].

Using a combination of rational drug design, phenotypic screening and target deconvolution approaches, we have developed several generations of anticancer compounds. These small molecules share a common structure consisting of three pharmacophores derived from approved drugs, natural compounds and chemical moieties with known biological functions [39,[42], [43], [44], [45], [46],48]. Recently, we reported two small-molecule compounds as potential leads against drug-resistant PCa. Specifically, an aminobisphosphonate-containing compound BKM1972 effectively inhibited the skeletal growth of chemoresistant C4-2B-TaxR cells [44], and an α-phenylcinnamic acid derivative LG1836 significantly extended the survival of mice carrying ADT-resistant CWR22Rv1 tumors [48]. Although these results are promising, the anticancer potency of the two compounds remained suboptimal. For example, the IC50 values of BKM1972 in CWR22Rv1 and C4-2B-TaxR cells were 8.4 µM and 4.9 µM, and the IC50 of LG1836 in the two cell lines were 2.6 µM and 4.0 µM, respectively. To discover more effective candidates with the goal of targeting drug-resistant PCa, we conducted C/Si-exchange modifications to our 3-component chemicals and developed a panel of silicon-containing compounds. As we presented here, GH1504 had much improved cytotoxicity in these cells, specifically, with an IC50 of 0.89 µM in CWR22Rv1 and 2.89 µM in C4-2B-TaxR cells. Animal experiments further demonstrated that GH1504 alone was effective against the in vivo growth of CWR22Rv1 and C4-2B-TaxR xenografts. These studies indicated that GH1504 could be further evaluated as a potential lead for overcoming both ADT resistance and chemoresistance in advanced PCa.

Current drug discovery strategies can be largely categorized as target-based drug discovery and phenotypic drug discovery [72]. With the realization of a complex biology and high heterogeneity of human cancers, recent years have seen a renewed interest in the phenotypic approach that has the advantages of identifying first-in-class medicines with novel mechanisms of action. We have followed this route of drug discovery and conducted phenotypic screens of our compounds in-house and at the National Cancer Institute, as in the cases of GH1504 and several other potential leads [44]. We have made efforts to understand the mechanism of action of GH1504 in PCa cells. As described here, GH1504 was found to be capable of co-targeting AR, AR-v7 and survivin, which may collectively contribute to its anticancer activity in drug-resistant PCa cells. Furthermore, an important and remaining question is how GH1504 exerts its inhibitory effect in such a diverse array of human cancer cell lines, including both AR-positive and -negative cells (Fig. 2). While our current study identified several possible targets of GH1504 in these cells, such as HSP27, HSP90, and survivin (Figs. 3A, 5A, S1), it appeared that GH1504 may differentially affect these proteins in a cell context-dependent manner. For example, GH1504 effectively decreases HSP27 but does not alter HSP90 levels in CWR22Rv1 cells, whereas it does not reduce the protein levels of either HSP27 and HSP90 in C4-2B-TaxR cells. These results suggested that HSP27 and HSP90 may be controlled by different mechanisms in CWR22Rv1 and C4-2B-TaxR cells, and the effect of GH1504 on these proteins is a combined result of the compound in these cells. These observations are consistent with previous ones that HSP27 and HSP90 could be regulated at multiple levels by various factors [73,74]. We are in the process of identifying direct molecular target(s) of GH1504. These efforts could provide an explanation for the relatively high potency of GH1504 in a wide spectrum of cancer cell lines and a better understanding of different effects of GH1504 on downstream targets in heterogeneous human cancer cells.

Compared with conventional inhibitors of protein activities, such as enzalutamide and other antiandrogens, inhibition of protein expression may provide a more effective approach to block oncogenic signaling and improve therapeutic efficacy. Several strategies are being explored to harness the cancer cell's proteasomal machinery and achieve targeted degradation of oncogenic proteins, including those once considered as undruggable [75]. ARV-110, an orally bioavailable proteolysis-targeting chimera (PROTAC) degrader of AR, is being tested in heavily pretreated metastatic CRPC patients and has demonstrated a favorable preliminary efficacy [76]. Interestingly, our results showed that GH1504 can promote the protein turnover and degradation of AR, AR-v7 and survivin, suggesting that this compound may function as a “co-degrader” of these oncogenic factors. Given the important role of AR, AR-v7 and survivin in ADT resistance and chemoresistance, it is plausible to hypothesize that the simultaneous downregulation of these proteins by GH1504 could elicit higher anticancer activity against drug-resistant CRPC than current compounds targeting a single protein (such as ARV-110).

In conclusion, we have developed a novel anticancer compound GH1504 that effectively inhibited the in vitro and in vivo growth of drug-resistant PCa cells. This is the first silicon-containing small molecule that has been evaluated in preclinical models of PCa. GH1504 and its analogues may represent a new class of drug candidates to overcome therapeutic resistance in PCa patients.

CRediT authorship contribution statement

Rui Zhao: Formal analysis. Xiaowei Ma: Formal analysis. Lijuan Bai: Formal analysis. Xin Li: Formal analysis. Kenza Mamouni: Formal analysis. Yang Yang: Formal analysis. HongYan Liu: Conceptualization. Alira Danaher: Formal analysis. Nicholas Cook: Formal analysis. Omer Kucuk: Conceptualization. Robert S. Hodges: Conceptualization. Lajos Gera: Conceptualization, Formal analysis, Writing – original draft. Daqing Wu: Conceptualization, Writing – original draft.

Declaration of Competing Interest

The authors declare that there is no conflict of interest.

Acknowledgments

Acknowledgments

This paper is dedicated to the memory of our co-author Dr. Robert S. Hodges, a leading scientist in peptide chemistry and drug development. This work was supported by Georgia Research Alliance VentureLab grant, National Cancer Institute grants 1R41CA217491-01A1, 2R42CA217491-02A1, Emory University Winship Cancer Institute-Roswell Country Club Prostate Cancer Research Award, and the Department of Education Title III Program at Clark Atlanta University (D. Wu). The authors are grateful to the National Cancer Institute Developmental Therapeutics Program for the NCI-60 human tumor cell line screening. We also thank Emory University Winship Cancer Institute Cancer Tissue and Pathology core for histopathological studies, and Dr. Rhea-Beth Markowitz at Georgia Cancer Center for editorial assistance.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Footnotes

Supplementary material associated with this article can be found, in the online version, at doi:10.1016/j.neo.2021.11.006.

Appendix. Supplementary materials

mmc1.docx (1.6MB, docx)

References

  • 1.Litwin M.S., Tan H.J. The diagnosis and treatment of prostate cancer: a review. JAMA. 2017;317:2532–2542. doi: 10.1001/jama.2017.7248. [DOI] [PubMed] [Google Scholar]
  • 2.Siegel R.L., Miller K.D., Fuchs H.E., Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33. doi: 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
  • 3.Karantanos T., Corn P.G., Thompson T.C. Prostate cancer progression after androgen deprivation therapy: mechanisms of castrate resistance and novel therapeutic approaches. Oncogene. 2013;32:5501–5511. doi: 10.1038/onc.2013.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Berthold D.R., Pond G.R., Soban F., de Wit R., Eisenberger M., Tannock I.F. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: updated survival in the TAX 327 study. J Clin Oncol. 2008;26:242–245. doi: 10.1200/JCO.2007.12.4008. [DOI] [PubMed] [Google Scholar]
  • 5.Teo M.Y., Rathkopf D.E., Kantoff P. Treatment of advanced prostate cancer. Annu Rev Med. 2019;70:479–499. doi: 10.1146/annurev-med-051517-011947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.James N.D., Sydes M.R., Clarke N.W., Mason M.D., Dearnaley D.P., Spears M.R., Ritchie A.W., Parker C.C., Russell J.M., Attard G., et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet. 2016;387:1163–1177. doi: 10.1016/S0140-6736(15)01037-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Sweeney C.J., Chen Y.H., Carducci M., Liu G., Jarrard D.F., Eisenberger M., Wong Y.N., Hahn N., Kohli M., Cooney M.M., et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med. 2015;373:737–746. doi: 10.1056/NEJMoa1503747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Galletti G., Leach B.I., Lam L., Tagawa S.T. Mechanisms of resistance to systemic therapy in metastatic castration-resistant prostate cancer. Cancer Treat Rev. 2017;57:16–27. doi: 10.1016/j.ctrv.2017.04.008. [DOI] [PubMed] [Google Scholar]
  • 9.Park J.W., Lee J.K., Sheu K.M., Wang L., Balanis N.G., Nguyen K., Smith B.A., Cheng C., Tsai B.L., Cheng D., et al. Reprogramming normal human epithelial tissues to a common, lethal neuroendocrine cancer lineage. Science. 2018;362:91–95. doi: 10.1126/science.aat5749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ku S.Y., Rosario S., Wang Y., Mu P., Seshadri M., Goodrich Z.W., Goodrich M.M., Labbe D.P., Gomez E.C., Wang J., et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science. 2017;355:78–83. doi: 10.1126/science.aah4199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Frank S., Nelson P., Vasioukhin V. Recent advances in prostate cancer research: large-scale genomic analyses reveal novel driver mutations and DNA repair defects. F1000Research. 2018;7 doi: 10.12688/f1000research.14499.1. F1000 Faculty Rev-1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Anantharaman A., Friedlander T.W. Targeting the androgen receptor in metastatic castrate-resistant prostate cancer: a review. Urol Oncol. 2016;34:356–367. doi: 10.1016/j.urolonc.2015.11.003. [DOI] [PubMed] [Google Scholar]
  • 13.Coutinho I., Day T.K., Tilley W.D., Selth L.A. Androgen receptor signaling in castration-resistant prostate cancer: a lesson in persistence. Endocr Relat Cancer. 2016;23:T179–T197. doi: 10.1530/ERC-16-0422. [DOI] [PubMed] [Google Scholar]
  • 14.Aurilio G., Cimadamore A., Mazzucchelli R., Lopez-Beltran A., Verri E., Scarpelli M., Massari F., Cheng L., Santoni M., Montironi R. Androgen receptor signaling pathway in prostate cancer: from genetics to clinical applications. Cells. 2020;9:2693. doi: 10.3390/cells9122653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Paschalis A., Sharp A., Welti J.C., Neeb A., Raj G.V., Luo J., Plymate S.R., de Bono J.S. Alternative splicing in prostate cancer. Nat Rev Clin Oncol. 2018;15:663–675. doi: 10.1038/s41571-018-0085-0. [DOI] [PubMed] [Google Scholar]
  • 16.Antonarakis E.S., Lu C., Luber B., Wang H., Chen Y., Zhu Y., Silberstein J.L., Taylor M.N., Maughan B.L., Denmeade S.R., et al. Clinical significance of androgen receptor splice variant-7 mRNA detection in circulating tumor cells of men with metastatic castration-resistant prostate cancer treated with first- and second-line abiraterone and enzalutamide. J Clin Oncol. 2017;35:2149–2156. doi: 10.1200/JCO.2016.70.1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sharp A., Coleman I., Yuan W., Sprenger C., Dolling D., Nava Rodrigues D., Russo J.W., Figueiredo I., Bertan C., Seed G., et al. Androgen receptor splice variant-7 expression emerges with castration resistance in prostate cancer. J Clin Invest. 2018;129:192–208. doi: 10.1172/JCI122819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Antonarakis E.S., Lu C., Wang H., Luber B., Nakazawa M., Roeser J.C., Chen Y., Mohammad T.A., Chen Y., Fedor H.L., et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371:1028–1038. doi: 10.1056/NEJMoa1315815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Luo J., Attard G., Balk S.P., Bevan C., Burnstein K., Cato L., Cherkasov A., De Bono J.S., Dong Y., Gao A.C., et al. Role of androgen receptor variants in prostate cancer: report from the 2017 mission androgen receptor variants meeting. Eur Urol. 2018;73:715–723. doi: 10.1016/j.eururo.2017.11.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Scher H.I., Lu D., Schreiber N.A., Louw J., Graf R.P., Vargas H.A., Johnson A., Jendrisak A., Bambury R., Danila D., et al. Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate cancer. JAMA Oncol. 2016;2:1441–1449. doi: 10.1001/jamaoncol.2016.1828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zhu Y., Dalrymple S.L., Coleman I., Zheng S.L., Xu J., Hooper J.E., Antonarakis E.S., De Marzo A.M., Meeker A.K., Nelson P.S., et al. Role of androgen receptor splice variant-7 (AR-V7) in prostate cancer resistance to 2nd-generation androgen receptor signaling inhibitors. Oncogene. 2020;39:6935–6949. doi: 10.1038/s41388-020-01479-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lu C., Brown L.C., Antonarakis E.S., Armstrong A.J., Luo J. Androgen receptor variant-driven prostate cancer II: advances in laboratory investigations. Prostate Cancer Prostatic Dis. 2020;23:381–397. doi: 10.1038/s41391-020-0217-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gao L., Zhang W., Zhang J., Liu J., Sun F., Liu H., Hu J., Wang X., Wang X., Su P., et al. KIF15-mediated stabilization of AR and AR-V7 contributes to enzalutamide resistance in prostate cancer. Cancer Res. 2020;81:1026–1039. doi: 10.1158/0008-5472.CAN-20-1965. [DOI] [PubMed] [Google Scholar]
  • 24.Moon S.J., Jeong B.C., Kim H.J., Lim J.E., Kim H.J., Kwon G.Y., Jackman J.A., Kim J.H. Bruceantin targets HSP90 to overcome resistance to hormone therapy in castration-resistant prostate cancer. Theranostics. 2021;11:958–973. doi: 10.7150/thno.51478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liao Y., Liu Y., Xia X., Shao Z., Huang C., He J., Jiang L., Tang D., Liu J., Huang H. Targeting GRP78-dependent AR-V7 protein degradation overcomes castration-resistance in prostate cancer therapy. Theranostics. 2020;10:3366–3381. doi: 10.7150/thno.41849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kiliccioglu I., Konac E., Dikmen A.U., Sozen S., Bilen C.Y. Hsp-27 and NF-kappaB pathway is associated with AR/AR-V7 expression in prostate cancer cells. Gene. 2019;697:138–143. doi: 10.1016/j.gene.2019.02.055. [DOI] [PubMed] [Google Scholar]
  • 27.Lev A., Lulla A.R., Ross B.C., Ralff M.D., Makhov P.B., Dicker D.T., El-Deiry W.S. ONC201 targets AR and AR-V7 signaling, reduces PSA, and synergizes with everolimus in prostate cancer. Mol Cancer Res. 2018;16:754–766. doi: 10.1158/1541-7786.MCR-17-0614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wade C.A., Kyprianou N. Profiling prostate cancer therapeutic resistance. Int J Mol Sci. 2018;19:904. doi: 10.3390/ijms19030904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Takara K., Sakaeda T., Okumura K. An update on overcoming MDR1-mediated multidrug resistance in cancer chemotherapy. Curr Pharm Des. 2006;12:273–286. doi: 10.2174/138161206775201965. [DOI] [PubMed] [Google Scholar]
  • 30.Huang Y., Sadee W. Membrane transporters and channels in chemoresistance and -sensitivity of tumor cells. Cancer Lett. 2006;239:168–182. doi: 10.1016/j.canlet.2005.07.032. [DOI] [PubMed] [Google Scholar]
  • 31.Zhu Y., Liu C., Nadiminty N., Lou W., Tummala R., Evans C.P., Gao A.C. Inhibition of ABCB1 expression overcomes acquired docetaxel resistance in prostate cancer. Mol Cancer Ther. 2013;12:1829–1836. doi: 10.1158/1535-7163.MCT-13-0208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gottesman M.M., Fojo T., Bates S.E. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2:48–58. doi: 10.1038/nrc706. [DOI] [PubMed] [Google Scholar]
  • 33.Zhang M., Mukherjee N., Bermudez R.S., Latham D.E., Delaney M.A., Zietman A.L., Shipley W.U., Chakravarti A. Adenovirus-mediated inhibition of survivin expression sensitizes human prostate cancer cells to paclitaxel in vitro and in vivo. Prostate. 2005;64:293–302. doi: 10.1002/pros.20263. [DOI] [PubMed] [Google Scholar]
  • 34.Zhang M., Latham D.E., Delaney M.A., Chakravarti A. Survivin mediates resistance to antiandrogen therapy in prostate cancer. Oncogene. 2005;24:2474–2482. doi: 10.1038/sj.onc.1208490. [DOI] [PubMed] [Google Scholar]
  • 35.Kumari S., Senapati D., Heemers H.V. Rationale for the development of alternative forms of androgen deprivation therapy. Endocr Relat Cancer. 2017;24:R275–R295. doi: 10.1530/ERC-17-0121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sampath Kumar H.M., Herrmann L., Tsogoeva S.B. Structural hybridization as a facile approach to new drug candidates. Bioorg Med Chem Lett. 2020;30 doi: 10.1016/j.bmcl.2020.127514. [DOI] [PubMed] [Google Scholar]
  • 37.Ivasiv V., Albertini C., Goncalves A.E., Rossi M., Bolognesi M.L. Molecular hybridization as a tool for designing multitarget drug candidates for complex diseases. Curr Top Med Chem. 2019;19:1694–1711. doi: 10.2174/1568026619666190619115735. [DOI] [PubMed] [Google Scholar]
  • 38.Stewart J.M., Chan D.C., Simkeviciene V., Bunn P.A., Helfrich B., York E.J., Taraseviciene-Stewart L., Bironaite D., Gera L. Bradykinin antagonists as new drugs for prostate cancer. Int Immunopharmacol. 2002;2:1781–1786. doi: 10.1016/s1567-5769(02)00181-9. [DOI] [PubMed] [Google Scholar]
  • 39.Seo S.I., Gera L., Zhau H.E., Qian W.P., Iqbal S., Johnson N.A., Zhang S., Zayzafoon M., Stewart J., Wang R., et al. BKM1740, an acyl-tyrosine bisphosphonate amide derivative, inhibits the bone metastatic growth of human prostate cancer cells by inducing apoptosis. Clin Cancer Res. 2008;14:6198–6206. doi: 10.1158/1078-0432.CCR-08-1023. [DOI] [PubMed] [Google Scholar]
  • 40.Jutras S., Bachvarova M., Keita M., Bascands J.L., Mes-Masson A.M., Stewart J.M., Gera L., Bachvarov D. Strong cytotoxic effect of the bradykinin antagonist BKM-570 in ovarian cancer cells–analysis of the molecular mechanisms of its antiproliferative action. FEBS J. 2010;277:5146–5160. doi: 10.1111/j.1742-4658.2010.07928.x. [DOI] [PubMed] [Google Scholar]
  • 41.Gera L., Chan D., Hodges R., Bunn P. (2013). Flurbiprofen analogs and methods of use in treating cancer, U.S Patent US 8,575,170 B2.
  • 42.Gera L., Tentler J., Eckhardt G., Wu D., Hodges R.S. In: Proceedings of the 33rd European Peptide Symposium. Naydenova E., Pajpanova T., Danalev D., editors. Bulgarian Peptide Society; Sofia: 2015. Design and synthesis of novel peptidomimetic small molecule therapeutics with nanomolar anticancer activities; pp. 252–253. Peptides. 2015. [Google Scholar]
  • 43.Zhang S., Gera L., Mamouni K., Li X., Chen Z., Kucuk O., Wu D. Inhibition of skeletal growth of human prostate cancer by the combination of docetaxel and BKM1644: an aminobisphosphonate derivative. Oncotarget. 2016;7:27489–27498. doi: 10.18632/oncotarget.8481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chen Y., Gera L., Zhang S., Li X., Yang Y., Mamouni K., Wu A.Y., Liu H., Kucuk O., Wu D. Small molecule BKM1972 inhibits human prostate cancer growth and overcomes docetaxel resistance in intraosseous models. Cancer Lett. 2019;446:62–72. doi: 10.1016/j.canlet.2019.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gera L., Stewart J., Chung L.W., Wu D. (2010). Compositions and methods for treating bone cancer. 2010;US Patent 2010/0144678.
  • 46.Gera L., Hegyes P., Wu D., Hodges R.S. Proceedings of the 35th European Peptide Symposium. 2018. Design and synthesis of novel silicon-containing small molecule peptidomimetics with nanomolar anticancer activities. J Pept Scidoi.org/ [DOI] [Google Scholar]
  • 47.Stewart J.M., Gera L., Chan D.C., Bunn P.A., York E.J., Simkeviciene V., Helfrich B. Bradykinin-related compounds as new drugs for cancer and inflammation. Can J Physiol Pharmacol. 2002;80:275–280. doi: 10.1139/y02-030. [DOI] [PubMed] [Google Scholar]
  • 48.Chen Y., Li X., Mamouni K., Yang Y., Danaher A., White J., Liu H., Kucuk O., Gera L., Wu D. Novel small-molecule LG1836 inhibits the in vivo growth of castration-resistant prostate cancer. Prostate. 2020;80:993–1005. doi: 10.1002/pros.24032. [DOI] [PubMed] [Google Scholar]
  • 49.Bains W., Tacke R. Silicon chemistry as a novel source of chemical diversity in drug design. Curr Opin Drug Discov Dev. 2003;6:526–543. [PubMed] [Google Scholar]
  • 50.Pooni P.K., Showell G.A. Silicon switches of marketed drugs. Mini Rev Med Chem. 2006;6:1169–1177. doi: 10.2174/138955706778560120. [DOI] [PubMed] [Google Scholar]
  • 51.Meanwell N.A. Synopsis of some recent tactical application of bioisosteres in drug design. J Med Chem. 2011;54:2529–2591. doi: 10.1021/jm1013693. [DOI] [PubMed] [Google Scholar]
  • 52.Fujii S., Hashimoto Y. Progress in the medicinal chemistry of silicon: C/Si exchange and beyond. Future Med Chem. 2017;9:485–505. doi: 10.4155/fmc-2016-0193. [DOI] [PubMed] [Google Scholar]
  • 53.Bom D., Curran D.P., Kruszewski S., Zimmer S.G., Thompson Strode J., Kohlhagen G., Du W., Chavan A.J., Fraley K.A., Bingcang A.L., et al. The novel silatecan 7-tert-butyldimethylsilyl-10-hydroxycamptothecin displays high lipophilicity, improved human blood stability, and potent anticancer activity. J Med Chem. 2000;43:3970–3980. doi: 10.1021/jm000144o. [DOI] [PubMed] [Google Scholar]
  • 54.Bikzhanova G.A., Toulokhonova I.S., Gately S., West R. Novel silicon-containing drugs derived from the indomethacin scaffold: synthesis, characterization and evaluation of biological activity. Silicon Chem. 2007;3:209–217. [Google Scholar]
  • 55.Gately S., West R. Novel therapeutics with enhanced biological activity generated by the strategic introduction of silicon isosteres into known drug scaffolds. Drug Dev Res. 2007;68:156–163. [Google Scholar]
  • 56.Van Hattum A.H., Pinedo H.M., Schluper H.M., Hausheer F.H., Boven E. New highly lipophilic camptothecin BNP1350 is an effective drug in experimental human cancer. Int J Cancer. 2000;88:260–266. doi: 10.1002/1097-0215(20001015)88:2<260::aid-ijc18>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 57.Gera L., Hodges R.R., Hegyes P. (2017). Silylalkyloxyaryl compounds and methods for treating Cancer. United States Patent, US 9828393 B2.
  • 58.Hegyes P., Torocsik M. Alkoxy derivatives of benzoic acid esters and amides having antiplasmid activity, pharmaceutical compositions containing them and processes for their preparation. Hung. Pat. Appl. (2000), HU 9801407 A1 20000328.
  • 59.Liu C., Armstrong C., Zhu Y., Lou W., Gao A.C. Niclosamide enhances abiraterone treatment via inhibition of androgen receptor variants in castration resistant prostate cancer. Oncotarget. 2016;7:32210–32220. doi: 10.18632/oncotarget.8493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Shoemaker R.H. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6:813–823. doi: 10.1038/nrc1951. [DOI] [PubMed] [Google Scholar]
  • 61.McCarty D.J., Huang W., Kane M.A., Purushottamachar P., Gediya L.K., Njar V.C.O. Novel galeterone analogs act independently of AR and AR-V7 for the activation of the unfolded protein response and induction of apoptosis in the CWR22Rv1 prostate cancer cell model. Oncotarget. 2017;8:88501–88516. doi: 10.18632/oncotarget.19762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tan J., Sharief Y., Hamil K.G., Gregory C.W., Zang D.Y., Sar M., Gumerlock P.H., deVere White R.W., Pretlow T.G., Harris S.E., et al. Dehydroepiandrosterone activates mutant androgen receptors expressed in the androgen-dependent human prostate cancer xenograft CWR22 and LNCaP cells. Mol Endocrinol. 1997;11:450–459. doi: 10.1210/mend.11.4.9906. [DOI] [PubMed] [Google Scholar]
  • 63.Chen M.E., Lin S.H., Chung L.W., Sikes R.A. Isolation and characterization of PAGE-1 and GAGE-7. New genes expressed in the LNCaP prostate cancer progression model that share homology with melanoma-associated antigens. J Biol Chem. 1998;273:17618–17625. doi: 10.1074/jbc.273.28.17618. [DOI] [PubMed] [Google Scholar]
  • 64.Foster C.S., Dodson A.R., Ambroisine L., Fisher G., Moller H., Clark J., Attard G., De-Bono J., Scardino P., Reuter V.E., et al. Hsp-27 expression at diagnosis predicts poor clinical outcome in prostate cancer independent of ETS-gene rearrangement. Br J Cancer. 2009;101:1137–1144. doi: 10.1038/sj.bjc.6605227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Andrieu C., Taieb D., Baylot V., Ettinger S., Soubeyran P., De-Thonel A., Nelson C., Garrido C., So A., Fazli L., et al. Heat shock protein 27 confers resistance to androgen ablation and chemotherapy in prostate cancer cells through eIF4E. Oncogene. 2010;29:1883–1896. doi: 10.1038/onc.2009.479. [DOI] [PubMed] [Google Scholar]
  • 66.Coffey K., Robson C.N. Regulation of the androgen receptor by post-translational modifications. J Endocrinol. 2012;215:221–237. doi: 10.1530/JOE-12-0238. [DOI] [PubMed] [Google Scholar]
  • 67.Lee D.K., Chang C. Endocrine mechanisms of disease: expression and degradation of androgen receptor: mechanism and clinical implication. J Clin Endocrinol Metab. 2003;88:4043–4054. doi: 10.1210/jc.2003-030261. [DOI] [PubMed] [Google Scholar]
  • 68.Fortugno P., Beltrami E., Plescia J., Fontana J., Pradhan D., Marchisio P.C., Sessa W.C., Altieri D.C. Regulation of survivin function by Hsp90. Proc Natl Acad Sci U S A. 2003;100:13791–13796. doi: 10.1073/pnas.2434345100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhang J., Lu Y., Dai J., Yao Z., Kitazawa R., Kitazawa S., Zhao X., Hall D.E., Pienta K.J., Keller E.T. In vivo real-time imaging of TGF-beta-induced transcriptional activation of the RANK ligand gene promoter in intraosseous prostate cancer. Prostate. 2004;59:360–369. doi: 10.1002/pros.20019. [DOI] [PubMed] [Google Scholar]
  • 70.Bastos D.A., Antonarakis E.S. Darolutamide for castration-resistant prostate cancer. Onco Targets Ther. 2019;12:8769–8777. doi: 10.2147/OTT.S197244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.de Bono J.S., Piulats J.M., Pandha H.S., Petrylak D.P., Saad F., Aparicio L.M., Sandhu S.K., Fong P., Gillessen S., Hudes G.R., et al. Phase II randomized study of figitumumab plus docetaxel and docetaxel alone with crossover for metastatic castration-resistant prostate cancer. Clin Cancer Res. 2014;20:1925–1934. doi: 10.1158/1078-0432.CCR-13-1869. [DOI] [PubMed] [Google Scholar]
  • 72.Swinney D.C., Lee J.A. Recent advances in phenotypic drug discovery. F1000Research. 2020;9 doi: 10.12688/f1000research.25813.1. F1000 Faculty Rev-944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Prodromou C. Mechanisms of Hsp90 regulation. Biochem J. 2016;473:2439–2452. doi: 10.1042/BCJ20160005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Stetler R.A., Gao Y., Signore A.P., Cao G., Chen J. HSP27: mechanisms of cellular protection against neuronal injury. Curr Mol Med. 2009;9:863–872. doi: 10.2174/156652409789105561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ding Y., Fei Y., Lu B. Emerging new concepts of degrader technologies. Trends Pharmacol Sci. 2020;41:464–474. doi: 10.1016/j.tips.2020.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Proof-of-concept with PROTACs in prostate cancer. Cancer Discov. 2020;10:1084. doi: 10.1158/2159-8290.CD-NB2020-054. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

mmc1.docx (1.6MB, docx)

Data Availability Statement

All data generated or analyzed during this study are included in this published article.


Articles from Neoplasia (New York, N.Y.) are provided here courtesy of Neoplasia Press

RESOURCES