1
|
Wang R, Ma S, Xu N, Gan Y, Li P, Zhang J, Zhang Z, Gu Q, Xiang J. Developing a Novel Enzalutamide-Resistant Prostate Cancer Model via AR F877L Mutation in LNCaP Cells. Curr Protoc 2024; 4:e1033. [PMID: 38652202 DOI: 10.1002/cpz1.1033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Prostate cancer is a leading diagnosis and major cause of cancer-related deaths in men worldwide. As a typical hormone-responsive disease, prostate cancer is commonly managed with androgen deprivation therapy (ADT) to curb its progression and potential metastasis. Unfortunately, progression to castration-resistant prostate cancer (CRPC), a notably more aggressive phase of the disease, occurs within a timeframe of 2-3 years following ADT. Enzalutamide, a recognized androgen receptor (AR) antagonist, has been employed as a standard of care for men with metastatic castration-resistant prostate cancer (mCRPC) since it was first approved in 2012, due to its ability to prolong survival. However, scientific evidence suggests that sustained treatment with AR antagonists may induce acquired AR mutations or splice variants, such as AR F877L, T878A, and H875Y, leading to drug resistance and thereby diminishing the therapeutic efficacy of these agents. Thus, the establishment of prostate cancer models incorporating these particular mutations is essential for developing new therapeutic strategies to overcome such resistance and evaluate the efficacy of next-generation AR-targeting drugs. We have developed a CRISPR (clustered regularly interspaced short palindromic repeats)-based knock-in technology to introduce an additional F877L mutation in AR into the human prostate cell line LNCaP. This article provides comprehensive descriptions of the methodologies for cellular gene editing and establishment of an in vivo model. Using these methods, we successfully identified an enzalutamide-resistant phenotype in both in vitro and in vivo models. We also assessed the efficacy of target protein degraders (TPDs), such as ARV-110 and ARV-667, in both models, and the corresponding validation data are also included here. © 2024 Wiley Periodicals LLC. Basic Protocol 1: Generation of AR F877L-mutated LNCaP cell line using CRISPR technology Basic Protocol 2: Validation of drug resistance in AR F877L-mutated LNCaP cell line using the 2D CTG assay Support Protocol: Testing of sgRNA efficiency in HEK 293 cells Basic Protocol 3: Validation of drug resistance in AR F877L-mutated LNCaP cell line in vivo.
Collapse
Affiliation(s)
- Ruifeng Wang
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
- Organ Transplant Center & Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Shuhua Ma
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Nengwei Xu
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Yumiao Gan
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Pengya Li
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Jingying Zhang
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Zhixiang Zhang
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Qingyang Gu
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| | - Jian Xiang
- Oncology and Immunology Unit (OIU), WuXi Biology, WuXi AppTec, China
| |
Collapse
|
2
|
Efficacy of PACE4 pharmacotherapy in JHU-LNCaP-SM preclinical model of androgen independent prostate cancer. Sci Rep 2022; 12:17489. [PMID: 36261691 PMCID: PMC9582213 DOI: 10.1038/s41598-022-21593-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/29/2022] [Indexed: 01/12/2023] Open
Abstract
Prostate cancer (PCa) is a complex disease progressing from in situ to invasive or metastatic tumors while also being capable of modulating its androgen dependence. Understanding how novel therapies are working across the different stages of the disease is critical for their proper positioning in the spectrum of PCa treatments. The targeting of proprotein convertase PACE4 (Paired basic Amino Acid-Cleaving Enzyme 4) has been proposed as a novel approach to treat PCa. Animal studies performed on LNCaP xenografts, an androgen-dependent model, already yielded positive results. In this study, we tested PACE4 inhibition on JHU-LNCaP-SM, a newly described androgen-independent model, in cell-based and xenograft assays. Like LNCaP, JHU-LNCaP-SM cells express PACE4 and its oncogenic isoform PACE4-altCT. Using isoform-specific siRNAs, downregulation of PACE4-altCT resulted in JHU-LNCaP-SM growth inhibition. Furthermore, JHU-LNCaP-SM responded to the PACE4 pharmacological inhibitor known as C23 in cell-based assays as well as in athymic nude mice xenografts. These data support the efficacy of PACE4 inhibitors against androgen independent PCa thereby demonstrating that PACE4 is a key target in PCa. The JHU-LNCaP-SM cell line represents a model featuring important aspects of androgen-independent PCa, but it also represents a very convenient model as opposed to LNCaP cells for in vivo studies, as it allows rapid screening due to its high implantation rate and growth characteristics as xenografts.
Collapse
|
3
|
Phytochemical Profile and Biological Activity of the Ethanolic Extract from the Aerial Part of Crocus alatavicus Regel & Semen Growing Wildly in Southern Kazakhstan. Molecules 2022; 27:molecules27113468. [PMID: 35684407 PMCID: PMC9182240 DOI: 10.3390/molecules27113468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 02/05/2023] Open
Abstract
The composition of the ethanolic extract from the aerial parts of Crocus alatavicus Regel & Semen from southern Kazakhstan spontaneous flora was analyzed together with the determination of its antibacterial, antifungal, antiviral and anticancer activity. The phytochemical profile analysis by high-performance liquid chromatography-electrospray ionization-quadrupole-time of flight-mass spectrometry (HPLC/ESI-QTOF-MS) revealed the presence of multiple kaempferol derivatives. High-performance reverse-phase liquid chromatography combined with a photodiode-array detection (RP-HPLC/PDA) found that kaempferol 3-O-dihexoside and kaempferol 3-O-acyltetrahexoside accounted for 70.5% of the kaempferol derivatives. The minimum inhibitory concentration (MIC) values of the extract for all the tested reference microorganisms were high, reaching 10 mg/mL for yeasts and 20 mg/mL for bacteria. In contrast, antiviral activity was observed at 2 mg/mL, resulting in the inhibition of the HSV-1-induced cytopathic effect and the reduction in virus infectious titer by 1.96 log, as well as the viral load by 0.85 log. Among the tested prostate cancer cell lines, significant cytotoxic activity of the extract was noted only on the LNCaP cell line, with an IC50 value of 1.95 mg/mL. The LNCaP cell line treated with 2 mg/mL of the extract showed a noticeably reduced number of spindle-shaped cells with longer cellular projections, a significant increase in the peak corresponding to the population of apoptotic cells in the sub-G1 phase and a decreased intracellular glutathione (GSH) level, suggesting the prooxidative properties of the extract. The obtained data provide novel information about the flavonoids present in the aerial part of C. alatavicus and suggest its potential application as a source of the compounds active against HSV-1 and metastatic, androgen-sensitive prostate cancer.
Collapse
|
4
|
Kim J, Cho S, Park Y, Lee J, Park J. Evaluation of micro-RNA in extracellular vesicles from blood of patients with prostate cancer. PLoS One 2021; 16:e0262017. [PMID: 34972164 PMCID: PMC8719659 DOI: 10.1371/journal.pone.0262017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs) contain various types of molecules including micro-RNAs, so isolating EVs can be an effective way to analyze and diagnose diseases. A lot of micro-RNAs have been known in relation to prostate cancer (PCa), and we evaluate miR-21, miR-141, and miR-221 in EVs and compare them with prostate-specific antigen (PSA). EVs were isolated from plasma of 38 patients with prostate cancer and 8 patients with benign prostatic hyperplasia (BPH), using a method that showed the highest recovery of RNA. Isolation of EVs concentrated micro-RNAs, reducing the cycle threshold (Ct) value of RT-qPCR amplification of micro-RNA such as miR-16 by 5.12 and miR-191 by 4.65, compared to the values before EV isolation. Normalization of target micro-RNAs was done using miR-191. For miR-221, the mean expression level of patients with localized PCa was significantly higher than that of the control group, having 33.45 times higher expression than the control group (p < 0.01). Area under curve (AUC) between BPH and PCa for miR-221 was 0.98 (p < 0.0001), which was better than AUC for prostate-specific antigen (PSA) level in serum for the same patients. The levels of miR-21 and miR-141 in EVs did not show significant changes in patients with PCa compared to the control group in this study. This study suggests isolating EVs can be a helpful approach in analyzing micro-RNAs with regard to disease.
Collapse
Affiliation(s)
- Jiyoon Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| | - Siwoo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| | - Yonghyun Park
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jiyoul Lee
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaesung Park
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Gyeong-buk, Republic of Korea
| |
Collapse
|
5
|
Aslan M, Hsu EC, Liu S, Stoyanova T. Quantifying the invasion and migration ability of cancer cells with a 3D Matrigel drop invasion assay. Biol Methods Protoc 2021; 6:bpab014. [PMID: 34377838 PMCID: PMC8346651 DOI: 10.1093/biomethods/bpab014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022] Open
Abstract
Metastasis is the main cause of cancer-associated morbidity which will account for ∼ 600,000 deaths in the USA in 2021. Defining new mechanisms that drive cancer metastasis is vital for developing new therapeutic strategies and improving clinical outcomes for cancer patients. Herein, we describe a recently established 3D Matrigel drop invasion assay to measure cancer cell invasion and migration capability in vitro. This assay is a versatile and simple tool to test the ability of cells to invade and migrate, test the functional role of genes of interest in cell invasion and migration, analyze the localization of the target proteins at the cell invasion edge in situ, and screen drug effects on cancer cell invasion and migration.
Collapse
Affiliation(s)
- Merve Aslan
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - En-Chi Hsu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - Shiqin Liu
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| | - Tanya Stoyanova
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, 3155 Porter Drive. Palo Alto, CA 94304, USA
| |
Collapse
|
6
|
Subhawa S, Naiki-Ito A, Kato H, Naiki T, Komura M, Nagano-Matsuo A, Yeewa R, Inaguma S, Chewonarin T, Banjerdpongchai R, Takahashi S. Suppressive Effect and Molecular Mechanism of Houttuynia cordata Thunb. Extract against Prostate Carcinogenesis and Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13143403. [PMID: 34298624 PMCID: PMC8306559 DOI: 10.3390/cancers13143403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/20/2021] [Accepted: 07/04/2021] [Indexed: 01/17/2023] Open
Abstract
Simple Summary This study explored the chemopreventive effects of Houttuynia cordata Thunb. (HCT) extracts against prostate carcinogenesis in both androgen-sensitive prostate cancer and castration-resistant prostate cancer (CRPC) using the Transgenic Rat for Adenocarcinoma of Prostate (TRAP) model, CRPC xenograft mice, and prostate cancer cell lines. HCT suppressed cell proliferation and stimulated apoptosis via inactivation of AKT/ERK/MAPK in both androgen-sensitive prostate cancer and CRPC cell lines. HCT also inhibited cell migration and EMT phenotypes through the STAT3/Snail/Twist pathway. One of the active compounds of HCT was identified as rutin. Consistent with in vitro study, the incidence of adenocarcinoma in the TRAP model and CRPC tumor growth in the xenograft model were suppressed by induction of apoptosis and inactivation of AKT/ERK/MAPK by HCT intake. Our data demonstrated that HCT attenuated androgen-sensitive prostate cancer and CRPC by mechanisms that may involve inhibition of cell growth and caspase-dependent apoptosis pathways. Abstract Houttuynia cordata Thunb. (HCT) is a well-known Asian medicinal plant with biological activities used in the treatment of many diseases including cancer. This study investigated the effects of HCT extract and its ethyl acetate fraction (EA) on prostate carcinogenesis and castration-resistant prostate cancer (CRPC). HCT and EA induced apoptosis in androgen-sensitive prostate cancer cells (LNCaP) and CRPC cells (PCai1) through activation of caspases, down-regulation of androgen receptor, and inactivation of AKT/ERK/MAPK signaling. Rutin was found to be a major component in HCT (44.00 ± 5.61 mg/g) and EA (81.34 ± 5.21 mg/g) in a previous study. Rutin had similar effects to HCT/EA on LNCaP cells and was considered to be one of the active compounds. Moreover, HCT/EA inhibited cell migration and epithelial-mesenchymal transition phenotypes via STAT3/Snail/Twist pathways in LNCaP cells. The consumption of 1% HCT-mixed diet significantly decreased the incidence of adenocarcinoma in the lateral prostate lobe of the Transgenic rat for adenocarcinoma of prostate model. Similarly, tumor growth of PCai1 xenografts was significantly suppressed by 1% HCT treatment. HCT also induced caspase-dependent apoptosis via AKT inactivation in both in vivo models. Together, the results of in vitro and in vivo studies indicate that HCT has inhibitory effects against prostate carcinogenesis and CRPC. This plant therefore should receive more attention as a source for the future development of non-toxic chemopreventive agents against various cancers.
Collapse
Affiliation(s)
- Subhawat Subhawa
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Intravaroros Rd., Sripoom, Muang, Chiang Mai 50200, Thailand;
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
- Correspondence: (A.N.-I.); (R.B.); Tel.: +81-52-853-8156 (A.N.-I.); +66-53-93-5325 (R.B.); Fax: +81-52-842-0817 (A.N.-I.); +66-53-894-031 (R.B.)
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| | - Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| | - Masayuki Komura
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| | - Aya Nagano-Matsuo
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| | - Ranchana Yeewa
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Intravaroros Rd., Sripoom, Muang, Chiang Mai 50200, Thailand;
| | - Shingo Inaguma
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| | - Teera Chewonarin
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Intravaroros Rd., Sripoom, Muang, Chiang Mai 50200, Thailand;
| | - Ratana Banjerdpongchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 110 Intravaroros Rd., Sripoom, Muang, Chiang Mai 50200, Thailand;
- Correspondence: (A.N.-I.); (R.B.); Tel.: +81-52-853-8156 (A.N.-I.); +66-53-93-5325 (R.B.); Fax: +81-52-842-0817 (A.N.-I.); +66-53-894-031 (R.B.)
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan; (S.S.); (H.K.); (T.N.); (M.K.); (A.N.-M.); (R.Y.); (S.I.); (S.T.)
| |
Collapse
|
7
|
Stenberg VY, Juzeniene A, Bruland ØS, Larsen RH. In situ Generated <sup>212</sup>Pb-PSMA Ligand in a <sup>224</sup>Ra-Solution for Dual Targeting of Prostate Cancer Sclerotic Stroma and PSMA-positive Cells. Curr Radiopharm 2021; 13:130-141. [PMID: 32389119 PMCID: PMC7527546 DOI: 10.2174/1874471013666200511000532] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 12/31/2022]
Abstract
Background: New treatments combating bone and extraskeletal metastases are needed for patients with metastatic castration-resistant prostate cancer. The majority of metastases overexpress prostate-specific membrane antigen (PSMA), making it an ideal candidate for targeted radionuclide therapy. Objective: The aim of this study was to test a novel liquid 224Ra/212Pb-generator for the rapid preparation of a dual-alpha targeting solution. Here, PSMA-targeting ligands are labelled with 212Pb in the 224Ra-solution in transient equilibrium with daughter nuclides. Thus, natural bone-seeking 224Ra targeting sclerotic bone metastases and 212Pb-chelated PSMA ligands targeting PSMA-expressing tumour cells are obtained. Methods: Two PSMA-targeting ligands, the p-SCN-Bn-TCMC-PSMA ligand (NG001), specifically developed for chelating 212Pb, and the most clinically used DOTA-based PSMA-617 were labelled with 212Pb. Radiolabelling and targeting potential were investigated in situ, in vitro (PSMA-positive C4-2 human prostate cancer cells) and in vivo (athymic mice bearing C4-2 xenografts). Results: NG001 was rapidly labelled with 212Pb (radiochemical purity >94% at concentrations of ≥15 µg/ml) using the liquid 224Ra/212Pb-generator. The high radiochemical purity and stability of [212Pb]Pb-NG001 were demonstrated over 48 hours in the presence of ascorbic acid and albumin. Similar binding abilities of the 212Pb-labelled ligands were observed in C4-2 cells. The PSMA ligands displayed comparable tumour uptake after 2 hours, but NG001 showed a 3.5-fold lower kidney uptake than PSMA-617. Radium-224 was not chelated and, hence, showed high uptake in bones. Conclusion: A fast method for the labelling of PSMA ligands with 212Pb in the 224Ra/212Pb-solution was developed. Thus, further in vivo studies with dual tumour targeting by alpha-particles are warranted.
Collapse
Affiliation(s)
- Vilde Y Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway,Nucligen AS, Oslo, Norway,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Øyvind S Bruland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway,Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
8
|
Synthesis and evaluation of tumor-homing peptides for targeting prostate cancer. Amino Acids 2021; 53:645-652. [PMID: 33846842 DOI: 10.1007/s00726-021-02971-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/26/2021] [Indexed: 10/21/2022]
Abstract
High toxicity caused by chemotherapeutic drugs and the acquisition of drug resistance by cancer cells are the major drawbacks in cancer therapy. A promising approach to overcome the posed barriers is conjugating tumor-homing peptides to drugs or nanocarriers. Such high-affinity peptides can specifically target surface markers overexpressed by cancer cells, ensuring a rapid and cancer-specific uptake of the drugs. Since prostate-specific membrane antigen (PSMA) is overexpressed by aggressive prostate cancer cells, targeting this surface protein with peptide conjugates can lead to the development of effective strategies against prostate cancer. In this study, we aimed to determine which PSMA-binding peptide among peptides 563, 562 and 9-mer, show the highest selectivity towards PSMA using 22Rv1 prostate cancer cells, a cell line with moderate PSMA levels. Tumor-homing peptides were synthesized by fluorenylmethoxycarbonyl-based solid-phase peptide synthesis (Fmoc-SPPS) strategy, and evaluated for their prostate cancer cell-specific targeting efficiencies by flow cytometry. Our results showed that the PSMA-binding capacity of peptide 563 was superior to those of 562, 9-mer, and 5-mer; therefore, can be utilized as a potent-targeting agent not only in the treatment of high PSMA positive but also moderate PSMA positive prostate cancer tumors.
Collapse
|
9
|
Guo Q, Dong Y, Zhang Y, Fu H, Chen C, Wang L, Yang X, Shen M, Yu J, Chen M, Zhang J, Duan Y. Sequential Release of Pooled siRNAs and Paclitaxel by Aptamer-Functionalized Shell-Core Nanoparticles to Overcome Paclitaxel Resistance of Prostate Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:13990-14003. [PMID: 33739080 DOI: 10.1021/acsami.1c00852] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Paclitaxel (PTX) is a first-line chemotherapeutic agent to treat prostate cancer (PCa), but a large number of patients acquired drug resistance after short-term treatment. To develop combinational therapeutics to overcome PTX-resistant PCa, we established PTX-resistant LNCaP (LNCaP/PTX) cells and found that the LNCaP/PTX cells exhibited epithelial-mesenchymal transition (EMT) and enhanced metastasis during the selection process. We revealed that β-tubulin III, androgen receptor, and CXCR4 expressions were significantly increased in LNCaP/PTX cells and directly contributed to PTX resistance and EMT. Therefore, we developed prostate-specific membrane antigen aptamer (Apt)-functionalized shell-core nanoparticles (PTX/siRNAs NPs-Apt); the hydrophobic DSPE encapsulating PTX formed the dense inner core and the hydrophilic Apt-PEG2K with calcium phosphate (CaP) absorbing siRNAs formed the outer shell to sequentially release siRNAs and PTX, where CaP could trigger lysosomal escape to ensure that pooled siRNAs efficiently released into the cytoplasm to reverse EMT and resensitize PTX, while the PTX located in the core was subsequently released to exert the killing effect of chemotherapy to achieve the best synergistic effect. PTX/siRNAs NPs-Apt showed an enhanced tumor-targeting ability and achieved superior efficacy in the subcutaneous and orthotopic PCa tumor model with minimal side effects.
Collapse
Affiliation(s)
- Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chuanrong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liting Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xupeng Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Jiali Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
10
|
Multicomponent synthesis and anti-proliferative screening of biaryl triazole-containing cyclophanes. Bioorg Med Chem Lett 2021; 40:127899. [PMID: 33722739 DOI: 10.1016/j.bmcl.2021.127899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 11/20/2022]
Abstract
We report a practical two-step approach involving a Ugi 4-CR/ azide-alkyne cycloaddition for the synthesis of biaryl-containing cyclophanes. The series represents an extension of our previously reported macrocycles as an effort to enhance the anti-proliferative activity of this scaffold. In this variant, we incorporate a biphenyl moiety in the framework, thus enhancing the macrocycle size, lipophilicity, and structural diversity. Macrocycles were tested against different cell lines, being more cytotoxic against prostate (PC-3 and DU-145) and breast (MCF-7) tumor cells. Gratifyingly, the most active compound showed a significative enhancement of PC-3 growth inhibition with respect to our previous series, reaffirming the potential anti-proliferative activity of this kind of cyclophanes.
Collapse
|
11
|
Montazeri-Najafabady N, Chatrabnous N, Arabnezhad MR, Azarpira N. Anti-androgenic effect of astaxanthin in LNCaP cells is mediated through the aryl hydrocarbon-androgen receptors cross talk. J Food Biochem 2021; 45:e13702. [PMID: 33694182 DOI: 10.1111/jfbc.13702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 11/30/2022]
Abstract
The aim of this study was to investigate the anti-androgenic effects of astaxanthin (AST) on human prostatic cancer cell growth, and its impact on androgen receptor (AR) signaling using prostate cancer (PCa) cell line LNCaP. LNCaP cells were treated with AST alone and in combination with CH223191 and flutamide (Flu) in the presence and absence of testosterone. MTT assay, cellular prostate-specific antigen (PSA) and dihydrotestosterone (DHT) production, mRNA levels of CYP1A1, PSA, Kallikrein-Related Peptidase 2 (KLK2), Transmembrane Serine Protease 2 (TMPRSS2), and AR genes were measured as endpoints. The expression of CYP1A1, PSA, KLK2, TMPRSS2, and AR mRNA levels was decreased which results in reducing the production of PSA and DHT in the presence of testosterone. Our data clearly demonstrate that AST has a potential ability to suppress the human prostate LNCaP cells growth at high concentrations. AST was able to repress the testosterone-induced transcription of AR-target genes. PRACTICAL APPLICATIONS: Astaxanthin is a natural compound with the most potent antioxidant activity among other antioxidants. In the current study, ASX suppressed the LNCaP cells at high concentrations. Furthermore, AST inhibited testosterone-induced transcriptional activation of androgen-related genes. AST induced the expression of CYP1A1, which is able to metabolize the steroid hormones. It seems that AST can act as AhR exogenous ligand by induction of CYP1A1, which results in testosterone metabolism and consequent suppression of AR genes. So that, AST could prevent the growth of testosterone-dependent PCa cells, downregulate downstream genes in testosterone pathways, and enhance the metabolism of testosterone via AhR pathway. Collectively, AST could be considered as a potential candidate for the treatment of PCa.
Collapse
Affiliation(s)
| | - Nazanin Chatrabnous
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad-Reza Arabnezhad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Department of Pathology, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Ikwu FA, Shallangwa GA, Mamza PA. Ligand Based Design, ADMET and Molecular Docking Studies of Arylpiperazine Derivatives as Potent Anti-Proliferate Agents Against LNCAP Prostate Cancer Cell Lines. CHEMISTRY AFRICA 2021. [DOI: 10.1007/s42250-020-00210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
13
|
Hope JM, Bersi MR, Dombroski JA, Clinch AB, Pereles RS, Merryman WD, King MR. Circulating prostate cancer cells have differential resistance to fluid shear stress-induced cell death. J Cell Sci 2021; 134:jcs.251470. [PMID: 33526716 PMCID: PMC7929932 DOI: 10.1242/jcs.251470] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are exposed to fluid shear stress (FSS) of greater than 1000 dyn/cm2 (100 Pa) in circulation. Normally, CTCs that are exposed to FSS of this magnitude die. However, some CTCs develop resistance to this FSS, allowing them to colonize distant organs. We explored how prostate CTCs can resist cell death in response to forces of this magnitude. The DU145, PC3 and LNCaP human prostate cancer cell lines were used to represent cells of different metastatic origins. The cell lines were briefly treated with an average FSS of 3950 dyn/cm2 (395 Pa) using a 30 G needle and a syringe pump. DU145 cells had no change in cell viability, PC3 cells had some cell death and LNCaP cells exhibited significant cell death. These cell death responses correlated with increased cell membrane damage, less efficient membrane repair and increased stiffness. Additionally, FSS treatment prevented the LNCaP FSS-sensitive cell line from forming a growing tumor in vivo. This suggests that these properties play a role in FSS resistance and could represent potential targets for disrupting blood-borne metastasis. Summary: Prostate cancer cells have different sensitivities to fluid forces that alter their resistance to elevated blood flow-level fluid shear stress.
Collapse
Affiliation(s)
- Jacob M Hope
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Andrea B Clinch
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Rebecca S Pereles
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| |
Collapse
|
14
|
Copeland BT, Shallal H, Shen C, Pienta KJ, Foss CA, Pomper MG. Imaging and Characterization of Macrophage Distribution in Mouse Models of Human Prostate Cancer. Mol Imaging Biol 2020; 21:1054-1063. [PMID: 30805886 DOI: 10.1007/s11307-019-01318-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
PURPOSE Prostate carcinoma consists of tumor epithelium and malignant stroma. Until recently, diagnostic and therapeutic efforts have focused exclusively on targeting characteristics of the tumor epithelium, ignoring opportunities to target inflammatory infiltrate and extracellular matrix components. Prostate tumors are rich in tumor-associated macrophages (TAMs), which can be either of the cytotoxic M1 or protumorigenic M2 phenotype. We have quantified the proportion of each in seven common human prostate tumor lines grown subcutaneously in athymic nude mice and have imaged macrophage densities in vivo in xenografts derived from these lines. PROCEDURES A panel of seven human prostate cancer xenografts was generated in intact male athymic nude mice reflecting variable expression of the androgen receptor (AR) and prostate-specific membrane antigen (PSMA). Mice were imaged ex vivo using near-infrared fluorescence (NIRF) imaging for PSMA expression and total macrophage densities to enable direct comparison between the two. Tumors were harvested for sectioning and additional staining to delineate M1 and M2 phenotype along with vascular density. RESULTS Macrophage polarization analysis of sections revealed that all xenografts were > 94% M2 phenotype, and the few M1-polarized macrophages present were confined to the periphery. Xenografts displaying the fastest growth were associated with the highest densities of macrophages while the slowest growing tumors were characterized by focal, tumor-infiltrating macrophage densities. Xenograft sections displayed a strong positive spatial relationship between macrophages, vasculature, and PSMA expression. CONCLUSIONS Prostate TAM disposition can be imaged ex vivo and is associated with growth characteristics of a variety of tumor subtypes regardless of PSMA or AR expression.
Collapse
Affiliation(s)
- Ben T Copeland
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Hassan Shallal
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chentian Shen
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kenneth J Pienta
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Catherine A Foss
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| | - Martin G Pomper
- The Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
15
|
Bakrania A, Aubé M, Desjardins R, Sabbagh R, Day R. Upregulation of PACE4 in prostate cancer is not dependent on E2F transcription factors. Can J Physiol Pharmacol 2020; 98:477-481. [PMID: 32119574 DOI: 10.1139/cjpp-2019-0668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies in prostate cancer have identified PACE4, a proprotein convertase enzyme, as an emerging therapeutic target. Inhibition of PACE4-altCT, an oncogenic isoform of PACE4, using molecular or pharmacological approaches results in decreased cell proliferation and tumor progression in xenograft models. Although several validations have confirmed PACE4-altCT as a novel therapeutic target, the transcriptional regulation of PACE4 isoforms and mechanism of action remain a challenge. Previously, it has been reported that the human PACE4 promoter possesses potential binding sites for the E2F family of transcription factors, all of which are involved in cell cycle regulation and synthesis of DNA in mammalian cells. Therefore, we attempted to conduct in-depth evaluation of E2Fs on PACE4 and PACE4 isoform expression in prostate cancer. We conducted in vitro molecular silencing studies in various prostate cancer cell lines and determined the change in PACE4 expression levels. The results clearly show that the E2Fs alone do not alter PACE4 expression.
Collapse
Affiliation(s)
- Anita Bakrania
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Department of Surgery, Division of Urology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Mélanie Aubé
- Department of Surgery, Division of Urology, McGill University, Montréal, QC H3A 0G4, Canada
| | - Roxane Desjardins
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Department of Surgery, Division of Urology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Robert Sabbagh
- Department of Surgery, Division of Urology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - Robert Day
- Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Department of Surgery, Division of Urology, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada.,Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
16
|
Stenberg VY, Juzeniene A, Chen Q, Yang X, Bruland ØS, Larsen RH. Preparation of the alpha-emitting prostate-specific membrane antigen targeted radioligand [ 212 Pb]Pb-NG001 for prostate cancer. J Labelled Comp Radiopharm 2020; 63:129-143. [PMID: 31919866 DOI: 10.1002/jlcr.3825] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/17/2019] [Accepted: 01/06/2020] [Indexed: 12/26/2022]
Abstract
Prostate-specific membrane antigen (PSMA) is the most promising target for radioligand therapy of prostate cancer. The aim of this study was to prepare a small molecular ligand p-SCN-Bn-TCMC-PSMA (NG001) and compare it with the commonly used DOTA-based PSMA-617. The PSMA-targeting ability of the 212 Pb-labelled ligands was evaluated using PSMA-positive C4-2 human prostate cancer cells. Lead-212 is an in vivo generator of alpha particles by its daughter nuclides 212 Bi and 212 Po. NG001 was synthesized by conjugating the isothiocyanato group of p-SCN-Bn-TCMC to the amino group of a glutamate-urea-based PSMA-binding entity. Molecular size, chelator unit and chelator linking method are different in NG001 and PSMA-617. Both ligands were efficiently labelled with 212 Pb using a 224 Ra/212 Pb-solution generator in transient equilibrium with progeny. Lead-212-labelled NG001 was purified with a yield of 85.9±4.7% and with 0.7±0.2% of 224 Ra. Compared with [212 Pb]Pb-PSMA-617, [212 Pb]Pb-NG001 displayed a similar binding and internalization in C4-2 cells, with comparable tumour uptake in mice bearing C4-2 tumours, but almost a 2.5-fold lower kidney uptake. Due to the rapid normal tissue clearance and tumour cell internalization, any significant translocalization of 212 Bi was not detected in mice. In conclusion, the obtained results warrant further preclinical studies to evaluate the therapeutic efficacy of [212 Pb]Pb-NG001.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Research and Development, Nucligen AS, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Qingqi Chen
- Department of Drug Synthesis, MedKoo Biosciences, Morrisville, North Carolina
| | - Xiaoming Yang
- Department of Drug Synthesis, MedKoo Biosciences, Morrisville, North Carolina
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
17
|
Wu P, Xu R, Chen X, Zhao Y, Tan D, Zhao Y, Qin W, Zhang C, Ge X, Shi C. Establishment and characterization of patient-derived xenografts for hormone-naïve and castrate-resistant prostate cancers to improve treatment modality evaluation. Aging (Albany NY) 2020; 12:3848-3861. [PMID: 32092044 PMCID: PMC7066917 DOI: 10.18632/aging.102854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/28/2020] [Indexed: 04/12/2023]
Abstract
Prostate cancer (PC) is a heterogeneous disease characterized by variable morphological patterns. Thus, establishing a patient-derived xenograft (PDX) model that retains the key features of the primary tumor for each type of PC is important for appropriate evaluation. In this study, we established PDX models of hormone-naïve (D17225) and castration-resistant (B45354) PC by implanting fresh tumor samples, obtained from patients with advanced PC under the renal capsule of immune-compromised mice. Supplementation with exogenous androgens shortened the latent period of tumorigenesis and increased the tumor formation rate. The PDX models exhibited the same major genomic and phenotypic features of the disease in humans and maintained the main pathological features of the primary tumors. Moreover, both PDX models showed different outcomes after castration or docetaxel treatment. The hormone-naïve D17225 PDX model displayed a range of responses from complete tumor regression to overt tumor progression, and the development of castrate-resistant PC was induced after castration. The responses of the two PDX models to androgen deprivation and docetaxel were similar to those observed in patients with advanced PC. These new preclinical PC models will facilitate research on the mechanisms underlying treatment response and resistance.
Collapse
Affiliation(s)
- Pengpeng Wu
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Rong Xu
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xue Chen
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Ya Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Biomedicine Application Laboratory, School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yong Zhao
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xu Ge
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
18
|
Copeland BT, Du J, Pal SK, Jones JO. Factors that influence the androgen receptor cistrome in benign and malignant prostate cells. Mol Oncol 2019; 13:2616-2632. [PMID: 31520575 PMCID: PMC6887583 DOI: 10.1002/1878-0261.12572] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/14/2019] [Accepted: 09/11/2019] [Indexed: 01/24/2023] Open
Abstract
The androgen receptor (AR) plays key roles in the development of prostate tissue and the development and progression of prostate cancer (PC). AR guides cytodifferentiation and homeostasis in benign luminal epithelial cells; however, in PC, AR instead drives the uncontrolled proliferation of these cells. This ‘AR malignancy shift’ (AMS) is a central event in tumorigenesis. Using a ChIP‐seq approach in primary human tissues, cell lines, and mouse models, we demonstrate that the AMS occurs in every sample analyzed, suggesting that it is necessary for PC development. Using molecular and genetic techniques, we demonstrate that forkhead box (FOX)A1, HOXB13, GATA2, and c‐JUN are involved in the regulation of the AMS. AR‐binding sites (ARBS) are enriched for FOX, HOX, and GATA motifs in PC cells but not for c‐JUN motifs in benign cells. We show that the SPOP mutation commonly found in localized PCs can cause the AMS but is not transformative on its own and must be coupled to another mutation to transform cells. We show that the AMS occurs in mouse models of PC as well and that chronic low T, which is associated with increased PC risk and aggressiveness in humans, also causes the AMS in mice. We have discovered a previously unrecognized, fundamental tenet of PC, one which explains how and why AR signaling is different in cancer and benign cells. Our work has the potential to be used to stratify patients with localized PC for specific treatments. Furthermore, our work suggests that the AMS is a novel target for the treatment and/or prevention of PC.
Collapse
Affiliation(s)
- Ben T Copeland
- Deparment of Medical Oncology, City of Hope, Duarte, CA, USA
| | - Juan Du
- Integrative Genomics Core, City of Hope, Duarte, CA, USA
| | - Sumanta K Pal
- Deparment of Medical Oncology, City of Hope, Duarte, CA, USA
| | - Jeremy O Jones
- Deparment of Medical Oncology, City of Hope, Duarte, CA, USA
| |
Collapse
|
19
|
Shi C, Chen X, Tan D. Development of patient-derived xenograft models of prostate cancer for maintaining tumor heterogeneity. Transl Androl Urol 2019; 8:519-528. [PMID: 31807428 DOI: 10.21037/tau.2019.08.31] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Prostate cancer (Pca) is a heterogeneous disease with multiple morphological patterns. Thus, the establishment of a patient-derived xenograft (PDX) model that retains key features of the primary tumor is of great significance. This review demonstrates the characteristics and advantages of the Pca PDX model and summarizes the main factors affecting the establishment of the model. Because this model well recapitulates the diverse heterogeneity observed in the clinic, it was extensively utilized to discover new therapeutic targets, screen drugs, and explore metastatic mechanisms. In the future, clinical phenotype and different stages of the Pca patient might be faithfully reflected by PDX model, which provides tremendous potential for understanding Pca biology and achieving individualized treatment.
Collapse
Affiliation(s)
- Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Xue Chen
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China.,School of Basic Medical Sciences, the Chengdu Medical University, Xindu 610500, China
| | - Dengxu Tan
- Division of Cancer Biology, Laboratory Animal Center, the Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
20
|
Wang M, Zeng X, Li S, Sun Z, Yu J, Chen C, Shen X, Pan W, Luo H. A Novel Tanshinone Analog Exerts Anti-Cancer Effects in Prostate Cancer by Inducing Cell Apoptosis, Arresting Cell Cycle at G2 Phase and Blocking Metastatic Ability. Int J Mol Sci 2019; 20:4459. [PMID: 31510010 PMCID: PMC6770861 DOI: 10.3390/ijms20184459] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/31/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer (PCa), an epithelial malignant tumor, is the second common cause of cancer death among males in western countries. Thus, the development of new strategies is urgently needed. Tanshinones isolated from Salvia miltiorrhiza and its synthetic analogs show various biological activities including anticancer effects. Among them, the tanshinone analog 2-((Glycine methyl ester)methyl)-naphtho (TC7) is the most effective, with better selectivity and lower toxicity. Therefore, in this work, the effect of TC7 against PCa was investigated through assessing the molecular mechanisms regulating the growth, metastasis, and invasion of PCa cells. Human PCa cells, PC3 and LNCAP, were used to evaluate TC7 mechanisms of action in vitro, while male BALB/c nude mice were used for in vivo experiments by subjecting each mouse to a subcutaneous injection of PC3 cells into the right flank to evaluate TC7 effects on tumor volume. Our in vitro results showed that TC7 inhibited cell proliferation by arresting the cell cycle at G2/M through the regulation of cyclin b1, p53, GADD45A, PLK1, and CDC2/cyclin b1. In addition, TC7 induced cell apoptosis by regulating apoptosis-associated genes such as p53, ERK1, BAX, p38, BCL-2, caspase-8, cleaved-caspase-8, PARP1, and the phosphorylation level of ERK1 and p38. Furthermore, it decreased DNA synthesis and inhibited the migration and invasion ability by regulating VEGF-1 and MMP-9 protein expression. Our in vivo evidence supports the conclusion that TC7 could be considered as a potential promising chemotherapeutic candidate in the treatment of PCa.
Collapse
Affiliation(s)
- Mengling Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- College of pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Xueyi Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Shengyou Li
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
- College of pharmacy, Guizhou University, Guiyang 550025, China
| | - Zekun Sun
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
- College of pharmacy, Guizhou University, Guiyang 550025, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Chao Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Xiangchun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- College of pharmacy, Guizhou Medical University, Guiyang 550025, China
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| |
Collapse
|
21
|
Proteomic analysis of human prostate cancer PC-3M-1E8 cells and PC-3M-2B4 cells of same origin but with different metastatic potential. PLoS One 2018; 13:e0206139. [PMID: 30379883 PMCID: PMC6209233 DOI: 10.1371/journal.pone.0206139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/08/2018] [Indexed: 12/29/2022] Open
Abstract
Prostate cancer (PCa) is the second most frequently diagnosed cancer and the fifth leading cause of death from cancer in men worldwide. Increased understanding of the prostate cancer metastasis mechanisms will help identify more efficient intervention strategies to prevent or treat this deadly disease in the future. To identify the candidate proteins that contribute to metastasis of PCa, isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic analysis was performed to explore differentially expressed proteins between two homologous human prostate cancer cell lines including highly-metastatic PC-3M-1E8 cell line and poorly-metastatic PC-3M-2B4 cell line. Here, a total of 58 proteins were identified to be significantly differentially expressed between PC-3M-1E8 and PC-3M-2B4 cells, which were further verified using real-time quantitative PCR and western blot analysis. The bioinformatic analysis suggested that the differentially expressed proteins, like MMP1 and FHL1, may contribute to the higher metastatic ability of PC-3M-1E8 cells than PC-3M-2B4 cells. In addition, functional analyses proved MMP1’s positive effect on the higher metastatic ability of PC-3M-1E8 cells than PC-3M-2B4 cells. These findings provided a unique resource to specifically reveal the complex molecular regulatory mechanisms underlying the progression of prostate cancer from poorly-metastatic to highly-metastatic stage.
Collapse
|
22
|
Harmatys KM, Overchuk M, Chen J, Ding L, Chen Y, Pomper MG, Zheng G. Tuning Pharmacokinetics to Improve Tumor Accumulation of a Prostate-Specific Membrane Antigen-Targeted Phototheranostic Agent. Bioconjug Chem 2018; 29:3746-3756. [PMID: 30350576 DOI: 10.1021/acs.bioconjchem.8b00636] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We describe a simple and effective bioconjugation strategy to extend the plasma circulation of a low molecular weight targeted phototheranostic agent, which achieves high tumor accumulation (9.74 ± 2.26%ID/g) and high tumor-to-background ratio (10:1). Long-circulating pyropheophorbide (LC-Pyro) was synthesized with three functional building blocks: (1) a porphyrin photosensitizer for positron-emission tomography (PET)/fluorescence imaging and photodynamic therapy (PDT), (2) a urea-based prostate-specific membrane antigen (PSMA) targeting ligand, and (3) a peptide linker to prolong the plasma circulation time. With porphyrin's copper-64 chelating and optical properties, LC-Pyro demonstrated its dual-modality (fluorescence/PET) imaging potential for selective and quantitative tumor detection in subcutaneous, orthotopic, and metastatic murine models. The peptide linker in LC-Pyro prolonged its plasma circulation time about 8.5 times compared to its truncated analog. High tumor accumulation of LC-Pyro enabled potent PDT, which resulted in significantly delayed tumor growth in a subcutaneous xenograft model. This approach can be applied to improve the pharmacokinetics of existing and future targeted PDT agents for enhanced tumor accumulation and treatment efficacy.
Collapse
Affiliation(s)
- Kara M Harmatys
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada.,Department of Medical Biophysics , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Marta Overchuk
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada.,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - Juan Chen
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Lili Ding
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| | - Ying Chen
- Johns Hopkins Medical School , 1550 Orleans Street, 492 CRB II , Baltimore , Maryland 21287 , United States
| | - Martin G Pomper
- Johns Hopkins Medical School , 1550 Orleans Street, 492 CRB II , Baltimore , Maryland 21287 , United States
| | - Gang Zheng
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada.,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada.,Department of Medical Biophysics , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| |
Collapse
|