1
|
Vijayakumar S, González-Sánchez ZI, Amanullah M, Sonamuthu J, Rajkumar M, Divya M, Durán-Lara EF, Li M. Shark chondroitin sulfate gold nanoparticles: A biocompatible apoptotic agent for osteosarcoma. Int J Biol Macromol 2024; 290:138793. [PMID: 39689798 DOI: 10.1016/j.ijbiomac.2024.138793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/19/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
Osteosarcoma is a highly aggressive tumor that originates in the bone and often infiltrates nearby bone cells. It is the most prevalent type of primary bone cancer among the various bone malignancies. Traditional cancer treatment methods such as surgery, chemotherapy, immunotherapy, and radiotherapy have had restricted success. However, the integration of nanotechnology into cancer research has led to notable progress. One promising area is the use of marine-derived polysaccharide-based nano formulations for treating various human diseases, including cancer. This study presents a straightforward method for synthesizing biocompatible gold nanoparticles (AuNPs), utilizing sodium borohydride as a reducing agent and a cost-effective, water-soluble chondroitin sulfate (CS) derived from shark cartilage as a stabilizing agent. The synthesized CS-Au NPs appeared purple and were mainly spherical, with 40.768 nm of average size. Cytotoxicity assays (MTT) indicated that CS-Au NPs significantly reduced the viability of human osteosarcoma cells (MG63) at 100 μg/mL, while it showed no cytotoxic effects on mouse embryonic fibroblast cells (NIH3T3) at the same concentration. The observed toxicity of the CS-Au NPs was linked to a rise in the production of reactive oxygen species (ROS) within damaged mitochondria. ROS generation and changes in mitochondrial membrane potential were detected in MG63 cells treated with CS-Au NPs. Furthermore, apoptotic analysis through ethidium bromide dual staining and flow cytometry demonstrated that CS-Au NPs at higher concentrations significantly increased the amount of apoptotic cells, as demonstrated by acridine orange/ethidium bromide staining. Flow cytometry also confirmed that CS-Au NPs activated the apoptotic pathway in MG63 cells.
Collapse
Affiliation(s)
- Sekar Vijayakumar
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| | - Zaira I González-Sánchez
- Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, PUCMM, Autopista Duarte Km 1 ½, Santiago de los Caballeros, Dominican Republic; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Jegatheeswaran Sonamuthu
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mangaiyarkarasi Rajkumar
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Mani Divya
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi 630 003, Tamilnadu, India
| | - Esteban F Durán-Lara
- Bio&NanoMaterialsLab Drug Delivery and Controlled Release, Universidad de Talca, Talca 3460000, Maule, Chile; Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Maule, Chile
| | - Mingchun Li
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| |
Collapse
|
2
|
Fan D, Li J, Li L, An M, Yang H, Zhou G, Gao S, Bottini M, Zhang J, Ge K. Phosphate Ion-Responsive and Calcium Peroxide-Based Nanomedicine for Bone-Targeted Treatment of Breast Cancer Bone Metastasis. Adv Healthc Mater 2024; 13:e2402216. [PMID: 39109966 DOI: 10.1002/adhm.202402216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Indexed: 12/18/2024]
Abstract
The treatment of breast cancer bone metastasis is an unresolved clinical challenge, mostly because currently therapeutic approaches cannot simultaneously block the tumor growth and repair the osteolytic bone injuries at the metastatic site. Herein, the study develops a novel nanomedicine to treat breast cancer bone metastasis. The nanomedicine is based on phosphate ion-responsive and calcium peroxide-based nanoparticles carrying the bone-targeting agent zoledronic acid on the surface and loaded with the photosensitizer indocyanine green. Following intravenous administration to a mouse model of breast cancer bone metastasis, the nanoparticles efficiently accumulate at the bone metastasis site, react with free phosphate ions, and form hydroxyapatite nanoaggregates and O2, while releasing the photosensitizer. Hydroxyapatite nanoaggregates elicit the remineralization of the collagenous bone matrix and trigger tumor cell apoptosis. Upon irradiating tumor-bearing legs with an 808 nm laser source, the O2 and free photosensitizer produced 1O2 by the reaction of the nanoparticles with phosphate ions, further boosting the anti-tumor effect. Tumor killing hampers the vicious cycle at the site of bone metastasis, translating to osteolysis blockade and further encouraging the remineralization of bone matrix. This work sheds light on the development of a novel, safe, and efficient approach for the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Dehui Fan
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Jing Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Luwei Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Ming An
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
- Orthopedics Department, Bao Ding NO.1 Central Hospital, Baoding, 071000, China
| | - Hua Yang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Guoqiang Zhou
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
- Hebei University, Baoding, 071002, China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding, 071002, China
| | - Massimo Bottini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
- Sanford Burnham Prebys, La Jolla, CA, 92037, USA
| | - Jinchao Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| | - Kun Ge
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Material Science, Hebei University, Baoding, 071002, China
| |
Collapse
|
3
|
Wani SA, Qudrat S, Zubair H, Iqbal Z, Gulzar B, Aziz S, Inayat A, Safi D, Kamran A. Role of osteoclast inhibitors in prostate cancer bone metastasis; a narrative review. J Oncol Pharm Pract 2024:10781552241275943. [PMID: 39169855 DOI: 10.1177/10781552241275943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
OBJECTIVE To study the role of Osteoclast inhibitors in advanced prostate cancer metastasis treatment and their efficacy in reducing skeletal related events. METHODS: DATA SOURCE A comprehensive search was done using search terms as "osteoclast inhibitors" "Bisphosphonates" "Zoledronic acid" " pamidronate" " Alendronate" "Denosumab" " Prostate cancer metastasis" in pubmed and Google scholar. Relevant articles were screened and collected . The collected articles were used to frame the review and data showing use of Osteoclast inhibitors In prostate cancer bone metastasis was collected. DATA SUMMARY Prostate cancer metastasizes most commonly to the skeleton thus leading to significant morbidity ranging from pain, pathological fractures to spinal cord compression and are the primary cause of patient disability and reduced quality of life.Initially, radiation therapy and radiopharmaceuticals were the mainstay of treatment however the role of Bisphosphonates and denosumab has become an integral part of therapy to manage metastatic prostate cancer. These agents significantly decrease skeletal related events and enhance patients quality of life. Emerging therapies like Radium-223 have also shown promise in reducing skeletal related events and also improving survival rates in patients with bone metastasis. Other treatment options which are being used are systemic agents like Docetaxel, cabazitaxel, hormonal therapies like abiraterone and enzalutamide. Immunotherapy with sipuleucel-T has demonstrated a reduction in mortality among prostate cancer patients with metastasis, highlighting the need for further research in this area. Ongoing studies are investigating novel agents that target both tumor cells and the bone microenvironment. CONCLUSION Osteoclast inhibitors are effective in reducing skeletal related events in advanced bone metastasis and improve the quality of life of patients.
Collapse
Affiliation(s)
- Shariq Ahmad Wani
- Department of Internal Medicine, Government medical college, Srinagar, India
| | - Salma Qudrat
- internal Medicine, Khyber teaching hospital, Peshawar, Pakistan
| | - Hina Zubair
- Internal Medicine, Rawalpindi medical university, Rawalpindi, Pakistan
| | - Zahra Iqbal
- Internal Medicine, Virginia commonwealth university, Richmond, USA
| | - Babar Gulzar
- Department of Internal Medicine, Government medical college, Srinagar, India
| | - Sundal Aziz
- Internal Medicine, Khyber medical university, Peshawar, Pakistan
| | - Arsalan Inayat
- Internal Medicine, HSHS St Mary's Hospital, Decatur, Illinois, USA
| | - Danish Safi
- Internal Medicine, WVU hospital, Morgantown, WV, USA
| | - Amir Kamran
- Internal Medicine, Charleston Area Medical center, Charleston, USA
| |
Collapse
|
4
|
Jiang H, Liu M, Deng Y, Zhang C, Dai L, Zhu B, Ou Y, Zhu Y, Hu C, Yang L, Li J, Bai Y, Yang D. Identification of prostate cancer bone metastasis related genes and potential therapy targets by bioinformatics and in vitro experiments. J Cell Mol Med 2024; 28:e18511. [PMID: 39098992 PMCID: PMC11298316 DOI: 10.1111/jcmm.18511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
The aetiology of bone metastasis in prostate cancer (PCa) remains unclear. This study aims to identify hub genes involved in this process. We utilized machine learning, GO, KEGG, GSEA, Single-cell analysis, ROC methods to identify hub genes for bone metastasis in PCa using the TCGA and GEO databases. Potential drugs targeting these genes were identified. We validated these results using 16 specimens from patients with PCa and analysed the relationship between the hub genes and clinical features. The impact of APOC1 on PCa was assessed through in vitro experiments. Seven hub genes with AUC values of 0.727-0.926 were identified. APOC1, CFH, NUSAP1 and LGALS1 were highly expressed in bone metastasis tissues, while NR4A2, ADRB2 and ZNF331 exhibited an opposite trend. Immunohistochemistry further confirmed these results. The oxidative phosphorylation pathway was significantly enriched by the identified genes. Aflatoxin B1, benzo(a)pyrene, cyclosporine were identified as potential drugs. APOC1 expression was correlated with clinical features of PCa metastasis. Silencing APOC1 significantly inhibited PCa cell proliferation, clonality, and migration in vitro. This study identified 7 hub genes that potentially facilitate bone metastasis in PCa through mitochondrial metabolic reprogramming. APOC1 emerged as a promising therapeutic target and prognostic marker for PCa with bone metastasis.
Collapse
Affiliation(s)
- Haiyang Jiang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Yingfei Deng
- Pathology‐DepartmentThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chongjian Zhang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Longguo Dai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Bingyu Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yitian Ou
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Yong Zhu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Chen Hu
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Libo Yang
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Jun Li
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Yu Bai
- Department of Urology IThe Third Affiliated Hospital of Kunming Medical University (Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Cancer Center of Yunnan Province)KunmingYunnanChina
| | - Delin Yang
- Department of Urology IIThe second Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
5
|
Ahmed ME, Mahmoud AM, Reitano G, Zeina W, Lehner K, Day CA, Riaz I, Childs DS, Orme JJ, Tuba Kendi A, Johnson GB, Jeffrey Karnes R, Kwon ED, Andrews JR. Survival Patterns Based on First-site-specific Visceral Metastatic Prostate Cancer: Are Outcomes of Visceral Metastases the Same? EUR UROL SUPPL 2024; 66:38-45. [PMID: 39040620 PMCID: PMC11260861 DOI: 10.1016/j.euros.2024.06.006] [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] [Accepted: 06/11/2024] [Indexed: 07/24/2024] Open
Abstract
Background and objective Visceral metastatic disease in prostate cancer patients conveys a poor prognosis. Using advanced imaging techniques, studies have demonstrated increasing detection rates of visceral metastasis. Visceral metastases are now seen in up to 30-60% of prostate cancer patients. Survival patterns of site-specific visceral metastasis are described poorly in the literature. Here, we sought to investigate survival patterns in prostate cancer patients according to their first detected site of visceral metastasis. Methods Retrospectively, we identified 203 prostate cancer patients with visceral metastases from the Mayo Clinic Advanced Prostate Cancer Registry. Patients were divided into three groups according to the first site of visceral metastases detected: lung, brain, or liver. Visceral metastases were detected primarily on either metabolic imaging (C-11 choline) or prostate-specific membrane antigen positron emission tomography computed tomography (CT) scan. Confirmation of visceral metastasis diagnosis was established with either biopsy when feasible or focused conventional imaging, including focused CT or magnetic resonance imaging. Overall survival and cancer-specific survival were estimated using the Kaplan-Meier method. Univariate and multivariate Cox regression model was conducted to assess different variables that affect overall and cancer-specific survival. Key findings and limitations Over a median (interquartile range) follow-up duration of 16.2 (3.9-49.8) mo, the overall and cancer-specific survival of the entire cohort suggests better survival patterns in patients with first-site lung metastases than in patients with first-site brain or liver metastases (p < 0.0001). In univariate and multivariate analyses of factors impacting patients' overall and cancer-specific survival, a high prostate-specific antigen level at diagnosis of visceral metastasis, concomitant bone and lymph node disease, and more than four visceral metastases were associated with poor overall and cancer-specific survival (p < 0.05). On the contrary, first-site lung metastasis was associated with improved overall and cancer-specific survival, compared with first-site liver and brain metastases (p < 0.001). Conclusions and clinical implications These data suggest that prostate cancer patients with visceral metastatic disease have varying survival patterns according to first-site detected visceral metastasis. In our cohort, patients with first-site lung metastasis demonstrated better survival outcomes than patients with first-site brain or liver metastasis. Patient summary Our study explored the survival outcomes among patients with visceral metastatic prostate cancer employing cutting-edge imaging methods. Prostate cancer patients with metastases to different organs have different survival rates. Patients with cancer spreading to the lungs first showed better survival than those with cancer spreading to the brain or liver first.
Collapse
Affiliation(s)
| | | | | | - Wael Zeina
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Kelly Lehner
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Carter A. Day
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Irbaz Riaz
- Department of Medical Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | | | - Jacob J. Orme
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - A. Tuba Kendi
- Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | - Geoffrey B. Johnson
- Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Jack R. Andrews
- Department of Urology, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
6
|
Belue MJ, Harmon SA, Yang D, An JY, Gaur S, Law YM, Turkbey E, Xu Z, Tetreault J, Lay NS, Yilmaz EC, Phelps TE, Simon B, Lindenberg L, Mena E, Pinto PA, Bagci U, Wood BJ, Citrin DE, Dahut WL, Madan RA, Gulley JL, Xu D, Choyke PL, Turkbey B. Deep Learning-Based Detection and Classification of Bone Lesions on Staging Computed Tomography in Prostate Cancer: A Development Study. Acad Radiol 2024; 31:2424-2433. [PMID: 38262813 PMCID: PMC11214604 DOI: 10.1016/j.acra.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/25/2024]
Abstract
RATIONALE AND OBJECTIVES Efficiently detecting and characterizing metastatic bone lesions on staging CT is crucial for prostate cancer (PCa) care. However, it demands significant expert time and additional imaging such as PET/CT. We aimed to develop an ensemble of two automated deep learning AI models for 1) bone lesion detection and segmentation and 2) benign vs. metastatic lesion classification on staging CTs and to compare its performance with radiologists. MATERIALS AND METHODS This retrospective study developed two AI models using 297 staging CT scans (81 metastatic) with 4601 benign and 1911 metastatic lesions in PCa patients. Metastases were validated by follow-up scans, bone biopsy, or PET/CT. Segmentation AI (3DAISeg) was developed using the lesion contours delineated by a radiologist. 3DAISeg performance was evaluated with the Dice similarity coefficient, and classification AI (3DAIClass) performance on AI and radiologist contours was assessed with F1-score and accuracy. Training/validation/testing data partitions of 70:15:15 were used. A multi-reader study was performed with two junior and two senior radiologists within a subset of the testing dataset (n = 36). RESULTS In 45 unseen staging CT scans (12 metastatic PCa) with 669 benign and 364 metastatic lesions, 3DAISeg detected 73.1% of metastatic (266/364) and 72.4% of benign lesions (484/669). Each scan averaged 12 extra segmentations (range: 1-31). All metastatic scans had at least one detected metastatic lesion, achieving a 100% patient-level detection. The mean Dice score for 3DAISeg was 0.53 (median: 0.59, range: 0-0.87). The F1 for 3DAIClass was 94.8% (radiologist contours) and 92.4% (3DAISeg contours), with a median false positive of 0 (range: 0-3). Using radiologist contours, 3DAIClass had PPV and NPV rates comparable to junior and senior radiologists: PPV (semi-automated approach AI 40.0% vs. Juniors 32.0% vs. Seniors 50.0%) and NPV (AI 96.2% vs. Juniors 95.7% vs. Seniors 91.9%). When using 3DAISeg, 3DAIClass mimicked junior radiologists in PPV (pure-AI 20.0% vs. Juniors 32.0% vs. Seniors 50.0%) but surpassed seniors in NPV (pure-AI 93.8% vs. Juniors 95.7% vs. Seniors 91.9%). CONCLUSION Our lesion detection and classification AI model performs on par with junior and senior radiologists in discerning benign and metastatic lesions on staging CTs obtained for PCa.
Collapse
Affiliation(s)
- Mason J Belue
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Stephanie A Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Dong Yang
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Julie Y An
- Department of Radiology, University of California, San Diego, California, USA (J.Y.A.)
| | - Sonia Gaur
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA (S.G.)
| | - Yan Mee Law
- Department of Radiology, Singapore General Hospital, Singapore (Y.M.L.)
| | - Evrim Turkbey
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA (E.T., B.J.W.)
| | - Ziyue Xu
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Jesse Tetreault
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Nathan S Lay
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Enis C Yilmaz
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Tim E Phelps
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Benjamin Simon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (P.A.P.)
| | - Ulas Bagci
- Radiology and Biomedical Engineering Department, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA (U.B.)
| | - Bradford J Wood
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA (E.T., B.J.W.); Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (B.J.W.)
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (D.E.C.)
| | - William L Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (W.L.D., R.A.M.)
| | - Ravi A Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (W.L.D., R.A.M.)
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA (J.L.G.)
| | - Daguang Xu
- NVIDIA Corporation, Santa Clara, California, USA (D.Y., Z.X., J.T., D.X.)
| | - Peter L Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.)
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, 10 Center Dr., MSC 1182, Building 10, Room B3B85, Bethesda, Maryland, USA (M.J.B., S.A.H., N.S.L., E.C.Y., T.E.P., B.S., L.L., E.M., P.L.C., B.T.).
| |
Collapse
|
7
|
Zhang B, Liu M, Mai F, Li X, Wang W, Huang Q, Du X, Ding W, Li Y, Barwick BG, Ni JJ, Osunkoya AO, Chen Y, Zhou W, Xia S, Dong JT. Interruption of KLF5 acetylation promotes PTEN-deficient prostate cancer progression by reprogramming cancer-associated fibroblasts. J Clin Invest 2024; 134:e175949. [PMID: 38781024 PMCID: PMC11245161 DOI: 10.1172/jci175949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
Inactivation of phosphatase and tensin homolog (PTEN) is prevalent in human prostate cancer and causes high-grade adenocarcinoma with a long latency. Cancer-associated fibroblasts (CAFs) play a pivotal role in tumor progression, but it remains elusive whether and how PTEN-deficient prostate cancers reprogram CAFs to overcome the barriers for tumor progression. Here, we report that PTEN deficiency induced Krüppel-like factor 5 (KLF5) acetylation and that interruption of KLF5 acetylation orchestrated intricate interactions between cancer cells and CAFs that enhance FGF receptor 1 (FGFR1) signaling and promote tumor growth. Deacetylated KLF5 promoted tumor cells to secrete TNF-α, which stimulated inflammatory CAFs to release FGF9. CX3CR1 inhibition blocked FGFR1 activation triggered by FGF9 and sensitized PTEN-deficient prostate cancer to the AKT inhibitor capivasertib. This study reveals the role of KLF5 acetylation in reprogramming CAFs and provides a rationale for combined therapies using inhibitors of AKT and CX3CR1.
Collapse
Affiliation(s)
- Baotong Zhang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Fengyi Mai
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Xiawei Li
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Inner Mongolia Institute of Quality and Standardization, Inner Mongolia Administration for Market Regulation, Hohhot, China
| | - Wenzhou Wang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Qingqing Huang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Xiancai Du
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Weijian Ding
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jianping Jenny Ni
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Adeboye O. Osunkoya
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Departments of Pathology and Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yuanli Chen
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Siyuan Xia
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Li G, Zhao R, Xie Z, Qu X, Duan Y, Zhu Y, Liang H, Tang D, Li Z, He W. Mining bone metastasis related key genes of prostate cancer from the STING pathway based on machine learning. Front Med (Lausanne) 2024; 11:1372495. [PMID: 38835789 PMCID: PMC11148254 DOI: 10.3389/fmed.2024.1372495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/29/2024] [Indexed: 06/06/2024] Open
Abstract
Background Prostate cancer (PCa) is the second most prevalent malignant tumor in male, and bone metastasis occurs in about 70% of patients with advanced disease. The STING pathway, an innate immune signaling mechanism, has been shown to play a key role in tumorigenesis, metastasis, and cancerous bone pain. Hence, exploring regulatory mechanism of STING in PCa bone metastasis will bring novel opportunities for treating PCa bone metastasis. Methods First, key genes were screened from STING-related genes (SRGs) based on random forest algorithm and their predictive performance was evaluated. Subsequently, a comprehensive analysis of key genes was performed to explore their roles in prostate carcinogenesis, metastasis and tumor immunity. Next, cellular experiments were performed to verify the role of RELA in proliferation and migration in PCa cells, meanwhile, based on immunohistochemistry, we verified the difference of RELA expression between PCa primary foci and bone metastasis. Finally, based on the key genes to construct an accurate and reliable nomogram, and mined targeting drugs of key genes. Results In this study, three key genes for bone metastasis were mined from SRGs based on the random forest algorithm. Evaluation analysis showed that the key genes had excellent prediction performance, and it also showed that the key genes played a key role in carcinogenesis, metastasis and tumor immunity in PCa by comprehensive analysis. In addition, cellular experiments and immunohistochemistry confirmed that overexpression of RELA significantly inhibited the proliferation and migration of PCa cells, and RELA was significantly low-expression in bone metastasis. Finally, the constructed nomogram showed excellent predictive performance in Receiver Operating Characteristic (ROC, AUC = 0.99) curve, calibration curve, and Decision Curve Analysis (DCA) curve; and the targeted drugs showed good molecular docking effects. Conclusion In sum, this study not only provides a new theoretical basis for the mechanism of PCa bone metastasis, but also provides novel therapeutic targets and novel diagnostic tools for advanced PCa treatment.
Collapse
Affiliation(s)
- Guiqiang Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Urology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Runhan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhou Xie
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Qu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingtao Duan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yafei Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Liang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dagang Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zefang Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Qianjiang Hospital Affiliated with Chongqing University, Chongqing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Sun J, Tian T, Wang N, Jing X, Qiu L, Cui H, Liu Z, Liu J, Yan L, Li D. Pretreatment level of serum sialic acid predicts both qualitative and quantitative bone metastases of prostate cancer. Front Endocrinol (Lausanne) 2024; 15:1338420. [PMID: 38384968 PMCID: PMC10880016 DOI: 10.3389/fendo.2024.1338420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/18/2024] [Indexed: 02/23/2024] Open
Abstract
Background Recently, serum sialic acid (SA) has emerged as a distinct prognostic marker for prostate cancer (PCa) and bone metastases, warranting differential treatment and prognosis for low-volume (LVD) and high-volume disease (HVD). In clinical settings, evaluating bone metastases can prove advantageous. Objectives We aimed to establish the correlation between SA and both bone metastasis and HVD in newly diagnosed PCa patients. Methods We conducted a retrospective analysis of 1202 patients who received a new diagnosis of PCa between November 2014 and February 2021. We compared pretreatment SA levels across multiple groups and investigated the associations between SA levels and the clinical parameters of patients. Additionally, we compared the differences between HVD and LVD. We utilized several statistical methods, including the non-parametric Mann-Whitney U test, Spearman correlation, receiver operating characteristic (ROC) curve analysis, and logistic regression. Results The results indicate that SA may serve as a predictor of bone metastasis in patients with HVD. ROC curve analysis revealed a cut-off value of 56.15 mg/dL with an area under the curve of 0.767 (95% CI: 0.703-0.832, P < 0.001) for bone metastasis versus without bone metastasis and a cut-off value of 65.80 mg/dL with an area under the curve of 0.766 (95% CI: 0.644-0.888, P = 0.003) for HVD versus LVD. Notably, PCa patients with bone metastases exhibited significantly higher SA levels than those without bone metastases, and HVD patients had higher SA levels than LVD patients. In comparison to the non-metastatic and LVD cohorts, the cohort with HVD exhibited higher levels of alkaline phosphatase (AKP) (median, 122.00 U/L), fibrinogen (FIB) (median, 3.63 g/L), and prostate-specific antigen (PSA) (median, 215.70 ng/mL), as well as higher Gleason scores (> 7). Multivariate logistic regression analysis demonstrated that an SA level of > 56.15 mg/dL was independently associated with the presence of bone metastases in PCa patients (OR = 2.966, P = 0.018), while an SA level of > 65.80 mg/dL was independently associated with HVD (OR = 1.194, P = 0.048). Conclusion The pretreatment serum SA level is positively correlated with the presence of bone metastases.
Collapse
Affiliation(s)
- Jingtao Sun
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Tian Tian
- Respiratory and Critical Care Medicine Department, Qilu Hospital of Shandong University, Jinan, China
| | - Naiqiang Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xuehui Jing
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
- Department of Urology, Yucheng People’s Hospital, Dezhou, China
| | - Laiyuan Qiu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Haochen Cui
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Jikai Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Lei Yan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Dawei Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
10
|
Rehman K, Iqbal Z, Zhiqin D, Ayub H, Saba N, Khan MA, Yujie L, Duan L. Analysis of genetic biomarkers, polymorphisms in ADME-related genes and their impact on pharmacotherapy for prostate cancer. Cancer Cell Int 2023; 23:247. [PMID: 37858151 PMCID: PMC10585889 DOI: 10.1186/s12935-023-03084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/24/2023] [Indexed: 10/21/2023] Open
Abstract
Prostate cancer (PCa) is a non-cutaneous malignancy in males with wide variation in incidence rates across the globe. It is the second most reported cause of cancer death. Its etiology may have been linked to genetic polymorphisms, which are not only dominating cause of malignancy casualties but also exerts significant effects on pharmacotherapy outcomes. Although many therapeutic options are available, but suitable candidates identified by useful biomarkers can exhibit maximum therapeutic efficacy. The single-nucleotide polymorphisms (SNPs) reported in androgen receptor signaling genes influence the effectiveness of androgen receptor pathway inhibitors and androgen deprivation therapy. Furthermore, SNPs located in genes involved in transport, drug metabolism, and efflux pumps also influence the efficacy of pharmacotherapy. Hence, SNPs biomarkers provide the basis for individualized pharmacotherapy. The pharmacotherapeutic options for PCa include hormonal therapy, chemotherapy (Docetaxel, Mitoxantrone, Cabazitaxel, and Estramustine, etc.), and radiotherapy. Here, we overview the impact of SNPs reported in various genes on the pharmacotherapy for PCa and evaluate current genetic biomarkers with an emphasis on early diagnosis and individualized treatment strategy in PCa.
Collapse
Affiliation(s)
- Khurram Rehman
- Faculty of Pharmacy, Gomal University, D.I.Khan, Pakistan
| | - Zoya Iqbal
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, ShenzhenShenzhen, 518035, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China
| | - Deng Zhiqin
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, ShenzhenShenzhen, 518035, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China
| | - Hina Ayub
- Department of Gynae, Gomal Medical College, D.I.Khan, Pakistan
| | - Naseem Saba
- Department of Gynae, Gomal Medical College, D.I.Khan, Pakistan
| | | | - Liang Yujie
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, 518035, Guangdong, China.
| | - Li Duan
- Department of Orthopedics, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, ShenzhenShenzhen, 518035, Guangdong, China.
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital, Shenzhen, 518035, Guangdong, China.
| |
Collapse
|
11
|
Zeng Y, Pan Z, Yuan J, Song Y, Feng Z, Chen Z, Ye Z, Li Y, Bao Y, Ran Z, Li X, Ye H, Zhang K, Liu X, He Y. Inhibiting Osteolytic Breast Cancer Bone Metastasis by Bone-Targeted Nanoagent via Remodeling the Bone Tumor Microenvironment Combined with NIR-II Photothermal Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301003. [PMID: 37211708 DOI: 10.1002/smll.202301003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/08/2023] [Indexed: 05/23/2023]
Abstract
Bone is one of the prone metastatic sites of patients with advanced breast cancer. The "vicious cycle" between osteoclasts and breast cancer cells plays an essential role in osteolytic bone metastasis from breast cancer. In order to inhibit bone metastasis from breast cancer, NIR-II photoresponsive bone-targeting nanosystems (CuP@PPy-ZOL NPs) are designed and synthesized. CuP@PPy-ZOL NPs can trigger the photothermal-enhanced Fenton response and photodynamic effect to enhance the photothermal treatment (PTT) effect and thus achieve synergistic anti-tumor effect. Meanwhile, they exhibit a photothermal enhanced ability to inhibit osteoclast differentiation and promote osteoblast differentiation, which reshaped the bone microenvironment. CuP@PPy-ZOL NPs effectively inhibited the proliferation of tumor cells and bone resorption in the in vitro 3D bone metastases model of breast cancer. In a mouse model of breast cancer bone metastasis, CuP@PPy-ZOL NPs combined with PTT with NIR-II significantly inhibited the tumor growth of breast cancer bone metastases and osteolysis while promoting bone repair to achieve the reversal of osteolytic breast cancer bone metastases. Furthermore, the potential biological mechanisms of synergistic treatment are identified by conditioned culture experiments and mRNA transcriptome analysis. The design of this nanosystem provides a promising strategy for treating osteolytic bone metastases.
Collapse
Affiliation(s)
- Yaoxun Zeng
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhenxing Pan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Jiongpeng Yuan
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yuqiong Song
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510300, P. R. China
| | - Zhenzhen Feng
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zefeng Chen
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, SAR, 999077, P. R. China
| | - Zhaoyi Ye
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yushan Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Ying Bao
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhili Ran
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xinyi Li
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Huiling Ye
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Kun Zhang
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xujie Liu
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Yan He
- Allan H. Conney Laboratory for Anticancer Research, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| |
Collapse
|
12
|
Khan M, Parshad S, Naimi MF, Sidhu AK, Lyons F, Hardisty MR, Whyne CM, Smoragiewicz M, Phillips CM, Briones J, Emmenegger U. Sarcopenia in Men With Bone-Predominant Metastatic Castration-Resistant Prostate Cancer Undergoing Ra-223 Therapy. Clin Genitourin Cancer 2023; 21:e228-e235.e1. [PMID: 36849325 DOI: 10.1016/j.clgc.2023.01.009] [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] [Received: 06/24/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Osteosarcopenia is the progressive loss of musculoskeletal structure and functionality, contributing to disability and mortality. Despite complex interactions between bone and muscle, osteosarcopenia prevention and treatment in men with metastatic castration-resistant prostate cancer (mCRPC) focuses predominantly on bone health. It is unknown whether Radium-223 (Ra-223) therapy affects sarcopenia. METHODS We identified 52 patients with mCRPC who had received Ra-223 and had a baseline plus ≥1 follow-up abdominopelvic CT scan. The total contour area (TCA) and averaged Hounsfield units (HU) of the left and right psoas muscles were obtained at the inferior L3 endplate, and the psoas muscle index (PMI) was calculated therefrom. Intrapatient musculoskeletal changes were analyzed across various time points. RESULTS TCA and PMI gradually declined over the study period (P = .002, P = .003, respectively), but Ra-223 therapy did not accelerate sarcopenia, nor the decline of HU compared to the pre-Ra-223 period. The median overall survival of patients with baseline sarcopenia was numerically worse (14.93 vs. 23.23 months, HR 0.612, P = .198). CONCLUSIONS Ra-223 does not accelerate sarcopenia. Thus, worsening muscle parameters in men with mCRPC undergoing Ra-223 therapy are attributable to other factors. Further research is needed to determine whether baseline sarcopenia predicts poor overall survival in such patients.
Collapse
Affiliation(s)
- Maira Khan
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada
| | - Shruti Parshad
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada
| | - Mahdi F Naimi
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada
| | - Amanjot K Sidhu
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada
| | - Frank Lyons
- Department of Orthopaedic Surgery, Mater University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - Michael R Hardisty
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Cari M Whyne
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Martin Smoragiewicz
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Cameron M Phillips
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Department of Medical Oncology, Niagara Health Systems, St. Catharines, Ontario, Canada
| | - Juan Briones
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada
| | - Urban Emmenegger
- Sunnybrook Research Institute and Odette Cancer Centre, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Janta S, Pranweerapaiboon K, Vivithanaporn P, Plubrukarn A, Chairoungdua A, Prasertsuksri P, Apisawetakan S, Chaithirayanon K. Holothurin A Inhibits RUNX1-Enhanced EMT in Metastasis Prostate Cancer via the Akt/JNK and P38 MAPK Signaling Pathway. Mar Drugs 2023; 21:345. [PMID: 37367670 DOI: 10.3390/md21060345] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Due to the challenge of prostate cancer (PCa) management, there has been a surge in efforts to identify more safe and effective compounds that can modulate the epithelial-mesenchymal transition (EMT) for driving metastasis. Holothurin A (HA), a triterpenoid saponin isolated from Holothuria scabra, has now been characterized for its diverse biological activities. However, the mechanisms of HA in EMT-driven metastasis of human PCa cell lines has not yet been investigated. Moreover, runt-related transcription factor 1 (RUNX1) acts as an oncogene in prostate cancer, but little is known about its role in the EMT. Thus, the purpose of this study was to determine how RUNX1 influences EMT-mediated metastasis, as well as the potential effect of HA on EMT-mediated metastasis in endogenous and exogenous RUNX1 expressions of PCa cell lines. The results demonstrated that RUNX1 overexpression could promote the EMT phenotype with increased EMT markers, consequently driving metastatic migration and invasion in PC3 cell line through the activation of Akt/MAPK signaling pathways. Intriguingly, HA treatment could antagonize the EMT program in endogenous and exogenous RUNX1-expressing PCa cell lines. A decreasing metastasis of both HA-treated cell lines was evidenced through a downregulation of MMP2 and MMP9 via the Akt/P38/JNK-MAPK signaling pathway. Overall, our approach first demonstrated that RUNX1 enhanced EMT-driven prostate cancer metastasis and that HA was capable of inhibiting the EMT and metastatic processes and should probably be considered as a candidate for metastasis PCa treatment.
Collapse
Affiliation(s)
- Sirorat Janta
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Kanta Pranweerapaiboon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10540, Thailand
| | - Anuchit Plubrukarn
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Songkhla 09112, Thailand
| | - Arthit Chairoungdua
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Somjai Apisawetakan
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Wattana, Bangkok 10110, Thailand
| | | |
Collapse
|
14
|
Zhang Q, Zhang P, Zhao Z, Wang J, Zhang H. Exploring the role of differentially expressed metabolic genes and their mechanisms in bone metastatic prostate cancer. PeerJ 2023; 11:e15013. [PMID: 37070095 PMCID: PMC10105558 DOI: 10.7717/peerj.15013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/16/2023] [Indexed: 04/19/2023] Open
Abstract
Background Approximately 10-20% of patients diagnosed with prostate cancer (PCa) evolve into castration-resistant prostate cancer (CRPC), while nearly 90% of patients with metastatic CRPC (mCRPC) exhibit osseous metastases (BM). These BM are intimately correlated with the stability of the tumour microenvironment. Purpose This study aspires to uncover the metabolism-related genes and the underlying mechanisms responsible for bone metastatic prostate cancer (BMPCa). Methods Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets of PCa and BM were analyzed through R Studio software to identify differentially expressed genes (DEGs). The DEGs underwent functional enrichment via Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO), with key factors screened by a random forest utilized to establish a prognostic model for PCa. The study explored the relationship between DEGs and the stability of the immune microenvironment. The action and specificity of CRISP3 in PCa was validated through western blot analysis, CCK-8 assay, scratch assay, and cellular assay. Results The screening of GEO and TCGA datasets resulted in the identification of 199 co-differential genes. Three DEGs, including DES, HBB, and SLPI, were selected by random forest classification model and cox regression model. Immuno-infiltration analysis disclosed that a higher infiltration of naïve B cells and resting CD4 memory T cells occurred in the high-expression group of DES, whereas infiltration of resting M1 macrophages and NK cells was greater in the low-expression group of DES. A significant infiltration of neutrophils was observed in the high-expression group of HBB, while greater infiltration of gamma delta T cells and M1 macrophages was noted in the low-expression group of HBB. Resting dendritic cells, CD8 T cells, and resting T regulatory cells (Tregs) infiltrated significantly in the high-expression group of SLPI, while only resting mast cells infiltrated significantly in the low-expression group of SLPI. CRISP3 was established as a critical gene in BMPCa linked to DES expression. Targeting CRISP3, d-glucopyranose may impact tumour prognosis. During the mechanistic experiments, it was established that CRISP3 can advance the proliferation and metastatic potential of PCa by advancing epithelial-to-mesenchymal transition (EMT). Conclusion By modulating lipid metabolism and maintaining immunological and microenvironmental balance, DES, HBB, and SLPI suppress prostate cancer cell growth. The presence of DES-associated CRISP3 is a harbinger of unfavorable outcomes in prostate cancer and may escalate tumor proliferation and metastatic capabilities by inducing epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Qingfu Zhang
- Department of Urology, Tai ’an Central Hospital, Tai ’an, Shandong, China
| | - Peng Zhang
- Department of Spine Surgery, Tai ’an Central Hospital, Tai ’an, Shandong, China
| | - Zhongting Zhao
- Department of Spinal Surgery, The Third People’s Hospital of Jinan, Jinan, Shandong, China
| | - Jun Wang
- Department of Emergency, Qingdao Eighth People’s Hospital, Qingdao, China
| | - Hepeng Zhang
- Department of Urology, Tai ’an Central Hospital, Tai ’an, Shandong, China
| |
Collapse
|
15
|
Hegde M, Naliyadhara N, Unnikrishnan J, Alqahtani MS, Abbas M, Girisa S, Sethi G, Kunnumakkara AB. Nanoparticles in the diagnosis and treatment of cancer metastases: Current and future perspectives. Cancer Lett 2023; 556:216066. [PMID: 36649823 DOI: 10.1016/j.canlet.2023.216066] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Metastasis accounts for greater than 90% of cancer-related deaths. Despite recent advancements in conventional chemotherapy, immunotherapy, targeted therapy, and their rational combinations, metastatic cancers remain essentially untreatable. The distinct obstacles to treat metastases include their small size, high multiplicity, redundancy, therapeutic resistance, and dissemination to multiple organs. Recent advancements in nanotechnology provide the numerous applications in the diagnosis and prophylaxis of metastatic diseases, including the small particle size to penetrate cell membrane and blood vessels and their capacity to transport complex molecular 'cargo' particles to various metastatic regions such as bones, brain, liver, lungs, and lymph nodes. Indeed, nanoparticles (NPs) have demonstrated a significant ability to target specific cells within these organs. In this regard, the purpose of this review is to summarize the present state of nanotechnology in terms of its application in the diagnosis and treatment of metastatic cancer. We intensively reviewed applications of NPs in fluorescent imaging, PET scanning, MRI, and photoacoustic imaging to detect metastasis in various cancer models. The use of targeted NPs for cancer ablation in conjunction with chemotherapy, photothermal treatment, immuno therapy, and combination therapy is thoroughly discussed. The current review also highlights the research opportunities and challenges of leveraging engineering technologies with cancer cell biology and pharmacology to fabricate nanoscience-based tools for treating metastases.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nikunj Naliyadhara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Jyothsna Unnikrishnan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia; Computers and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa, 35712, Egypt
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
16
|
Bonanni D, Pinzi L, Rastelli G. Development of machine learning classifiers to predict compound activity on prostate cancer cell lines. J Cheminform 2022; 14:77. [PMID: 36348374 PMCID: PMC9641853 DOI: 10.1186/s13321-022-00647-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022] Open
Abstract
Prostate cancer is the most common type of cancer in men. The disease presents good survival rates if treated at the early stages. However, the evolution of the disease in its most aggressive variant remains without effective therapeutic answers. Therefore, the identification of novel effective therapeutics is urgently needed. On these premises, we developed a series of machine learning models, based on compounds with reported highly homogeneous cell-based antiproliferative assay data, able to predict the activity of ligands towards the PC-3 and DU-145 prostate cancer cell lines. The data employed in the development of the computational models was finely-tuned according to a series of thresholds for the classification of active/inactive compounds, to the number of features to be implemented, and by using 10 different machine learning algorithms. Models’ evaluation allowed us to identify the best combination of activity thresholds and ML algorithms for the classification of active compounds, achieving prediction performances with MCC values above 0.60 for PC-3 and DU-145 cells. Moreover, in silico models based on the combination of PC-3 and DU-145 data were also developed, demonstrating excellent precision performances. Finally, an analysis of the activity annotations reported for the ligands in the curated datasets were conducted, suggesting associations between cellular activity and biological targets that might be explored in the future for the design of more effective prostate cancer antiproliferative agents.
Collapse
|
17
|
Tong Y, Cao Y, Jin T, Huang Z, He Q, Mao M. Role of Interleukin-1 family in bone metastasis of prostate cancer. Front Oncol 2022; 12:951167. [PMID: 36237303 PMCID: PMC9552844 DOI: 10.3389/fonc.2022.951167] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022] Open
Abstract
Prostate cancer (PCa) is one of the most fatal diseases in male patients with high bone metastatic potential. Bone metastasis severely shortens overall survival and brings skeletal-related events (SREs) which reduces the life quality of patients, and this situation is currently regarded as irreversible and incurable. The progression and metastasis of PCa are found to be closely associated with inflammatory cytokines and chemokines. As pivotal members of inflammatory cytokines, Interleukin-1 (IL-1) family plays a crucial role in this process. Elevated expression of IL-1 family was detected in PCa patients with bone metastasis, and accumulating evidences proved that IL-1 family could exert vital effects on the progression and bone metastasis of many cancers, while some members have dual effects. In this review, we discuss the role of IL-1 family in the bone metastasis of PCa. Furthermore, we demonstrate that many members of IL-1 family could act as pivotal biomarkers to predict the clinical stage and prognosis of PCa patients. More importantly, we have elucidated the role of IL-1 family in the bone metastasis of PCa, which could provide potential targets for the treatment of PCa bone metastasis and probable directions for future research.
Collapse
Affiliation(s)
- Yuanhao Tong
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinghao Cao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianzhe Jin
- Department of Gynecologic Oncology, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengwei Huang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Qinyuan He
- Organization Department, Suzhou Traditional Chinese Medicine Hospital, Suzhou, China
| | - Min Mao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Min Mao,
| |
Collapse
|
18
|
Solimando AG, Kalogirou C, Krebs M. Angiogenesis as Therapeutic Target in Metastatic Prostate Cancer - Narrowing the Gap Between Bench and Bedside. Front Immunol 2022; 13:842038. [PMID: 35222436 PMCID: PMC8866833 DOI: 10.3389/fimmu.2022.842038] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis in metastatic castration-resistant prostate cancer (mCRPC) has been extensively investigated as a promising druggable biological process. Nonetheless, targeting angiogenesis has failed to impact overall survival (OS) in patients with mCRPC despite promising preclinical and early clinical data. This discrepancy prompted a literature review highlighting the tumor heterogeneity and biological context of Prostate Cancer (PCa). Narrowing the gap between the bench and bedside appears critical for developing novel therapeutic strategies. Searching clinicaltrials.gov for studies examining angiogenesis inhibition in patients with PCa resulted in n=20 trials with specific angiogenesis inhibitors currently recruiting (as of September 2021). Moreover, several other compounds with known anti-angiogenic properties - such as Metformin or Curcumin - are currently investigated. In general, angiogenesis-targeting strategies in PCa include biomarker-guided treatment stratification - as well as combinatorial approaches. Beyond established angiogenesis inhibitors, PCa therapies aiming at PSMA (Prostate Specific Membrane Antigen) hold the promise to have a substantial anti-angiogenic effect - due to PSMA´s abundant expression in tumor vasculature.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine "G. Baccelli", University of Bari Medical School, Bari, Italy.,Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany
| | - Markus Krebs
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Moretti A. What are the efficacy and safety of bisphosphonates and RANK-ligand-inhibitors for men with prostate cancer and bone metastases? - A Cochrane Review summary with commentary. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2021; 21:451-454. [PMID: 34854383 PMCID: PMC8672400 DOI: pmid/34854383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Antimo Moretti
- Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Italy
- Corresponding author: Antimo Moretti, Department of Medical and Surgical Specialties and Dentistry, University of Campania “Luigi Vanvitelli”, Italy E-mail:
| |
Collapse
|
20
|
Fonseca NM, Roberts ME, Wyatt AW. A marrow-minded look at immune checkpoint blockade resistance in metastatic castration resistant prostate cancer. Transl Androl Urol 2021; 10:4009-4013. [PMID: 34804843 PMCID: PMC8575591 DOI: 10.21037/tau-20-1205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/30/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Nicolette M Fonseca
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Morgan E Roberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Endolysosomal ion channel MCOLN2 (Mucolipin-2) promotes prostate cancer progression via IL-1β/NF-κB pathway. Br J Cancer 2021; 125:1420-1431. [PMID: 34548638 PMCID: PMC8575885 DOI: 10.1038/s41416-021-01537-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/13/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prostate cancer (Pca) is the most common cancer type among males worldwide. Dysregulation of Ca2+ signaling plays important roles during Pca progression. However, there is lack of information about the role of endolysosomal Ca2+ -permeable channels in Pca progression. METHODS The expression pattern of MCOLN2 was studied by immunohistochemistry and western blot. Cell viability assay, transwell assay and in vivo tumorigenesis were performed to evaluate the functional role of MCOLN2. Downstream targets of MCOLN2 were investigated by cytokine array, enzyme-linked immunosorbent assay, Ca2+ release experiments and luciferase reporter assays. RESULTS We report that MCOLN2 expression is significantly elevated in Pca tissues, and associated with poor prognosis. Overexpression of MCOLN2 promoted Pca cells proliferation, migration and invasion. Importantly, knockdown of MCOLN2 inhibited Pca xenograft tumor growth and bone lesion development in vivo. In addition, MCOLN2 promoted the production and release of IL-1β. Moreover, luciferase reporter assay and western blot revealed that MCOLN2 promoted Pca development by regulating the IL-1β/NF-κB pathway. CONCLUSION In summary, MCOLN2 is crucially involved in Pca progression. Mechanistically, MCOLN2 regulates Pca progression via IL-1β/NF-κB pathway. Our study highlights an intriguing possibility of targeting MCOLN2 as potential therapeutic strategy in Pca treatment.
Collapse
|
22
|
Ban J, Fock V, Aryee DNT, Kovar H. Mechanisms, Diagnosis and Treatment of Bone Metastases. Cells 2021; 10:2944. [PMID: 34831167 PMCID: PMC8616226 DOI: 10.3390/cells10112944] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Bone and bone marrow are among the most frequent metastatic sites of cancer. The occurrence of bone metastasis is frequently associated with a dismal disease outcome. The prevention and therapy of bone metastases is a priority in the treatment of cancer patients. However, current therapeutic options for patients with bone metastatic disease are limited in efficacy and associated with increased morbidity. Therefore, most current therapies are mainly palliative in nature. A better understanding of the underlying molecular pathways of the bone metastatic process is warranted to develop novel, well-tolerated and more successful treatments for a significant improvement of patients' quality of life and disease outcome. In this review, we provide comparative mechanistic insights into the bone metastatic process of various solid tumors, including pediatric cancers. We also highlight current and innovative approaches to biologically targeted therapy and immunotherapy. In particular, we discuss the role of the bone marrow microenvironment in the attraction, homing, dormancy and outgrowth of metastatic tumor cells and the ensuing therapeutic implications. Multiple signaling pathways have been described to contribute to metastatic spread to the bone of specific cancer entities, with most knowledge derived from the study of breast and prostate cancer. However, it is likely that similar mechanisms are involved in different types of cancer, including multiple myeloma, primary bone sarcomas and neuroblastoma. The metastatic rate-limiting interaction of tumor cells with the various cellular and noncellular components of the bone-marrow niche provides attractive therapeutic targets, which are already partially exploited by novel promising immunotherapies.
Collapse
Affiliation(s)
- Jozef Ban
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Valerie Fock
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
| | - Dave N. T. Aryee
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (J.B.); (V.F.); (D.N.T.A.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
23
|
van der Zande K, Oyen WJG, Zwart W, Bergman AM. Radium-223 Treatment of Patients with Metastatic Castration Resistant Prostate Cancer: Biomarkers for Stratification and Response Evaluation. Cancers (Basel) 2021; 13:cancers13174346. [PMID: 34503156 PMCID: PMC8431634 DOI: 10.3390/cancers13174346] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Radium-223 dichloride ([223Ra]RaCl2; Ra-223) is an alpha-emitting radiopharmaceutical treatment for patients with metastatic castration resistant prostate cancer (mCRPC) with predominantly bone metastases. While responses to chemotherapeutic and antihormonal mCRPC treatments can be assessed by serum PSA levels, a decrease of serum PSA levels is not expected during Ra-223 therapy. Moreover, radiographic evaluation of bone metastases response is challenging. Therefore, novel biomarkers to select patients for Ra-223 treatment and monitoring response are urgently needed. In this review, we discuss the currently used and exploratory biomarkers for this purpose, including soluble and cellular factors detected in the peripheral blood, genetic defects and radiographic assessments. We conclude that some biomarkers, including metabolic products of collagen degradation and novel PET scan techniques, might hold promise as predictors of response to Ra-223 treatment. However, these biomarkers have not been extensively studied. Consequently, currently, no biomarker has established a place in patient stratification and response evaluation. Abstract Radium-223 dichloride ([223Ra]RaCl2; Ra-223) is a targeted alpha-emitting radiopharmaceutical which results in an overall survival and health related quality of life (HRQoL) benefit in symptomatic patients with metastatic castration resistant prostate cancer (mCRPC) and predominantly bone metastasis. Although effective, options to select patients who will derive treatment benefit and to monitor and predict treatment outcomes are limited. PSA response and radiographic evaluation are commonly used in mCRPC treatment assessment but are not informative in Ra-223 treated patients. Consequently, there is a clear need for predictive and prognostic tools. In this review, we discuss the physiology of bone metastases and the mechanism of action and efficacy of Ra-223 treatment, as well as offering an outline of current innovative prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Kim van der Zande
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Wim J. G. Oyen
- Department of Nuclear Medicine, Rijnstate Hospital, Wagnerlaan 55, 6815 AD Arnhem, The Netherlands;
| | - Wilbert Zwart
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Correspondence: (W.Z.); (A.M.B.); Tel.: +31-2051-28156 (W.Z.); +31-2051-22569 (A.M.B.)
| | - Andries M. Bergman
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Correspondence: (W.Z.); (A.M.B.); Tel.: +31-2051-28156 (W.Z.); +31-2051-22569 (A.M.B.)
| |
Collapse
|
24
|
Lovell S, Zhang L, Kryza T, Neodo A, Bock N, De Vita E, Williams ED, Engelsberger E, Xu C, Bakker AT, Maneiro M, Tanaka RJ, Bevan CL, Clements JA, Tate EW. A Suite of Activity-Based Probes To Dissect the KLK Activome in Drug-Resistant Prostate Cancer. J Am Chem Soc 2021; 143:8911-8924. [PMID: 34085829 PMCID: PMC9282638 DOI: 10.1021/jacs.1c03950] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Kallikrein-related
peptidases (KLKs) are a family of secreted serine
proteases, which form a network (the KLK activome) with an important
role in proteolysis and signaling. In prostate cancer (PCa), increased
KLK activity promotes tumor growth and metastasis through multiple
biochemical pathways, and specific quantification and tracking of
changes in the KLK activome could contribute to validation of KLKs
as potential drug targets. Herein we report a technology platform
based on novel activity-based probes (ABPs) and inhibitors enabling
simultaneous orthogonal analysis of KLK2, KLK3, and KLK14 activity
in hormone-responsive PCa cell lines and tumor homogenates. Importantly,
we identifed a significant decoupling of KLK activity and abundance
and suggest that KLK proteolysis should be considered as an additional
parameter, along with the PSA blood test, for accurate PCa diagnosis
and monitoring. Using selective inhibitors and multiplexed fluorescent
activity-based protein profiling (ABPP), we dissect the KLK activome
in PCa cells and show that increased KLK14 activity leads to a migratory
phenotype. Furthermore, using biotinylated ABPs, we show that active
KLK molecules are secreted into the bone microenvironment by PCa cells
following stimulation by osteoblasts suggesting KLK-mediated signaling
mechanisms could contribute to PCa metastasis to bone. Together our
findings show that ABPP is a powerful approach to dissect dysregulation
of the KLK activome as a promising and previously underappreciated
therapeutic target in advanced PCa.
Collapse
Affiliation(s)
- Scott Lovell
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Leran Zhang
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Thomas Kryza
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Anna Neodo
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elena De Vita
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elisabeth Engelsberger
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Congyi Xu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Alexander T Bakker
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Maria Maneiro
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Charlotte L Bevan
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.,The Francis Crick Institute, London NW1 1AT, U.K
| |
Collapse
|
25
|
Bock N, Kryza T, Shokoohmand A, Röhl J, Ravichandran A, Wille ML, Nelson CC, Hutmacher DW, Clements JA. In vitro engineering of a bone metastases model allows for study of the effects of antiandrogen therapies in advanced prostate cancer. SCIENCE ADVANCES 2021; 7:eabg2564. [PMID: 34193425 PMCID: PMC8245033 DOI: 10.1126/sciadv.abg2564] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/17/2021] [Indexed: 05/05/2023]
Abstract
While androgen-targeted therapies are routinely used in advanced prostate cancer (PCa), their effect is poorly understood in treating bone metastatic lesions and ultimately results in the development of metastatic castrate resistant prostate cancer (mCRPC). Here, we used an all-human microtissue-engineered model of mineralized metastatic tissue combining human osteoprogenitor cells, 3D printing and prostate cancer cells, to assess the effects of the antiandrogens, bicalutamide, and enzalutamide in this microenvironment. We demonstrate that cancer/bone stroma interactions and antiandrogens drive cancer progression in a mineralized microenvironment. Probing the bone microenvironment with enzalutamide led to stronger cancer cell adaptive responses and osteomimicry than bicalutamide. Enzalutamide presented with better treatment response, in line with enzalutamide delaying time to bone-related events and enzalutamide extending survival in mCRPC. The all-human microtissue-engineered model of mineralized metastatic tissue presented here represents a substantial advance to dissect the role of the bone tumor microenvironment and responses to therapies for mCPRC.
Collapse
Affiliation(s)
- Nathalie Bock
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, 4059 QLD, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), QUT, Kelvin Grove, 4059 QLD, Australia
| | - Thomas Kryza
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
| | - Ali Shokoohmand
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, 4059 QLD, Australia
| | - Joan Röhl
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
| | - Akhilandeshwari Ravichandran
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, 4059 QLD, Australia
| | - Marie-Luise Wille
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), QUT, Kelvin Grove, 4059 QLD, Australia
- Bone and Joint Disorders Program, School of Mechanical Medical, and Process Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, 4000 QLD, Australia
| | - Colleen C Nelson
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
| | - Dietmar W Hutmacher
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia.
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, 4059 QLD, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D Innovation), QUT, Kelvin Grove, 4059 QLD, Australia
- Bone and Joint Disorders Program, School of Mechanical Medical, and Process Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, 4000 QLD, Australia
- ARC Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, 4059 QLD, Australia
| | - Judith A Clements
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Brisbane 4000, QLD, Australia.
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
- Translational Research Institute (TRI), QUT, Woolloongabba, 4102 QLD, Australia
| |
Collapse
|
26
|
Marques IA, Abrantes AM, Pires AS, Neves AR, Caramelo FJ, Rodrigues T, Matafome P, Tavares-da-Silva E, Gonçalves AC, Pereira CC, Teixeira JP, Seiça R, Costa G, Figueiredo A, Botelho MF. Kinetics of radium-223 and its effects on survival, proliferation and DNA damage in lymph-node and bone metastatic prostate cancer cell lines. Int J Radiat Biol 2021; 97:714-726. [PMID: 33764249 DOI: 10.1080/09553002.2021.1906462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is associated with a very unfavorable prognosis. At this advanced stage of the disease, there are several therapeutic strategies approved in recent times, being one of them Radium-223 dichloride (Radium-223). However, its mechanisms of action and the process that conducts to cell death are not fully understood. Given this, our main goal is to characterize the radiobiological effects induced by Radium-223 and to evaluate its kinetics on metastatic Prostate Cancer (mPCa) cells. MATERIALS AND METHODS In vitro studies were conducted using two mPCa cell lines, the LNCaP and PC3, the first being derived from lymph node metastasis and the second from bone metastasis. Kinetic studies were conducted to access the capacity of these cell lines to uptake, retain and internalize the Radium-223. For the assessment of radiobiological effects, cells were first exposed to different doses of Radium-223 and the clonogenic assay was done to evaluate cell survival and to determine lethal doses (LD50). Then, the effects were also evaluated in terms of proliferation, oxidative stress, morphological changes and cell damage. RESULTS Radium-223 is uptaken by mPCa cells and reaches the nucleus, where it is retained over time. Irradiation decreases cell survival and proliferation, with LNCaP cells (LD50 = 1.73mGy) being more radiosensitive than PC3 cells (LD50 = 4.20mGy). Irradiated cells showed morphological changes usually associated with apoptosis and a dose-dependent increase in DNA damage. Moreover, activation of cell cycle checkpoints occurs through ATM/CHK2 pathway, which is involved in cell cycle arrest and cell death. CONCLUSIONS The cytotoxic and anti-proliferative effects on both cell lines showed that Radium-223 can decrease the aggressiveness of tumor cells by decreasing the cell survival and proliferation and, also, by increasing the DNA damage. The similar results observed in both cell lines indicated that Radium-223 may have the potential to be used as a therapeutic option also for mCRPC patients with lymph node metastasis. The activation of DNA Damage Response pathways allows the possibility to understand the importance of these checkpoints as targets for new combined therapies.
Collapse
Affiliation(s)
- Inês A Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Ana M Abrantes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana S Pires
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana R Neves
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Project Development Office, Department of Mathematics and Computer Science, Eindhoven University of Technology (TU/e), Eindhoven, The Netherlands
| | - Francisco J Caramelo
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Biostatistics and Medical Informatics of Faculty of Medicine, Coimbra, Portugal
| | - Tiago Rodrigues
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Paulo Matafome
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Edgar Tavares-da-Silva
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Urology and Renal Transplantation, Coimbra, Portugal
| | - Ana C Gonçalves
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Oncobiology and Hematology and University Clinic of Hematology of Faculty of Medicine, Coimbra, Portugal
| | - Cristiana C Pereira
- National Institute of Health, Environmental Health Department, Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - João P Teixeira
- National Institute of Health, Environmental Health Department, Porto, Portugal
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
| | - Raquel Seiça
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Coimbra, Portugal
| | - Grancinda Costa
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Nuclear Medicine, Coimbra, Portugal
| | - Arnaldo Figueiredo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Centro Hospitalar e Universitário de Coimbra (CHUC), Department of Urology and Renal Transplantation, Coimbra, Portugal
| | - Maria F Botelho
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
27
|
Zhang B, Li Y, Wu Q, Xie L, Barwick B, Fu C, Li X, Wu D, Xia S, Chen J, Qian WP, Yang L, Osunkoya AO, Boise L, Vertino PM, Zhao Y, Li M, Chen HR, Kowalski J, Kucuk O, Zhou W, Dong JT. Acetylation of KLF5 maintains EMT and tumorigenicity to cause chemoresistant bone metastasis in prostate cancer. Nat Commun 2021; 12:1714. [PMID: 33731701 PMCID: PMC7969754 DOI: 10.1038/s41467-021-21976-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/18/2021] [Indexed: 12/25/2022] Open
Abstract
Advanced prostate cancer (PCa) often develops bone metastasis, for which therapies are very limited and the underlying mechanisms are poorly understood. We report that bone-borne TGF-β induces the acetylation of transcription factor KLF5 in PCa bone metastases, and acetylated KLF5 (Ac-KLF5) causes osteoclastogenesis and bone metastatic lesions by activating CXCR4, which leads to IL-11 secretion, and stimulating SHH/IL-6 paracrine signaling. While essential for maintaining the mesenchymal phenotype and tumorigenicity, Ac-KLF5 also causes resistance to docetaxel in tumors and bone metastases, which is overcome by targeting CXCR4 with FDA-approved plerixafor. Establishing a mechanism for bone metastasis and chemoresistance in PCa, these findings provide a rationale for treating chemoresistant bone metastasis of PCa with inhibitors of Ac-KLF5/CXCR4 signaling.
Collapse
Affiliation(s)
- Baotong Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yixiang Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Qiao Wu
- Department of Genetics and Cell Biology, Nankai University College of Life Sciences, Tianjin, China
| | - Lin Xie
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Third Affiliated Hospital of Kunming Medical University, Cancer Hospital of Yunnan Province, Kunming, China
| | - Benjamin Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Changying Fu
- Department of Genetics and Cell Biology, Nankai University College of Life Sciences, Tianjin, China
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China
| | - Xin Li
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Daqing Wu
- Molecular Oncology and Biomarkers Program, Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Siyuan Xia
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jing Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wei Ping Qian
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Adeboye O Osunkoya
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Pathology and Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lawrence Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Paula M Vertino
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yichao Zhao
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Menglin Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hsiao-Rong Chen
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jeanne Kowalski
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jin-Tang Dong
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology School of Medicine, Shenzhen, China.
| |
Collapse
|
28
|
Tan X, Chen WB, Lv DJ, Yang TW, Wu KH, Zou LB, Luo J, Zhou XM, Liu GC, Shu FP, Mao XM. LncRNA SNHG1 and RNA binding protein hnRNPL form a complex and coregulate CDH1 to boost the growth and metastasis of prostate cancer. Cell Death Dis 2021; 12:138. [PMID: 33542227 PMCID: PMC7862296 DOI: 10.1038/s41419-021-03413-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
The interaction between LncRNA and RNA-binding protein (RBPs) plays an essential role in the regulation over the malignant progression of tumors. Previous studies on the mechanism of SNHG1, an emerging lncRNA, have primarily focused on the competing endogenous RNA (ceRNA) mechanism. Nevertheless, the underlying mechanism between SNHG1 and RBPs in tumors remains to be explored, especially in prostate cancer (PCa). SNHG1 expression profiles in PCa were determined through the analysis of TCGA data and tissue microarray at the RNA level. Gain- and loss-of-function experiments were performed to investigate the biological role of SNHG1 in PCa initiation and progression. RNA-seq, immunoblotting, RNA pull-down and RNA immunoprecipitation analyses were utilized to clarify potential pathways with which SNHG1 might be involved. Finally, rescue experiments were carried out to further confirm this mechanism. We found that SNHG1 was dominantly expressed in the nuclei of PCa cells and significantly upregulated in PCa patients. The higher expression level of SNHG1 was dramatically correlated with tumor metastasis and patient survival. Functionally, overexpression of SNHG1 in PCa cells induced epithelial-mesenchymal transition (EMT), accompanied by down-regulation of the epithelial marker, E-cadherin, and up-regulation of the mesenchymal marker, vimentin. Increased proliferation and migration, as well as accelerated xenograft tumor growth, were observed in SNHG1-overexpressing PCa cells, while opposite effects were achieved in SNHG1-silenced cells. Mechanistically, SNHG1 competitively interacted with hnRNPL to impair the translation of protein E-cadherin, thus activating the effect of SNHG1 on the EMT pathway, eventually promoting the metastasis of PCa. Our findings demonstrate that SNHG1 is a positive regulator of EMT activation through the SNHG1-hnRNPL-CDH1 axis. SNHG1 may serve as a novel potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wen-Bin Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Dao-Jun Lv
- Department of Urology, Minimally Invasive Surgery Center, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Guangzhou, Guangdong, China
| | - Tao-Wei Yang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kai-Hui Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Li-Bin Zou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junqi Luo
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xu-Min Zhou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guo-Chang Liu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Fang-Peng Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Xiang-Ming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Mollica V, Rizzo A, Rosellini M, Marchetti A, Ricci AD, Cimadamore A, Scarpelli M, Bonucci C, Andrini E, Errani C, Santoni M, Montironi R, Massari F. Bone Targeting Agents in Patients with Metastatic Prostate Cancer: State of the Art. Cancers (Basel) 2021; 13:cancers13030546. [PMID: 33535541 PMCID: PMC7867059 DOI: 10.3390/cancers13030546] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 02/03/2023] Open
Abstract
Simple Summary Over the disease course of metastatic prostate cancer, approximately the 90% of patients develops bone metastases, with bone involvement frequently leading to various skeletal complications including pathological fractures, spinal cord compression, and pain. Notably enough, the peculiar inclination of prostate cancer cells to seed the bone is considered an important cause of morbidity for prostate cancer patients. Recent years have witnessed the advent of several novel treatments for prostate cancer, and therapeutic paradigms are rapidly shifting. In this review, we aim at giving an overview of current knowledge on the relationship between prostate cancer and bone, especially focusing on the use of bone-targeted agents in this setting. Abstract Bone health represents a major issue in castration-resistant prostate cancer (CRPC) patients with bone metastases; in fact, the frequently prolonged use of hormonal agents causes important modifications in physiological bone turnover and most of these men will develop skeletal-related events (SREs), including spinal cord compression, pathologic fractures and need for surgery or radiation to bone, which are estimated to occur in almost half of this patient population. In the last decade, several novel therapeutic options have entered into clinical practice of bone metastatic CRPC, with recent approval of enzalutamide and abiraterone acetate, cabazitaxel chemotherapy and radium-223, on the basis of survival benefit suggested by landmark Phase III trials assessing these agents in this setting. Conversely, although bone-targeted agents (BTAs)—such as the bisphosphonate zoledronic acid and the receptor activator of nuclear factor kappa-B (RANK) ligand inhibitor denosumab—are approved for the prevention of SREs, these compounds have not shown benefit in terms of overall survival. However, emerging evidence has suggested that the combination of BTAs and abiraterone acetate, enzalutamide and the radiopharmaceutical radium-223 could result in improved clinical outcomes and prolonged survival in bone metastatic CRPC. In this review, we will provide an overview on bone tropism of prostate cancer and on the role of BTAs in metastatic hormone-sensitive and castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Veronica Mollica
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Alessandro Rizzo
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Matteo Rosellini
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Andrea Marchetti
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Angela Dalia Ricci
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60121 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60121 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Chiara Bonucci
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Elisa Andrini
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
| | - Costantino Errani
- Department of Musculo–Skeletal Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60121 Ancona, Italy; (A.C.); (M.S.); (R.M.)
| | - Francesco Massari
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, 40138 Bologna, Italy; (V.M.); (A.R.); (M.R.); (A.M.); (A.D.R.); (C.B.); (E.A.)
- Correspondence: or
| |
Collapse
|
30
|
Abstract
Theranostics in men with metastatic castration-resistant prostate cancer (mCRPC) has been developed to target bone and the tumor itself. Currently, bone-directed targeted alpha therapy with radium-223 (223Ra) is the only theranostic agent proven to prolong survival in men with mCRPC who have symptomatic bone metastases and no known visceral metastases. The clinical utility and therapeutic success of 223Ra has encouraged the development of other tumor-targeting theranostic agents in mCRPC, primarily targeting prostate-specific membrane antigen (PSMA) with radioligand therapy (RLT). There is increasing evidence of promising response rates and a low toxicity profile with 177Lu-labeled PSMA RLT in patients with mCRPC. A phase III randomized study of 177Lu-labeled PSMA RLT has completed accrual and is awaiting results as to whether the drug improves radiographic progression-free survival and overall survival in men with mCRPC receiving standard of care treatments. Additional early clinical trials are investigating the role of tumor-directed targeted alpha therapy with radiotracers such as 225Ac. In this article, we review the current status of theranostics in prostate cancer, discussing the challenges and opportunities of combination therapies with more conventional agents such as androgen receptor inhibitors, cytotoxic chemotherapy, and immunotherapy.
Collapse
|
31
|
Ordikhani F, Zandi N, Mazaheri M, Luther GA, Ghovvati M, Akbarzadeh A, Annabi N. Targeted nanomedicines for the treatment of bone disease and regeneration. Med Res Rev 2020; 41:1221-1254. [PMID: 33347711 DOI: 10.1002/med.21759] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 10/14/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022]
Abstract
Targeted delivery by either passive or active targeting of therapeutics to the bone is an attractive treatment for various bone related diseases such as osteoporosis, osteosarcoma, multiple myeloma, and metastatic bone tumors. Engineering novel drug delivery carriers can increase therapeutic efficacy and minimize the risk of side effects. Developmnet of nanocarrier delivery systems is an interesting field of ongoing studies with opportunities to provide more effective therapies. In addition, preclinical nanomedicine research can open new opportunities for preclinical bone-targeted drug delivery; nevertheless, further research is needed to progress these therapies towards clinical applications. In the present review, the latest advancements in targeting moieties and nanocarrier drug delivery systems for the treatment of bone diseases are summarized. We also review the regeneration capability and effective delivery of nanomedicines for orthopedic applications.
Collapse
Affiliation(s)
- Farideh Ordikhani
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nooshin Zandi
- Institute for Nanoscience and Nanotechnology, Sharif University of Technology, Tehran, Iran.,Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Mozhdeh Mazaheri
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | - Gaurav A Luther
- Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahsa Ghovvati
- Department of Chemical and Biomolecular Engineering, University of California- Los Angeles, California, Los Angeles, USA
| | - Abolfazl Akbarzadeh
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.,Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California- Los Angeles, California, Los Angeles, USA
| |
Collapse
|
32
|
Heidegger I, Necchi A, Pircher A, Tsaur I, Marra G, Kasivisvanathan V, Kretschmer A, Mathieu R, Ceci F, van den Bergh RCN, Thibault C, Tilki D, Valerio M, Surcel C, Gandaglia G. A Systematic Review of the Emerging Role of Immune Checkpoint Inhibitors in Metastatic Castration-resistant Prostate Cancer: Will Combination Strategies Improve Efficacy? Eur Urol Oncol 2020; 4:745-754. [PMID: 33243663 DOI: 10.1016/j.euo.2020.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022]
Abstract
CONTEXT The role of immune checkpoint inhibition (ICI) in the treatment of prostate cancer (PC) still remains elusive. It has been proposed that combination of ICI with other molecules increases the efficacy of immunotherapy in PC. OBJECTIVE To systematically review the literature to assess the potential role of ICI in combination with additional therapies for the management of metastatic castration-resistant PC (mCRPC). EVIDENCE ACQUISITION A systematic review using Medline and scientific meeting records was carried out in September 2020 according to the Preferred Reporting Items for Systematic Review and Meta-analyses guidelines. Ongoing trials of immunotherapy with standard mCRPC therapeutics were identified via a systematic search on ClinicalTrials.gov. EVIDENCE SYNTHESIS A total of five full-text papers, ten congress abstracts, and 15 trials on ClinicalTrials.gov were identified. Preclinical evidence suggests that combinational approaches might be considered to enhance the efficacy of ICI in PC patients. This led to the design of more than 50 immunotherapy-based clinical trials. The majority of the studies focus on ICI combinations with vaccines, androgen deprivation therapy, chemotherapy, PARP inhibition, radiotherapy, and prostate-specific membrane antigen-guided radioligand therapy. Preliminary analyses reported promising findings for the use of ICI in combination with other anticancer therapies. However, no phase 3 trial has yet reported final results, so no level 1 evidence with long-term outcomes currently supports the combination of ICI with mCRPC therapies. CONCLUSIONS Preclinical and clinical trials have demonstrated that combining immunotherapy with standard mCRPC treatment options has the potential to provide a synergistic effect. Nonetheless, a better understanding of the mechanism and of the optimal treatment approach is still needed. PATIENT SUMMARY We reviewed the literature on immunotherapy in combination with standard treatments for patients with metastatic castration-resistant prostate cancer (mCRPC). Current evidence supports the hypothesis that immunotherapeutic drugs might be effective in mCRPC if combined with other treatment options. However, results of ongoing trials are still awaited before this novel treatment approach can be implemented in the daily practice.
Collapse
Affiliation(s)
- Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria.
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Andreas Pircher
- Department of Internal Medicine V, Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, Mainz University Medicine, Mainz, Germany
| | - Giancarlo Marra
- Department of Urology, San Giovanni Battista Hospital, University of Turin, Turin, Italy
| | - Veeru Kasivisvanathan
- Division of Surgery and Interventional Science, University College London, London, UK
| | | | | | - Francesco Ceci
- Department of Nuclear Medicine, San Giovanni Battista Hospital, Turin, Italy
| | | | - Constance Thibault
- Department of Oncology, Hôpital Européen Georges Pompidou, Paris, France
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christian Surcel
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Giorgio Gandaglia
- Division of Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
33
|
Gao X, Li L, Cai X, Huang Q, Xiao J, Cheng Y. Targeting nanoparticles for diagnosis and therapy of bone tumors: Opportunities and challenges. Biomaterials 2020; 265:120404. [PMID: 32987273 DOI: 10.1016/j.biomaterials.2020.120404] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022]
Abstract
A variety of targeted nanoparticles were developed for the diagnosis and therapy of orthotopic and metastatic bone tumors during the past decade. This critical review will focus on principles and methods in the design of these bone-targeted nanoparticles. Ligands including bisphosphonates, aspartic acid-rich peptides and synthetic polymers were grafted on nanoparticles such as PLGA nanoparticles, liposomes, dendrimers and inorganic nanoparticles for bone targeting. Besides, other ligands such as monoclonal antibodies, peptides and aptamers targeting biomarkers on tumor/bone cells were identified for targeted diagnosis and therapy. Examples of targeted nanoparticles for the early detection of bone metastatic tumors and the ablation of cancer via chemotherapy, photothermal therapy, gene therapy and combination therapy will be intensively reviewed. The development of multifunctional nanoparticles to break down the "vicious" cycle between tumor cell proliferation and bone resorption, and the challenges and perspectives in this area will be discussed.
Collapse
Affiliation(s)
- Xin Gao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Lin Li
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Xiaopan Cai
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Quan Huang
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Jianru Xiao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Yiyun Cheng
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
34
|
Sariisik E, Zistl D, Docheva D, Schilling AF, Benoit M, Sudhop S, Clausen-Schaumann H. Inadequate tissue mineralization promotes cancer cell attachment. PLoS One 2020; 15:e0237116. [PMID: 32857787 PMCID: PMC7454967 DOI: 10.1371/journal.pone.0237116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/20/2020] [Indexed: 01/13/2023] Open
Abstract
Bone metastases are a frequent complication in prostate cancer, and several studies have shown that vitamin D deficiency promotes bone metastases. However, while many studies focus on vitamin D’s role in cell metabolism, the effect of chronically low vitamin D levels on bone tissue, i.e. insufficient mineralization of the tissue, has largely been ignored. To investigate, whether poor tissue mineralization promotes cancer cell attachment, we used a fluorescence based adhesion assay and single cell force spectroscopy to quantify the adhesion of two prostate cancer cell lines to well-mineralized and demineralized dentin, serving as biomimetic bone model system. Adhesion rates of bone metastases-derived PC3 cells increased significantly on demineralized dentin. Additionally, on mineralized dentin, PC3 cells adhered mainly via membrane anchored surface receptors, while on demineralized dentin, they adhered via cytoskeleton-anchored transmembrane receptors, pointing to an interaction via exposed collagen fibrils. The adhesion rate of lymph node derived LNCaP cells on the other hand is significantly lower than that of PC3 and not predominately mediated by cytoskeleton-linked receptors. This indicates that poor tissue mineralization facilitates the adhesion of invasive cancer cells by the exposure of collagen and emphasizes the disease modifying effect of sufficient vitamin D for cancer patients.
Collapse
Affiliation(s)
- Ediz Sariisik
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Chair of Applied Physics, Ludwig-Maximilians-Universität, Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität, Munich, Germany
| | - Domenik Zistl
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
| | - Denitsa Docheva
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Department of Trauma Surgery, Experimental Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Arndt F. Schilling
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Clinic for Trauma Surgery, Orthopaedics, and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Benoit
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Chair of Applied Physics, Ludwig-Maximilians-Universität, Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität, Munich, Germany
| | - Stefanie Sudhop
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität, Munich, Germany
- * E-mail:
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences, Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
35
|
Wu L, Xiang S, Hu X, Mo M, Zhao C, Cai Y, Tong S, Jiang H, Chen L, Wang Z, Xiong W, Ou Z. Prostate-specific antigen modulates the osteogenic differentiation of MSCs via the cadherin 11-Akt axis. Clin Transl Med 2020; 10:363-373. [PMID: 32508049 PMCID: PMC7240859 DOI: 10.1002/ctm2.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A high prevalence of osteoblastic bone metastases is characteristic of prostate cancer. Prostate-specific antigen (PSA) is a serine protease uniquely produced by prostate cancer cells and is an important serological marker for prostate cancer. However, whether PSA modulates the osteogenic process remains largely unknown. In this study, we explored the effect of PSA on modulating the osteoblastic differentiation of mesenchymal stem cells (MSCs). In this study, we used flow cytometry, CCK-8 assay, Alizarin red S (ARS) staining and quantification, alkaline phosphatase (ALP) activity and staining, Western blotting, and quantitative real-time PCR (qRT-PCR) to explore the effect of PSA on osteogenic differentiation of MSCs. RESULTS We first demonstrated that although PSA did not affect the proliferation, morphology, or phenotype of MSCs, it significantly promoted the osteogenic differentiation of MSCs in a concentration-dependent manner. Furthermore, we demonstrated that PSA promoted the osteogenic differentiation of MSCs by elevating the expression of Cadherin 11 in MSCs and, thus, activating the Akt signaling pathway. CONCLUSIONS In conclusion, we demonstrated that PSA could promote the osteogenesis of MSCs through Akt signaling pathway activation by elevating the expression of cadherin-11 in MSCs. These findings imply a possible role of PSA in osteoblastic bone metastases in prostate cancer.
Collapse
Affiliation(s)
- Longxiang Wu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiqi Xiang
- Department of OrthopedicsThe Second Xiangya Hospital of Central South UniversityChangshaP.R. China
| | - Xiheng Hu
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Miao Mo
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Cheng Zhao
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Yi Cai
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Shiyu Tong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Huichuan Jiang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Linxiao Chen
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhi Wang
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Wei Xiong
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| | - Zhenyu Ou
- Department of UrologyXiangya Hospital of Central South UniversityChangshaP.R. China
| |
Collapse
|
36
|
Chen J, Wu Z, Ding W, Xiao C, Zhang Y, Gao S, Gao Y, Cai W. SREBP1 siRNA enhance the docetaxel effect based on a bone-cancer dual-targeting biomimetic nanosystem against bone metastatic castration-resistant prostate cancer. Theranostics 2020; 10:1619-1632. [PMID: 32042326 PMCID: PMC6993241 DOI: 10.7150/thno.40489] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
Until recently, there have been limited options for patients with bone metastatic castration-resistant prostate cancer (BmCRPC) following the failure of or development of resistance to docetaxel (DTX), which is one of the frontline treatments. Sterol regulatory element-binding protein 1 (SREBP1) is reported to regulate abnormal lipid metabolism and to promote the progression and metastasis of prostate cancer (PCa). The siRNA interferes SREBP1 may provide an efficient treatment when combined with DTX. Methods: In this study, lipoic acid (LA) and cross-linked peptide-lipoic acid micelles were cross-linked (LC) for DTX and siSREBP1 delivery (LC/D/siR). Then, cell membrane of PCa cells (Pm) and bone marrow mesenchymal stem cells (Bm) were fused for cloaking LC/D/siR (PB@LC/D/siR). Finally, the synthesized PB@LC/D/siR was evaluated in vitro and in vivo. Results: PB@LC/D/siR is internalized in PCa cells by a mechanism of lysosome escape. Tumor targeting and bone homing studies are evaluated using bone metastatic CRPC (BmCRPC) models, both in vitro and in vivo. Moreover, the enhanced anti-proliferation, anti-migration and anti-invasion capacities of DTX- and siSREBP1- loaded PB@LC (PB@LC/D/siR) were observed in vitro. Furthermore, PB@LC/D/siR was able to suppress the growth of the tumor effectively with deep tumor penetration, high safety and good protection of the bone at the tumor site. Additionally, the mRNA levels and protein levels of SREBP1 and SCD1 were able to be significantly downregulated by PB@LC/D/siR. Conclusion: This study presented a bone-cancer dual-targeting biomimetic nanodelivery system for bone metastatic CRPC.
Collapse
Affiliation(s)
- Jiyuan Chen
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Zhenjie Wu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chengwu Xiao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yu Zhang
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shen Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yuan Gao
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Weimin Cai
- Department of Clinical Pharmacy and Drug Administration, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
37
|
Chowdhury S, Oudard S, Uemura H, Joniau S, Pilon D, Lefebvre P, McQuarrie K, Liu J, Dearden L, Sermon J, Van Sanden S, Diels J, Hadaschik BA. Matching-Adjusted Indirect Comparison of Health-Related Quality of Life and Adverse Events of Apalutamide Versus Enzalutamide in Non-Metastatic Castration-Resistant Prostate Cancer. Adv Ther 2020; 37:512-526. [PMID: 31813087 DOI: 10.1007/s12325-019-01157-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The present study aimed to indirectly compare apalutamide and enzalutamide with respect to tolerability and health-related quality of life (HRQoL) among men with non-metastatic castration-resistant prostate cancer (nmCRPC). METHODS Patient-level data from the SPARTAN study [apalutamide + androgen deprivation therapy (ADT) versus placebo + ADT] and aggregate published data from the PROSPER study (enzalutamide + ADT versus placebo + ADT) were used. Anchored matching-adjusted indirect comparison (MAIC) was conducted by weighting patients' baseline characteristics from SPARTAN to match aggregated baseline characteristics in PROSPER. Odds ratios (ORs) of reported adverse events (AEs) and baseline-to-follow-up least squares mean differences in HRQoL [measured with Functional Assessment of Cancer Therapy-Prostate (FACT-P) score] with 95% credible intervals were re-estimated for SPARTAN arms using weighted population and indirectly compared with those in PROSPER through a Bayesian framework. Events of special interest included fatigue, hot flush, nausea, diarrhea, hypertension, falls, dizziness, decreased appetite, arthralgia, asthenia and headache. In addition, any AEs and serious AEs were explored. RESULTS Of 1207 SPARTAN patients, 1171 were matched to 1401 PROSPER patients. Relative to enzalutamide, apalutamide demonstrated better tolerability as evidenced by the highest probability of reduced occurrence of fatigue [p(OR < 1) = 99.5%], hypertension [p(OR < 1) = 99.2%], decreased appetite [p(OR < 1) = 98.3%], fall [p(OR < 1) = 90.3%], headaches [p(OR < 1) = 86.7%], and nausea [p(OR < 1) = 80.0%]. The probabilities of reduced occurrence of any AEs and SAEs with apalutamide versus enzalutamide were 66.9% and 90.9%, respectively. Relative to enzalutamide, apalutamide treatment was associated with a higher probability of a better HRQoL based on the FACT-P total score [p(diff > 0) = 73.1%]. The probability of a better HRQoL with apalutamide versus enzalutamide was highest for the physical [p(diff > 0) = 97.3%] and functional [p(diff > 0) = 86.7%] wellbeing subscales, and the pain-related subscale [p(diff > 0) = 90.1%]. CONCLUSION Anchored MAIC suggests that treatment of men with nmCRPC with apalutamide is associated with a higher probability of better tolerability due to fewer AEs and better HRQoL than enzalutamide.
Collapse
Affiliation(s)
- Simon Chowdhury
- Department of Medical Oncology, Guy's, King's, and St. Thomas' Hospital, London, UK.
| | - Stéphane Oudard
- European Georges Pompidou Hospital, Paris Descartes University, Paris, France
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | | | | | | | | | - Jinan Liu
- Janssen Research & Development, Horsham, PA, USA
| | | | | | | | | | - Boris A Hadaschik
- University of Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
38
|
Morris MJ, Corey E, Guise TA, Gulley JL, Kevin Kelly W, Quinn DI, Scholz A, Sgouros G. Radium-223 mechanism of action: implications for use in treatment combinations. Nat Rev Urol 2019; 16:745-756. [PMID: 31712765 PMCID: PMC7515774 DOI: 10.1038/s41585-019-0251-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/16/2022]
Abstract
The targeted alpha therapy radium-223 (223Ra) can prolong survival in men with castration-resistant prostate cancer (CRPC) who have symptomatic bone metastases and no known visceral metastases. Preclinical studies demonstrate that 223Ra preferentially incorporates into newly formed bone matrix within osteoblastic metastatic lesions. The emitted high-energy alpha particles induce DNA double-strand breaks that might be irreparable and lead to cell death in nearby exposed tumour cells, osteoblasts and osteoclasts. Consequently, tumour growth and abnormal bone formation are inhibited by these direct effects and by the disruption of positive-feedback loops between tumour cells and the bone microenvironment. 223Ra might also modulate immune responses within the bone. The clinical utility of 223Ra has encouraged the development of other anticancer targeted alpha therapies. A thorough understanding of the mechanism of action could inform the design of new combinatorial treatment strategies that might be more efficacious than monotherapy. On the basis of the current mechanistic knowledge and potential clinical benefits, combination therapies of 223Ra with microtubule-stabilizing cytotoxic drugs and agents targeting the androgen receptor axis, immune checkpoint receptors or DNA damage response proteins are being explored in patients with CRPC and metastatic bone disease.
Collapse
Affiliation(s)
- Michael J Morris
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA.
| | - Eva Corey
- Department of Urology, University of Washington, School of Medicine, Seattle, WA, USA
| | - Theresa A Guise
- Indiana University, School of Medicine, Indianapolis, IN, USA
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - William Kevin Kelly
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - David I Quinn
- Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Arne Scholz
- Bayer AG, Drug Discovery, Pharmaceuticals, Berlin, Germany
| | - George Sgouros
- Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Zhang X. Interactions between cancer cells and bone microenvironment promote bone metastasis in prostate cancer. Cancer Commun (Lond) 2019; 39:76. [PMID: 31753020 PMCID: PMC6873445 DOI: 10.1186/s40880-019-0425-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/13/2019] [Indexed: 12/26/2022] Open
Abstract
Bone metastasis is the leading cause of death in prostate cancer patients, for which there is currently no effective treatment. Since the bone microenvironment plays an important role in this process, attentions have been directed to the interactions between cancer cells and the bone microenvironment, including osteoclasts, osteoblasts, and bone stromal cells. Here, we explained the mechanism of interactions between prostate cancer cells and metastasis-associated cells within the bone microenvironment and further discussed the recent advances in targeted therapy of prostate cancer bone metastasis. This review also summarized the effects of bone microenvironment on prostate cancer metastasis and the related mechanisms, and provides insights for future prostate cancer metastasis studies.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Pathology, Jining First People's Hospital, Jining Medical University, No. 6 Jiankang Road, Jining, 272000, Shandong, P. R. China.
| |
Collapse
|
40
|
Shokoohmand A, Ren J, Baldwin J, Atack A, Shafiee A, Theodoropoulos C, Wille ML, Tran PA, Bray LJ, Smith D, Chetty N, Pollock PM, Hutmacher DW, Clements JA, Williams ED, Bock N. Microenvironment engineering of osteoblastic bone metastases reveals osteomimicry of patient-derived prostate cancer xenografts. Biomaterials 2019; 220:119402. [PMID: 31400612 DOI: 10.1016/j.biomaterials.2019.119402] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 01/01/2023]
Abstract
Representative in vitro models that mimic the native bone tumor microenvironment are warranted to support the development of more successful treatments for bone metastases. Here, we have developed a primary cell 3D model consisting of a human osteoblast-derived tissue-engineered construct (hOTEC) indirectly co-cultured with patient-derived prostate cancer xenografts (PDXs), in order to study molecular interactions in a patient-derived microenvironment context. The engineered biomimetic microenvironment had high mineralization and embedded osteocytes, and supported a high degree of cancer cell osteomimicry at the gene, protein and mineralization levels when co-cultured with prostate cancer PDXs from a lymph node metastasis (LuCaP35) and bone metastasis (BM18) from patients with primary prostate cancer. This fully patient-derived model is a promising tool for the assessment of new molecular mechanisms and as a personalized pre-clinical platform for therapy testing for patients with prostate cancer bone metastases.
Collapse
Affiliation(s)
- Ali Shokoohmand
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Jiongyu Ren
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Jeremy Baldwin
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Anthony Atack
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Abbas Shafiee
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Christina Theodoropoulos
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Marie-Luise Wille
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Phong A Tran
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Laura J Bray
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia
| | - Deborah Smith
- Cancer Pathology Research Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia; Department of Anatomical Pathology, Mater Hospital Brisbane, QLD, Australia
| | - Naven Chetty
- Cancer Pathology Research Group, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia; Department of Anatomical Pathology, Mater Hospital Brisbane, QLD, Australia
| | - Pamela M Pollock
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Judith A Clements
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Elizabeth D Williams
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia
| | - Nathalie Bock
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Australian Prostate Cancer Research Centre, Queensland (APCRC-Q), QUT, Brisbane, QLD, Australia; Translational Research Institute (TRI), QUT, Brisbane, QLD, Australia; Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove, QLD, Australia; School of Biomedical Sciences, Faculty of Health, QUT, Brisbane, QLD, Australia.
| |
Collapse
|
41
|
Synergistic combination treatment to break cross talk between cancer cells and bone cells to inhibit progression of bone metastasis. Biomaterials 2019; 227:119558. [PMID: 31654872 DOI: 10.1016/j.biomaterials.2019.119558] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/05/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Advanced-stage cancers often metastasize to bone, and is the major cause of cancer-related morbidity and mortality. Due to poor biodistribution of intravenously administered anticancer drugs within the bone, chemotherapy is not optimally effective in treating bone metastasis. Additionally, overexpression of receptor activator of nuclear factor κB ligand (RANKL) in the bone microenvironment drives the vicious, destructive cycle of progression of bone metastasis and bone resorption. We hypothesized that the combination treatment - with docetaxel (TXT), an anticancer drug encapsulated in sustained release biodegradable nanoparticles (TXT-NPs) that are designed to localize in bone marrow, and denosumab monoclonal antibody (DNmb), which binds to RANKL - could be more effective than either treatment alone. We tested our hypothesis in intraosseous prostate cancer (PC-3) cell-induced osteolytic mouse model of bone metastasis with treatments given intravenously. The results demonstrated better efficacy with TXT-NPs than with TXT-CrEL or saline control in inhibiting progression of metastasis and improving survival. TXT-NPs showed ~3-fold higher drug levels in metastasized bone tissue at 1 wk post-administration than TXT-CrEL, thus explaining their efficacy. However, the combination treatment (TXT-NPs + DNmb) given simultaneously was significantly more effective in inhibiting metastatic progression; it caused early tumor regression and improved survival, and caused no body weight loss or tumor relapse, even when the treatment was discontinued, whereas TXT-NPs or DNmb alone treatments showed tumor relapse after an initial regression. Micro-CT analysis of the bone from the combination treatment showed no bone loss and normal bone mineral content, bone density, and bone volume fraction, whereas TXT-NPs or DNmb alone treatments showed bone loss. Confirming the above results, histochemical analysis of the bone from the combination treatment demonstrated normal bone morphology, and osteoblast and osteoclast cell activities. In conclusion, TXT-NPs and DNmb in combination, because of their complementary roles in breaking the cross talk between cancer cells and bone cells, was significantly effective in treating bone metastasis.
Collapse
|
42
|
Liolios C, Sachpekidis C, Schäfer M, Kopka K. Bispecific radioligands targeting prostate-specific membrane antigen and gastrin-releasing peptide receptors on the surface of prostate cancer cells. J Labelled Comp Radiopharm 2019; 62:510-522. [DOI: 10.1002/jlcr.3749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/15/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Christos Liolios
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Martin Schäfer
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| | - Klaus Kopka
- Division of Radiopharmaceutical Chemistry; German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
- German Cancer Consortium (DKTK); German Cancer Research Center (DKFZ); Heidelberg ]-->Germany
| |
Collapse
|
43
|
Tassone E, Bradaschia-Correa V, Xiong X, Sastre-Perona A, Josephson AM, Khodadadi-Jamayran A, Melamed J, Bu L, Kahler DJ, Ossowski L, Leucht P, Schober M, Wilson EL. KLF4 as a rheostat of osteolysis and osteogenesis in prostate tumors in the bone. Oncogene 2019; 38:5766-5777. [PMID: 31239516 PMCID: PMC6639130 DOI: 10.1038/s41388-019-0841-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
We previously showed that KLF4, a gene highly expressed in murine prostate stem cells, blocks the progression of indolent intraepithelial prostatic lesions into aggressive and rapidly growing tumors. Here, we show that the anti-tumorigenic effect of KLF4 extends to PC3 human prostate cancer cells growing in the bone. We compared KLF4 null cells with cells transduced with a DOX-inducible KLF4 expression system, and find KLF4 function inhibits PC3 growth in monolayer and soft agar cultures. Furthermore, KLF4 null cells proliferate rapidly, forming large, invasive, and osteolytic tumors when injected into mouse femurs, whereas KLF4 re-expression immediately after their intra-femoral inoculation blocks tumor development and preserves a normal bone architecture. KLF4 re-expression in established KLF4 null bone tumors inhibits their osteolytic effects, preventing bone fractures and inducing an osteogenic response with new bone formation. In addition to these profound biological changes, KLF4 also induces a transcriptional shift from an osteolytic program in KLF4 null cells to an osteogenic program. Importantly, bioinformatic analysis shows that genes regulated by KLF4 overlap significantly with those expressed in metastatic prostate cancer patients and in three individual cohorts with bone metastases, strengthening the clinical relevance of the findings in our xenograft model.
Collapse
Affiliation(s)
- Evelyne Tassone
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
| | - Vivian Bradaschia-Correa
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Xiaozhong Xiong
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
| | - Ana Sastre-Perona
- The Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, 10016, USA
| | - Anne Marie Josephson
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, NYU School of Medicine, New York, NY, 10016, USA
- Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY, 10016, USA
| | - Jonathan Melamed
- Department of Pathology, NYU School of Medicine, New York, NY, 10016, USA
| | - Lei Bu
- Department of Medicine, NYU School of Medicine, New York, NY, 10016, USA
| | - David J Kahler
- High Throughput Biology Laboratory, NYU School of Medicine, New York, NY, 10016, USA
| | - Liliana Ossowski
- Department of Medicine, Mt Sinai School of Medicine, New York, NY, 10029, USA
| | - Philipp Leucht
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA
- Department of Orthopedic Surgery, NYU School of Medicine, New York, NY, 10016, USA
| | - Markus Schober
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.
- The Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, 10016, USA.
| | - Elaine L Wilson
- Department of Cell Biology, NYU School of Medicine, New York, NY, 10016, USA.
- Department of Urology, NYU School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
44
|
Soldatos TG, Iakovou I, Sachpekidis C. Retrospective Toxicological Profiling of Radium-223 Dichloride for the Treatment of Bone Metastases in Prostate Cancer Using Adverse Event Data. ACTA ACUST UNITED AC 2019; 55:medicina55050149. [PMID: 31100964 PMCID: PMC6572036 DOI: 10.3390/medicina55050149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
Abstract
Background and Objective: Radium-223 dichloride (Xofigo®) is a calcium mimetic agent approved for the treatment of castration-resistant prostate cancer patients with symptomatic bone metastases and no known visceral metastatic disease. This targeted, α-particle-emitting therapy has demonstrated significant survival benefit accompanied by a favorable safety profile. Nevertheless, recent evidence suggests that its combined use with abiraterone and prednisone/prednisolone may be associated with increased risk of death and fractures. While the precise pathophysiologic mechanisms of these events are not yet clear, collecting evidence from more clinical trials and translational studies is necessary. The aim of our present study is to assess whether accessible sources of patient outcome data can help gain additional clinical insights to radium-223 dichloride’s safety profile. Materials and Methods: We performed a retrospective analysis of cases extracted from the FDA Adverse Event Reporting System and characterized side effect occurrence by using reporting ratios. Results: A total of ~1500 prostate cancer patients treated with radium-223 dichloride was identified, and side effects reported with the use of radium-223 dichloride alone or in combination with other therapeutic agents were extracted. Our analysis demonstrates that radium-223 dichloride may often come with hematological-related reactions, and that, when administered together with other drugs, its safety profile may differ. Conclusions: While more prospective studies are needed to fully characterize the toxicological profile of radium-223 dichloride, the present work constitutes perhaps the first effort to examine its safety when administered alone and in combination with other agents based on computational evidence from public real-world post marketing data.
Collapse
Affiliation(s)
| | - Ioannis Iakovou
- Department of Nuclear Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece.
| | - Christos Sachpekidis
- Department of Nuclear Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece.
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
45
|
Gallicchio R, Mastrangelo PA, Nardelli A, Mainenti PP, Colasurdo AP, Landriscina M, Guglielmi G, Storto G. Radium-223 for the treatment of bone metastases in castration-resistant prostate cancer: when and why. TUMORI JOURNAL 2019; 105:367-377. [PMID: 31096849 DOI: 10.1177/0300891619851376] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Radium-223 dichloride (223Ra) is the first, recently approved, α-particle-emitting radiopharmaceutical for the treatment of patients with bone metastases in castration-resistant prostate cancer (CRPC) and no evidence of visceral metastases. We explored MEDLINE, relevant congresses, and websites for data on 223Ra and prostate cancer therapies, focusing on therapeutic strategies and timing, bone metastases, and diagnostic assessment. 223Ra represents the only bone-targeting agent that has significantly extended patients' overall survival while reducing pain and symptomatic skeletal events. Unlike other radiopharmaceuticals, such as strontium-89 and samarium-153 EDTMP, 223Ra (11.4-days half-life) has shown a high biological efficiency mainly due to its short penetration range. These features potentially allow reduced bone marrow toxicity and limit undue exposure. 223Ra has been validated under the product name Xofigo® by the US Food and Drug Administration and the European Medicines Agency. Patient selection, management, and treatment sequencing is recommended to be discussed in the context of a multidisciplinary environment, including oncology, urology, nuclear medicine, and radiation therapy physicians. No consensus has been achieved regarding the optimal timing and its administration as single agent or in combination with zoledronic acid or chemotherapy, so far. This review aims to provide a rationale for the use of 223Ra in treating metastases from CRPC, highlighting the crucial role of a multidisciplinary approach, the disputed inclusion and exclusion criteria on the basis of agencies regulations, and the value of diagnostics for therapy assessment.
Collapse
Affiliation(s)
- Rosj Gallicchio
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, Italy
| | - Pietro A Mastrangelo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, Italy
| | - Anna Nardelli
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Pier Paolo Mainenti
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Antonio P Colasurdo
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, Italy
| | - Matteo Landriscina
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, Italy.,Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi di Foggia, Italy
| | - Giuseppe Guglielmi
- Dipartimento di Medicina Clinica e Sperimentale, Università degli Studi di Foggia, Italy.,IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| | - Giovanni Storto
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Centro di Riferimento Oncologico della Basilicata (CROB), Rionero in Vulture, Italy
| |
Collapse
|
46
|
Liu J, Zhao C, Liu B, Liu H, Wang L. Analgesia and curative effect of pamidronate disodium combined with chemotherapy on elderly patients with advanced metastatic bone cancer. Oncol Lett 2019; 18:771-775. [PMID: 31289553 PMCID: PMC6540329 DOI: 10.3892/ol.2019.10340] [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: 01/24/2019] [Accepted: 05/06/2019] [Indexed: 11/06/2022] Open
Abstract
The curative effect and adverse reactions of pamidronate disodium in elderly patients with advanced metastatic bone cancer were evaluated. A total of 160 elderly patients with advanced metastatic bone cancer admitted to Affiliated Hospital of Nantong University from February 2012 to January 2015, were divided into the chemotherapy group (n=60) that received routine therapy and the pamidronate disodium group (n=100) that received pamidronate disodium therapy based on the chemotherapy. Pain relief, analgesic time, analgesic duration and side effects were compared between the two groups after treatment. The effect of pain relief in the pamidronate disodium group was significantly higher than that in the chemotherapy group (P<0.001). The total effective rate of the pamidronate disodium group was significantly higher than that of the chemotherapy group (P<0.001). The analgesic onset time in the pamidronate disodium group was earlier than in the chemotherapy group (P<0.001). The analgesic duration in the pamidronate disodium group was longer than that in the chemotherapy group (P<0.001). The incidence of adverse reactions and complications after treatment in the pamidronate disodium group was significantly less than that in the chemotherapy group (P<0.001). The results indicated that pamidronate disodium is effective in the treatment of elderly patients with advanced metastatic bone cancer and patients are less prone to adverse reactions, complications and pain, which is worthy of clinical application.
Collapse
Affiliation(s)
- Jiayong Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Bone and Soft Tissue Tumor, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Chaoqian Zhao
- Department of Oncology, Zhuzhou Central Hospital, Zhuzhou, Hunan 412007, P.R. China
| | - Bin Liu
- Department of Oncology, The Second Hospital of Xiangya, Central South University, Changsha, Hunan 410011, P.R. China
| | - Hua Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lixin Wang
- Department of Integrated TCM and Western Medicine, Shanghai Pulmonary Hospital Affiliated to Tongji University, Shanghai 200043, P.R. China
| |
Collapse
|
47
|
Bock N, Shokoohmand A, Kryza T, Röhl J, Meijer J, Tran PA, Nelson CC, Clements JA, Hutmacher DW. Engineering osteoblastic metastases to delineate the adaptive response of androgen-deprived prostate cancer in the bone metastatic microenvironment. Bone Res 2019; 7:13. [PMID: 31044095 PMCID: PMC6486620 DOI: 10.1038/s41413-019-0049-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/13/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
While stromal interactions are essential in cancer adaptation to hormonal therapies, the effects of bone stroma and androgen deprivation on cancer progression in bone are poorly understood. Here, we tissue-engineered and validated an in vitro microtissue model of osteoblastic bone metastases, and used it to study the effects of androgen deprivation in this microenvironment. The model was established by culturing primary human osteoprogenitor cells on melt electrowritten polymer scaffolds, leading to a mineralized osteoblast-derived microtissue containing, in a 3D setting, viable osteoblastic cells, osteocytic cells, and appropriate expression of osteoblast/osteocyte-derived mRNA and proteins, and mineral content. Direct co-culture of androgen receptor-dependent/independent cell lines (LNCaP, C4-2B, and PC3) led cancer cells to display functional and molecular features as observed in vivo. Co-cultured cancer cells showed increased affinity to the microtissues, as a function of their bone metastatic potential. Co-cultures led to alkaline phosphatase and collagen-I upregulation and sclerostin downregulation, consistent with the clinical marker profile of osteoblastic bone metastases. LNCaP showed a significant adaptive response under androgen deprivation in the microtissues, with the notable appearance of neuroendocrine transdifferentiation features and increased expression of related markers (dopa decarboxylase, enolase 2). Androgen deprivation affected the biology of the metastatic microenvironment with stronger upregulation of androgen receptor, alkaline phosphatase, and dopa decarboxylase, as seen in the transition towards resistance. The unique microtissues engineered here represent a substantial asset to determine the involvement of the human bone microenvironment in prostate cancer progression and response to a therapeutic context in this microenvironment.
Collapse
Affiliation(s)
- Nathalie Bock
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
- Centre in Regenerative Medicine, QUT, Kelvin Grove, QLD 4059 Australia
| | - Ali Shokoohmand
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
- Centre in Regenerative Medicine, QUT, Kelvin Grove, QLD 4059 Australia
| | - Thomas Kryza
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
| | - Joan Röhl
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
| | - Jonelle Meijer
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
- Centre in Regenerative Medicine, QUT, Kelvin Grove, QLD 4059 Australia
| | - Phong A. Tran
- Centre in Regenerative Medicine, QUT, Kelvin Grove, QLD 4059 Australia
- Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD 4000 Australia
| | - Colleen C. Nelson
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
| | - Judith A. Clements
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
| | - Dietmar W. Hutmacher
- School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD 4000 Australia
- Translational Research Institute (TRI), Woolloongabba, QLD 4102 Australia
- Centre in Regenerative Medicine, QUT, Kelvin Grove, QLD 4059 Australia
- Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane, QLD 4000 Australia
- Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove, QLD 4059 Australia
| |
Collapse
|
48
|
Adjei IM, Temples MN, Brown SB, Sharma B. Targeted Nanomedicine to Treat Bone Metastasis. Pharmaceutics 2018; 10:E205. [PMID: 30366428 PMCID: PMC6320768 DOI: 10.3390/pharmaceutics10040205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Bone metastases are common complications of solid tumors, particularly those of the prostate, breast, and lungs. Bone metastases can lead to painful and devastating skeletal-related events (SREs), such as pathological fractures and nerve compressions. Despite advances in treatment for cancers in general, options for bone metastases remain inadequate and generally palliative. Anticancer drugs (chemotherapy and radiopharmaceuticals) do not achieve therapeutic concentrations in the bone and are associated with dose-limiting side effects to healthy tissues. Nanomedicines, with their tunable characteristics, have the potential to improve drug targeting to bone metastases while decreasing side effects for their effective treatment. In this review, we present the current state of the art for nanomedicines to treat bone metastases. We also discuss new treatment modalities enhanced by nanomedicine and their effects on SREs and disease progression.
Collapse
Affiliation(s)
- Isaac M Adjei
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Madison N Temples
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Shannon B Brown
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
49
|
Yamashita K, Mizugishi K, Takaori-Kondo A. Familial Mediterranean Fever Mutations in a Patient with Periodic Episodes of Systemic Pain Deriving from Cancer Bone Metastases. Intern Med 2018; 57:2901-2904. [PMID: 29780113 PMCID: PMC6207823 DOI: 10.2169/internalmedicine.0431-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Familial Mediterranean fever (FMF), the most common autoinflammatory disorder, is characterized by recurrent febrile attacks and polyserositis. FMF is caused by mutations in MEFV, which encodes pyrin. In this report, we present an atypical FMF case with E148Q/L110P mutations in MEFV. The patient experienced periodic episodes of systemic pain originating from prostate cancer bone metastases. The pain attacks were prevented by continuous prophylactic therapy with colchicine. In this case, the presence of atypical FMF may have modulated the clinical manifestations of cancer bone metastases. To our knowledge, this is the first report to demonstrate the potential modulatory effect of MEFV mutations on cancer manifestations.
Collapse
Affiliation(s)
- Kouhei Yamashita
- Department of Hematology and Oncology, Kyoto University Hospital, Japan
| | - Kiyomi Mizugishi
- Department of Hematology and Oncology, Kyoto University Hospital, Japan
| | | |
Collapse
|
50
|
Tsaur I, Heidegger I, Kretschmer A, Borgmann H, Mirvald C, Gandaglia G, Briganti A, van den Bergh R, Tilki D, Ost P, Ploussard G, Surcel C. Combining anticancer drugs with osteoprotective agents in prostate cancer-A contemporary update. Urol Oncol 2018; 36:488-497. [PMID: 30268712 DOI: 10.1016/j.urolonc.2018.08.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 08/20/2018] [Accepted: 08/25/2018] [Indexed: 11/29/2022]
Abstract
Recently, a plethora of life-prolonging cytotoxic, next-generation hormonal, immunotherapeutical as well as radionuclide therapies has emerged as a standard care for metastasized castration-resistant prostate cancer. Being strikingly effective in cancer control, these novel therapies might in fact exert a beneficial impact on skeletal events. Therefore, combining anticancer drugs with osteoprotective agents might lead to additional clinical advantage but must be weighed against simultaneously exposing patients to serious toxicities. In addition, further survival prolongation by changing treatment paradigm in both metastasized hormone-sensitive and nonmetastatic castration-resistant disease might potentially increase the risk for bone density reduction complications due to a growing efficacy of androgen ablation leading to prolonged exposure. To address both possible indications of combined treatment and to draw practical conclusions, we performed a comprehensive review of the currently available evidence.
Collapse
Affiliation(s)
- Igor Tsaur
- Department of Urology and Pediatric Urology, Mainz University Medicine, Mainz, Germany.
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Hendrik Borgmann
- Department of Urology and Pediatric Urology, Mainz University Medicine, Mainz, Germany
| | - Cristian Mirvald
- Department of Urology, Fundeni Clinical Institute, University of Medicine and Pharmacy, Carol Davila Bucharest, Bucharest, Romania
| | - Giorgio Gandaglia
- Department of Urology, Urological Research Institute, Vita-Salute University and San Raffaele Hospital, Milan, Italy
| | - Alberto Briganti
- Department of Urology, Urological Research Institute, Vita-Salute University and San Raffaele Hospital, Milan, Italy
| | | | - Derya Tilki
- Department of Urology, Martini Klinik, Hamburg, Germany
| | - Piet Ost
- Department of radiation oncology and experimental cancer research, Ghent University Hospital, Ghent, Belgium
| | - Guillaume Ploussard
- Department of Urology, Saint Jean Languedoc Hospital, Toulouse, France and Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Cristian Surcel
- Department of Urology, Fundeni Clinical Institute, University of Medicine and Pharmacy, Carol Davila Bucharest, Bucharest, Romania
| | | |
Collapse
|