1
|
Akkus E, Arslan Ç, Ürün Y. Advancements in platinum chemotherapy for metastatic castration-resistant prostate cancer: Insights and perspectives. Cancer Treat Rev 2024; 130:102818. [PMID: 39178612 DOI: 10.1016/j.ctrv.2024.102818] [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: 03/03/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
Despite improvements in survival, metastatic castration-resistant prostate cancer (mCRPC) remains a significant clinical challenge. While taxanes, new hormonal agents, radiopharmaceuticals, and PARP inhibitors offer valuable treatment options, this review explores the potential of platinum chemotherapies (carboplatin, cisplatin, and oxaliplatin) as alternative choices. Existing research demonstrates promising preliminary results for platinum-based therapies in mCRPC showing PSA response rates (7.7-95 %) and improved overall survival (8-26.6 months). However, chemotherapy-related cytopenias are a frequent side effect. Further research is underway to evaluate the efficacy of platinum regimens against specific mCRPC histopathological variants, particularly aggressive subtypes where the carboplatin and cabazitaxel combination is already recommended. The unique DNA-targeting action of platinum therapy holds promise for patients with deficient DNA repair (dDDR), especially those with BRCA mutations. This potential is supported by both preclinical and ongoing clinical research. Given the limited success of immunotherapy in mCRPC, researchers are exploring the potential for platinum therapies to enhance its efficacy. Additionally, trials are investigating the synergy of combining platinum therapy with both immunotherapy and PARP inhibitors. Further exploration into the effectiveness of platinum therapies in specific mCRPC subpopulations, particularly those with dDDR, is crucial for optimizing their future use. In conclusion, this review highlights the promising potential of platinum-based chemotherapy as a valuable treatment option for mCRPC. While current evidence is encouraging, ongoing research is essential to further optimize its efficacy, identify optimal combinations with other therapies, and better understand its impact on specific mCRPC subpopulations.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University, Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University, Cancer Research Institute, Ankara, Türkiye
| | - Çağatay Arslan
- İzmir University of Economics, Medicalpoint Hospital, Department of Medical Oncology, İzmir, Türkiye
| | - Yüksel Ürün
- Ankara University, Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University, Cancer Research Institute, Ankara, Türkiye.
| |
Collapse
|
2
|
Baude J, Niogret J, Jacob P, Bardet F, Desmoulins I, Zanetta S, Kaderbhai C, Galland L, Mayeur D, Delattre B, Cormier L, Ladoire S. Carboplatin and Etoposide for the Treatment of Metastatic Prostate Cancer with or without Neuroendocrine Features: A French Single-Center Experience. Cancers (Basel) 2024; 16:280. [PMID: 38254771 PMCID: PMC10813788 DOI: 10.3390/cancers16020280] [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: 11/15/2023] [Revised: 12/17/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Chemotherapy using carboplatin and etoposide (CE) is frequently pragmatically proposed to treat metastatic prostate cancer (mPC), both primary small-cell neuroendocrine (PSC-NE) carcinoma and adenocarcinoma with or without neuroendocrine (NE) marker elevation. However, the real benefit of CE is poorly reported in the recent therapeutic context. METHODS We retrospectively analyzed the efficacy and tolerance of CE chemotherapy in these three different groups of mPC patients. Efficacy endpoints included radiological response, progression-free survival (PFS), and overall survival (OS), as well as PSA response and PFS2/PFS1 ratio in patients with adenocarcinoma. RESULTS Sixty-nine patients were included in this single-center study (N = 18 with PSC-NE carcinoma and 51 with adenocarcinoma with (N = 18) or without (N = 33) NE marker elevation). Patients with adenocarcinoma were highly pretreated with next-generation hormonal agents (NHAs) and taxanes. In patients with adenocarcinoma, a PSA response ≥50% was observed in six patients (15.8%), four of whom had NE marker elevation. The radiological response was higher in PSC-NE and tended to be higher in adenocarcinoma when NE marker elevation was present. Comparing patients with adenocarcinoma with vs. without NE marker elevation, the median PFS was 3.7 and 2.1 months and the median OS was 7.7 and 4.7 months, respectively. Overall, 62.3% of patients experienced grade 3-4 adverse events (mainly hematological), and three treatment-related deaths were recorded. CONCLUSION Reports of the clinical results of CE suggest that we should not mix PSC-NE and castration-resistant adenocarcinoma of the prostate. In patients with heavily pretreated adenocarcinoma, the benefit/risk ratio of CE chemotherapy seems unfavorable due to poor response and high toxicity.
Collapse
Affiliation(s)
- Jérémy Baude
- Department of Radiation Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France;
| | - Julie Niogret
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Pierre Jacob
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Florian Bardet
- Department of Urology, University Hospital François Mitterrand, 21000 Dijon, France; (F.B.)
| | - Isabelle Desmoulins
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Courèche Kaderbhai
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Loïck Galland
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Didier Mayeur
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
| | - Benjamin Delattre
- Department of Urology, University Hospital François Mitterrand, 21000 Dijon, France; (F.B.)
| | - Luc Cormier
- Department of Urology, University Hospital François Mitterrand, 21000 Dijon, France; (F.B.)
- UFR des Sciences de Santé, Université of Bourgogne Franche-Comté, 21000 Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
- UFR des Sciences de Santé, Université of Bourgogne Franche-Comté, 21000 Dijon, France
- Platform of Transfer in Biological Oncology, Georges-François Leclerc Cancer Center, 21000 Dijon, France
- INSERM U1231 «Lipid, Nutrition, Cancer», 21000 Dijon, France
| |
Collapse
|
3
|
Feuerecker B, Kratochwil C, Ahmadzadehfar H, Morgenstern A, Eiber M, Herrmann K, Pomykala KL. Clinical Translation of Targeted α-Therapy: An Evolution or a Revolution? J Nucl Med 2023; 64:685-692. [PMID: 37055224 DOI: 10.2967/jnumed.122.265353] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/10/2023] [Indexed: 04/15/2023] Open
Abstract
The field of radioligand therapy has advanced greatly in recent years, driven largely by β-emitting therapies targeting somatostatin receptor-expressing tumors and the prostate-specific membrane antigen. Now, more clinical trials are under way to evaluate α-emitting targeted therapies as potential next-generation theranostics with even higher efficacy due to their high linear energy and short range in human tissues. In this review, we summarize the important studies ranging from the first Food and Drug Administration-approved α-therapy, 223Ra-dichloride, for treatment of bone metastases in castration-resistant prostate cancer, including concepts in clinical translation such as targeted α-peptide receptor radiotherapy and 225Ac-PSMA-617 for treatment of prostate cancer, innovative therapeutic models evaluating new targets, and combination therapies. Targeted α-therapy is one of the most promising fields in novel targeted cancer therapy, with several early- and late-stage clinical trials for neuroendocrine tumors and metastatic prostate cancer already in progress, along with significant interest and investment in additional early-phase studies. Together, these studies will help us understand the short- and long-term toxicity of targeted α-therapy and potentially identify suitable therapeutic combination partners.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Nuclear Medicine, Technische Universität München, München, Germany
- Department of Radiology, Technische Universität München, München, Germany
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Radiology, University Hospital, LMU München, München, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Hojjat Ahmadzadehfar
- Department of Nuclear Medicine, Klinikum Westfalen-Knappschaftskrankenhaus, Dortmund, Germany
| | | | - Matthias Eiber
- Department of Nuclear Medicine, Technische Universität München, München, Germany
| | - Ken Herrmann
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; and
| | - Kelsey L Pomykala
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
4
|
Chong ZX, Ho WY, Yeap SK. Delineating the tumour-regulatory roles of EYA4. Biochem Pharmacol 2023; 210:115466. [PMID: 36849065 DOI: 10.1016/j.bcp.2023.115466] [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: 12/29/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
Eyes absent homolog 4 (EYA4) is a protein that regulates many vital cellular processes and organogenesis pathways. It possesses phosphatase, hydrolase, and transcriptional activation functions. Mutations in the Eya4 gene can cause sensorineural hearing loss and heart disease. In most non-nervous system cancers such as those of the gastrointestinal tract (GIT), hematological and respiratory systems, EYA4 acts as a putative tumor suppressor. However, in nervous system tumors such as glioma, astrocytoma, and malignant peripheral nerve sheath tumor (MPNST), it plays a putative tumor-promoting role. EYA4 interacts with various signaling proteins of the PI3K/AKT, JNK/cJUN, Wnt/GSK-3β, and cell cycle pathways to exert its tumor-promoting or tumor-suppressing effect. The tissue expression level and methylation profiles of Eya4 can help predict the prognosis and anti-cancer treatment response among cancer patients. Targeting and altering Eya4 expression and activity could be a potential therapeutic strategy to suppress carcinogenesis. In conclusion, EYA4 may have both putative tumor-promoting and tumor-suppressing roles in different human cancers and has the potential to serve as a prognostic biomarker and therapeutic agent in various cancer types.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500 Semenyih, Selangor, Malaysia.
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900 Sepang, Selangor, Malaysia.
| |
Collapse
|
5
|
Liu F, Zhang H, Sun Z, Meng X, Ma Z, Wang Z. Effects of etoposide combined with cisplatin on prognosis of patients with castration-resistant prostate cancer who failed castration treatment. Am J Transl Res 2022; 14:1705-1713. [PMID: 35422924 PMCID: PMC8991174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To determine the influences of etoposide combined with cisplatin on prognosis of patients with castration-resistant prostate cancer (CRPC) who failed castration treatment. METHODS A total of 100 patients with metastatic CRPC who failed castration treatment in our hospital from January 2015 to January 2017 were retrospectively analyzed. The patients were divided into a control group (n=59) treated with docetaxel combined with prednisone and an experimental group (n=41) treated with etoposide combined with cisplatin (EP). The change in prostate-specific antigen (PSA) level was adopted as the evaluation criterion for efficacy, by which the total clinical effective rate of patients was calculated. The neurologic rating scale (NRS) was adopted to evaluate the pain of patients, and the incidence of adverse reactions was compared between the two groups. Cox regression was carried out to analyze independent prognostic factors impacting 3-year survival. RESULTS The experimental group showed a significantly better clinical improvement than the control group (P<0.05). According to further analysis, the experimental group had a significantly higher clinical efficacy rate than the control group (P<0.05). Life quality scores of the experimental group were higher than those of the control group (all P<0.05). The two groups were not greatly different in bone pain, or incidence of adverse reactions (both P>0.05). The median survival time of the control group was 15.9 months, while that of the experimental group was 18 months, and the control group experienced a greatly shorter median survival time than the experimental group (P=0.040). According to Cox regression analysis, Gleason score, clinical stage, and metastasis were independent factors impacting the patients' 3-year prognosis (all P<0.05). CONCLUSION EP regimen can strongly improve the 3-year survival rate of patients, without increasing adverse reactions.
Collapse
Affiliation(s)
- Feng Liu
- Department of Nephrology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| | - Huaiwei Zhang
- Department of Urology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| | - Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| | - Xiangdi Meng
- Department of Urology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| | - Zhaosen Ma
- Department of Urology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| | - Zhixin Wang
- Department of Urology, China-Japan Union Hospital of Jilin University126 Xiantai Street, Changchun 130031, Jilin, China
| |
Collapse
|
6
|
Chao OS, Goodman OB. DNA-PKc inhibition overcomes taxane resistance by promoting taxane-induced DNA damage in prostate cancer cells. Prostate 2021; 81:1032-1048. [PMID: 34297853 DOI: 10.1002/pros.24200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/15/2021] [Accepted: 07/09/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Overcoming taxane resistance remains a major clinical challenge in metastatic castrate-resistant prostate cancer (mCRPC). Loss of DNA repair proteins is associated with resistance to anti-microtubule agents. We propose that alterations in DNA damage response (DDR) pathway contribute to taxane resistance, and identification of these alterations may provide a potential therapeutic target to resensitize docetaxel-refractory mCRPC to taxane-based therapy. METHODS Alterations in DDR gene expression in our prostate cancer cell line model of docetaxel-resistance (DU145-DxR) derived from DU-145 cells were determined by DDR pathway-specific polymerase chain reaction array and immunoblotting. The PRKDC gene encoding DNA-PKc (DNA-dependent protein kinase catalytic unit), was noted to be overexpressed and evaluated for its role in docetaxel resistance. Cell viability and clonogenic survival of docetaxel-treated DU145-DxR cells were assessed after pharmacologic inhibition of DNA-PKc with three different inhibitors-NU7441, LTURM34, and M3814. Response to second-line cytotoxic agents, cabazitaxel and etoposide upon DNA-PKc inhibition was also tested. The impact of DNA-PKc upregulation on DNA damage repair was evaluated by comet assay and analysis of double-strand breaks marker, γH2AX and Rad51. Lastly, DNA-PKc inhibitor's effect on MDR1 activity was assessed by rhodamine 123 efflux assay. RESULTS DDR pathway-specific gene profiling revealed significant upregulation of PRKDC and CDK7, and downregulation of MSH3 in DU145-DxR cells. Compared to parental DU145, DU145-DxR cells sustained significantly less DNA damage when exposed to etoposide and docetaxel. Pharmacologic inhibition of DNA-PKc, a component of NHEJ repair machinery, with all three inhibitors, significantly resensitized DU145-DxR cells to docetaxel. Furthermore, DNA-PKc inhibition also resensitized DU145-DxR to cabazitaxel and etoposide, which demonstrated cross-resistance. Inhibition of DNA-PKc led to increased DNA damage in etoposide- and docetaxel-treated DU145-DxR cells. Finally, DNA-PKc inhibition did not affect MDR1 activity, indicating that DNA-PKc inhibitors resensitized taxane-resistant cells via an MDR1-independent mechanism. CONCLUSION This study supports a role of DDR genes, particularly, DNA-PKc in promoting resistance to taxanes in mCRPC. Targeting prostatic DNA-PKc may provide a novel strategy to restore taxane sensitivity in taxane-refractory mCRPC.
Collapse
Affiliation(s)
- Olivia S Chao
- College of Medicine, Roseman University of Health Sciences, Las Vegas, Nevada, USA
| | - Oscar B Goodman
- College of Medicine, Roseman University of Health Sciences, Las Vegas, Nevada, USA
- Comprehensive Cancer Centers of Nevada, Las Vegas, Nevada, USA
| |
Collapse
|
7
|
Hongo H, Kosaka T, Nakatsuka S, Oya M. A long-term survivor of metastatic neuroendocrine prostate cancer treated with multimodal therapy: genetic consideration from next-generation sequencing. Int Cancer Conf J 2021; 10:174-180. [PMID: 34221827 DOI: 10.1007/s13691-021-00482-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/31/2021] [Indexed: 11/30/2022] Open
Abstract
It is still unclear whether cell-free DNA (cfDNA) can replace solid-tissue biopsy. A 59-year-old man developed castration-resistant prostate cancer with a liver metastasis. We performed a liver biopsy and collected a cfDNA sample. Although he underwent radiofrequency ablation, tumors recurred and he was transferred to another hospital. We performed next-generation sequencing using DNA from the biopsy tissue and cfDNA. BRCA2 p.T3033fs and AURKA F31I were detected in both the biopsy tissue and the cfDNA. cfDNA may be useful as a liquid biopsy for monitoring the gene profile of aggressive prostate cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13691-021-00482-2.
Collapse
Affiliation(s)
- Hiroshi Hongo
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Seishi Nakatsuka
- Department of Diagnostic Radiology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| |
Collapse
|
8
|
Activity and Adverse Events of Actinium-225-PSMA-617 in Advanced Metastatic Castration-resistant Prostate Cancer After Failure of Lutetium-177-PSMA. Eur Urol 2020; 79:343-350. [PMID: 33293081 DOI: 10.1016/j.eururo.2020.11.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Beta-emitting Lu-177-labeled prostate-specific membrane antigen (PSMA) radioligand therapy (RLT) is a new option for metastatic castration-resistant prostate cancer (mCRPC), but its antitumor effect can decrease over time. OBJECTIVE To report the safety and activity of alpha-emitting Ac-225-PSMA-617 RLT in mCRPC that has progressed after Lu-177-PSMA. DESIGN, SETTING, AND PARTICIPANTS Twenty-six patients were treated under a compassionate use protocol. The eligibility criteria included previous treatment with abiraterone or enzalutamide, previous taxane-based chemotherapy, progression after Lu-177-PSMA, and positive PSMA-ligand uptake. The median number of previous mCRPC regimens was 6. Ac-225-PSMA-617 was given every 8 wk until progression/intolerable side effects. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Prostate-specific antigen (PSA) decline, PSA progression-free survival (PSA-PFS), clinical progression-free survival (cPFS), overall survival (OS), and toxicity were measured. RESULTS AND LIMITATIONS Sixty-one cycles of Ac-225-PSMA-617 (median number of cycles 2; median activity 9 MBq) were administered. A PSA decline of ≥50% was achieved in 17/26 patients. The median PSA-PFS, cPFS, and OS periods were 3.5 (95% confidence interval [CI] 1.8-11.2), 4.1 (95% CI 3-14.8), and 7.7 (95% CI 4.5-12.1) mo, respectively. Liver metastases were associated with shorter PSA-PFS (median 1.9 vs 4.0 mo; p = 0.02), cPFS (median 1.8 vs 5.2 mo; p = 0.001), and OS (median 4.3 vs 10.4 mo; p = 0.01). Hematological grade 3/4 toxicities were anemia (35%), leucopenia (27%), and thrombocytopenia (19%). All patients experienced grade 1/2 xerostomia. Two and six patients stopped due to hematological toxicity and xerostomia, respectively. A limitation is the retrospective design. CONCLUSIONS Ac-225-PSMA-617 showed measurable antitumor effect after Lu-177-PSMA failure in late-stage mCRPC. Grade 3/4 hematological side effects were observed in up to one-third of patients, and xerostomia led to treatment halt in a relevant number of patients. PATIENT SUMMARY Ac-225-labeled prostate-specific membrane antigen (PSMA)-617 therapy showed substantial antitumor effect in late metastatic castration-resistant prostate cancer after Lu-177-PSMA failure. However, dry mouth is a common side effect that caused about a quarter of patients to stop therapy.
Collapse
|
9
|
Erkisa M, Ari F, Ulku I, Khodadust R, Yar Y, Yagci Acar H, Ulukaya E. Etoposide Loaded SPION-PNIPAM Nanoparticles Improve the in vitro Therapeutic Outcome on Metastatic Prostate Cancer Cells via Enhanced Apoptosis. Chem Biodivers 2020; 17:e2000607. [PMID: 32918383 DOI: 10.1002/cbdv.202000607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/11/2020] [Indexed: 01/02/2023]
Abstract
Prostate cancer is among the leading causes of death worldwide because its metastatic form is a deadly disease. Therefore, the development of new chemotherapeutics is of immense importance. Nanoparticle technology seems to provide diverse options in this regard. Therefore, poly(N-isopropylacrylamide) (PNIPAM) coated superparamagnetic iron oxide nanoparticles (SPION) loaded with Etoposide were prepared in small sizes (57 nm) and with 3.5 % drug content to improve the efficiency of Etoposide in prostate cancer therapy. Sustained release of the drug was achieved, which found to be sensitive to low pH and high temperature. The anti-growth activity of SPION-PNIPAM-Etoposide formulation against metastatic prostate cancer cells (PC-3, LNCaP) were investigated by SRB assay, then, confirmed by ATP assay. Mode of cell death was evaluated by using flow cytometry analyses. A significant improvement of nanoformulated drug was observed at 5-10 μg/ml doses of the drug in both cell lines. More importantly, this formulation enhanced the cytotoxic effect of Etoposide on PC-3 cells, which is considered more resistant to Etoposide than LNCaP and reduced the IC50 value by 55 % reaching to 4.5 μg drug/ml, which is a very significant improvement in the literature. It was clearly shown that nanoformulated drug provided about 3-fold increases in caspase-dependent early apoptotic cells in PC-3 cells. The novel formulation seems to successfully cause cell death of especially PC-3 metastatic prostate cancer cells. It should therefore be taken into consideration for further animal studies as a novel potent anticancer agent.
Collapse
Affiliation(s)
- Merve Erkisa
- Bursa Uludag University, Science and Art Faculty, Department of Biology, 16059, Bursa, Turkey.,Istinye University, Faculty of Medicine, Molecular Cancer Research Center, 34010, Istanbul, Turkey
| | - Ferda Ari
- Bursa Uludag University, Science and Art Faculty, Department of Biology, 16059, Bursa, Turkey
| | - Irem Ulku
- Koc University, Department of Chemistry, 34450, Istanbul, Turkey
| | | | - Yasemin Yar
- Koc University, Materials Science and Engineering, 34450, Istanbul, Turkey
| | - Havva Yagci Acar
- Koc University, Department of Chemistry, 34450, Istanbul, Turkey.,Koc University, Materials Science and Engineering, 34450, Istanbul, Turkey
| | - Engin Ulukaya
- Istinye University, School of Medicine, Department of Clinical Biochemistry, 34010, Istanbul, Turkey
| |
Collapse
|
10
|
Maitland NJ, Frame FM, Rane JK, Erb HH, Packer JR, Archer LK, Pellacani D. Resolution of Cellular Heterogeneity in Human Prostate Cancers: Implications for Diagnosis and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1164:207-224. [PMID: 31576551 DOI: 10.1007/978-3-030-22254-3_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Prostate cancers have a justified reputation as one of the most heterogeneous human tumours. Indeed, there are some who consider that advanced and castration-resistant prostate cancers are incurable, as a direct result of this heterogeneity. However, tumour heterogeneity can be defined in different ways. To a clinician, prostate cancer is a number of different diseases, the treatments for which remain equally heterogeneous and uncertain. To the pathologist, the histopathological appearances of the tumours are notoriously heterogeneous. Indeed, the genius of Donald Gleason in the 1960s was to devise a classification system designed to take into account the heterogeneity of the tumours both individually and in the whole prostate context. To the cell biologist, a prostate tumour consists of multiple epithelial cell types, inter-mingled with various fibroblasts, neuroendocrine cells, endothelial cells, macrophages and lymphocytes, all of which interact to influence treatment responses in a patient-specific manner. Finally, genetic analyses of prostate cancers have been compromised by the variable gene rearrangements and paucity of activating mutations observed, even in large numbers of patient tumours with consistent clinical diagnoses and/or outcomes. Research into familial susceptibility has even generated the least tractable outcome of such studies: the genetic loci are of low penetrance and are of course heterogeneous. By fractionating the tumour (and patient-matched non-malignant tissues) heterogeneity can be resolved, revealing homogeneous markers of patient outcomes.
Collapse
Affiliation(s)
- Norman J Maitland
- Cancer Research Unit, Department of Biology, University of York, York, UK.
| | - Fiona M Frame
- Cancer Research Unit, Department of Biology, University of York, York, UK
| | - Jayant K Rane
- Cancer Research Unit, Department of Biology, University of York, York, UK
| | - Holger H Erb
- Cancer Research Unit, Department of Biology, University of York, York, UK
| | - John R Packer
- Cancer Research Unit, Department of Biology, University of York, York, UK
| | - Leanne K Archer
- Cancer Research Unit, Department of Biology, University of York, York, UK
| | - Davide Pellacani
- Cancer Research Unit, Department of Biology, University of York, York, UK.,Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
11
|
Watanabe K, Kosaka T, Aimono E, Hongo H, Mikami S, Nishihara H, Oya M. Japanese Case of Enzalutamide-Resistant Prostate Cancer Harboring a SPOP Mutation With Scattered Allelic Imbalance: Response to Platinum-Based Therapy. Clin Genitourin Cancer 2019; 17:e897-e902. [PMID: 31296452 DOI: 10.1016/j.clgc.2019.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Keitaro Watanabe
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan.
| | - Eriko Aimono
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Hongo
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Shuji Mikami
- Division of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Ali AS, Grönberg M, Langer SW, Ladekarl M, Hjortland GO, Vestermark LW, Österlund P, Welin S, Grønbæk H, Knigge U, Sorbye H, Janson ET. Intravenous versus oral etoposide: efficacy and correlation to clinical outcome in patients with high-grade metastatic gastroenteropancreatic neuroendocrine neoplasms (WHO G3). Med Oncol 2018; 35:47. [PMID: 29511910 PMCID: PMC5840252 DOI: 10.1007/s12032-018-1103-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 02/23/2018] [Indexed: 01/09/2023]
Abstract
High-grade gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs, G3) are aggressive cancers of the digestive system with poor prognosis and survival. Platinum-based chemotherapy (cisplatin/carboplatin + etoposide) is considered the first-line palliative treatment. Etoposide is frequently administered intravenously; however, oral etoposide may be used as an alternative. Concerns for oral etoposide include decreased bioavailability, inter- and intra-patient variability and patient compliance. We aimed to evaluate possible differences in progression-free survival (PFS) and overall survival (OS) in patients treated with oral etoposide compared to etoposide given as infusion. Patients (n = 236) from the Nordic NEC study were divided into three groups receiving etoposide as a long infusion (24 h, n = 170), short infusion (≤ 5 h, n = 33) or oral etoposide (n = 33) according to hospital tradition. PFS and OS were analyzed with Kaplan-Meier (log-rank), cox proportional hazard ratios and confidence intervals. No statistical differences were observed in PFS or OS when comparing patients receiving long infusion (median PFS 3.8 months, median OS 14.5 months), short infusion (PFS 5.6 months, OS 11.0 months) or oral etoposide (PFS 5.4 months, OS 11.3 months). We observed equal efficacy for the three administration routes suggesting oral etoposide may be safe and efficient in treating high-grade GEP-NEN, G3 patients scheduled for cisplatin/carboplatin + etoposide therapy.
Collapse
Affiliation(s)
- Abir Salwa Ali
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden.
| | - Malin Grönberg
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | - Seppo W Langer
- Departments of Surgery C and Endocrinology PE, Rigshospitalet, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Morten Ladekarl
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Pia Österlund
- Department of Oncology, Tampere University Hospital and Tampere University, Tampere, Finland.,Department of Oncology, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Staffan Welin
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | - Henning Grønbæk
- Departments of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Ulrich Knigge
- Departments of Surgery C and Endocrinology PE, Rigshospitalet, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark.,Department of Oncology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Eva Tiensuu Janson
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| |
Collapse
|