1
|
Ma J, Wang X, Hu Y, Ma J, Ma Y, Chen H, Han Z. Recent Advances in Augmenting the Therapeutic Efficacy of Peptide-Drug Conjugates. J Med Chem 2025; 68:9037-9056. [PMID: 40267310 PMCID: PMC12067445 DOI: 10.1021/acs.jmedchem.5c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
There is an urgent need for the development of safe and effective modalities for the treatment of diseases owing to drug resistance, undesired side effects, and poor clinical outcomes. Combining cell-targeting and efficient cell-killing properties, peptide-drug conjugates (PDCs) have demonstrated superior efficacy compared with peptides and payloads alone. However, innovative molecular designs of PDCs are essential for further improving targeting precision, protease resistance and stability, cell permeability, and overall treatment efficacy. Several strategies have been developed to address these challenges, such as multivalency approaches, bispecific targeting, and long-acting PDCs. Other novel strategies, including overcoming biological barriers, conjugating novel functional payloads, and targeting macropinocytosis, have also shown promise. This perspective compiles the most recent strategies for enhancing PDC treatment efficacy, highlights key advancements in PDC, and provides insights on future directions for the development of novel PDCs.
Collapse
Affiliation(s)
- Jiahui Ma
- Gansu
Provincial Key Laboratory of Environmental Oncology, Department of
Tumor Center, Lanzhou University Second Hospital, Second Clinical
Medical School, Lanzhou University, Lanzhou 730000, China
| | - Xuedan Wang
- School
of Life Sciences and Engineering, Lanzhou
University of Technology, Lanzhou 730050, China
| | - Yonghua Hu
- Gansu
Provincial Key Laboratory of Environmental Oncology, Department of
Tumor Center, Lanzhou University Second Hospital, Second Clinical
Medical School, Lanzhou University, Lanzhou 730000, China
- Gansu
University of Chinese Medicine, Lanzhou 730000, China
| | - Jianping Ma
- School
of Life Sciences and Engineering, Lanzhou
University of Technology, Lanzhou 730050, China
| | - Yaping Ma
- Shenzhen
DIVBIO Pharmaceutical, Shenzhen 518057, China
| | - Hao Chen
- Gansu
Provincial Key Laboratory of Environmental Oncology, Department of
Tumor Center, Lanzhou University Second Hospital, Second Clinical
Medical School, Lanzhou University, Lanzhou 730000, China
| | - Zhijian Han
- Gansu
Provincial Key Laboratory of Environmental Oncology, Department of
Tumor Center, Lanzhou University Second Hospital, Second Clinical
Medical School, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
2
|
Sagar B, Gupta S, Verma SK, Reddy YVM, Shukla S. Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles. Eur J Med Chem 2025; 283:117131. [PMID: 39647418 DOI: 10.1016/j.ejmech.2024.117131] [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: 01/30/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/10/2024]
Abstract
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Collapse
Affiliation(s)
- Bulbul Sagar
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarthak Gupta
- Department of Chemistry, Indian Institute of Technology, Delhi, 110016, New Delhi, India
| | - Sarvesh Kumar Verma
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, 302017, Rajasthan, India
| | | | - Shefali Shukla
- Sri Venkateswara College, University of Delhi, New Delhi, India.
| |
Collapse
|
3
|
Aavani F, Rahimi R, Goleij P, Rezaeizadeh H, Bahramsoltani R. Royal jelly and its hormonal effects in breast cancer: a literature review. Daru 2024; 32:745-760. [PMID: 38717683 PMCID: PMC11555035 DOI: 10.1007/s40199-024-00513-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 04/10/2024] [Indexed: 11/12/2024] Open
Abstract
BACKGROUND Breast cancer is the most common cancer in women which can be cured in most individuals with early-stage non-metastatic disease. Imbalance in estrogen signaling pathways and propagating levels of estrogens has important roles in breast cancer development. Targeting the estrogen receptor signaling pathway is linked to breast cancer treatment. Royal jelly is one of the bee products containing 10-hydroxy-2-decenoic acid, a structure similar to mammalian estrogen, allowing it to attach to estrogen receptors. It is considered as a general tonic and immunomodulator which may be helpful in reducing the side effects of cancer treatments. Currently, there are controversial data regarding the pros and cons of royal jelly in cancer. Here we provide an overview of the effects of royal jelly on sex hormones and its possible role in breast cancer. METHODS Electronic databases including PubMed, Scopus, and Web of Science were searched with the search terms royal jelly, cancer, and sexual hormones. All preclinical and clinical studies regarding the hormonal effects of royal jelly were included. RESULTS According to the collected preclinical data, consumption of royal jelly at daily doses below 200 mg/kg can be useful to decrease the risk of breast cancer since it reduces the serum level of estrogen; whereas increases progesterone, which subsequently decreases the expression of ERs on the ER-positive cells. CONCLUSION Future clinical studies are essential to confirm the safe dose of royal jelly as an adjuvant therapy in breast cancer.
Collapse
Affiliation(s)
- Farzaneh Aavani
- Institute of Experimental and Clinical Pharmacology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roja Rahimi
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, No. 27, North Sarparast, West Taleqani, Felestin Sq., P.O. Box: 1417653761, Tehran, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Sana Institute of Higher Education, Sari, Iran
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Sari, Iran
| | - Hossein Rezaeizadeh
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Roodabeh Bahramsoltani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, No. 27, North Sarparast, West Taleqani, Felestin Sq., P.O. Box: 1417653761, Tehran, Iran.
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
4
|
Garrido MP, Hernandez A, Vega M, Araya E, Romero C. Conventional and new proposals of GnRH therapy for ovarian, breast, and prostatic cancers. Front Endocrinol (Lausanne) 2023; 14:1143261. [PMID: 37056674 PMCID: PMC10086188 DOI: 10.3389/fendo.2023.1143261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
For many years, luteinizing hormone-releasing hormone or gonadotropin-releasing hormone (GnRH) analogs have been used to treat androgen or estrogen-dependent tumors. However, emerging evidence shows that the GnRH receptor (GnRH-R) is overexpressed in several cancer cells, including ovarian, endometrial, and prostate cancer cells, suggesting that GnRH analogs could exert direct antitumoral actions in tumoral tissues that express GnRH-R. Another recent approach based on this knowledge was the use of GnRH peptides for developing specific targeted therapies, improving the delivery and accumulation of drugs in tumoral cells, and decreasing most side effects of current treatments. In this review, we discuss the conventional uses of GnRH analogs, together with the recent advances in GnRH-based drug delivery for ovarian, breast, and prostatic cancer cells.
Collapse
Affiliation(s)
- Maritza P. Garrido
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago, Chile
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Andrea Hernandez
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Margarita Vega
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago, Chile
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Eyleen Araya
- Departamento de Ciencias Quimicas, Facultad de Ciencias Exactas, Universidad Andres Bello, Santiago, Chile
| | - Carmen Romero
- Laboratorio de Endocrinología y Biología de la Reproducción, Hospital Clínico Universidad de Chile, Santiago, Chile
- Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
5
|
Antitumor Activity and Mechanism of Action of Hormonotoxin, an LHRH Analog Conjugated to Dermaseptin-B2, a Multifunctional Antimicrobial Peptide. Int J Mol Sci 2021; 22:ijms222111303. [PMID: 34768734 PMCID: PMC8582938 DOI: 10.3390/ijms222111303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is the most common cancer in men. For patients with advanced or metastatic prostate cancer, available treatments can slow down its progression but cannot cure it. The development of innovative drugs resulting from the exploration of biodiversity could open new therapeutic alternatives. Dermaseptin-B2, a natural multifunctional antimicrobial peptide isolated from Amazonian frog skin, has been reported to possess antitumor activity. To improve its pharmacological properties and to decrease its peripheral toxicity and lethality we developed a hormonotoxin molecule composed of dermaseptin-B2 combined with d-Lys6-LHRH to target the LHRH receptor. This hormonotoxin has a significant antiproliferative effect on the PC3 tumor cell line, with an IC50 value close to that of dermaseptin-B2. Its antitumor activity has been confirmed in vivo in a xenograft mouse model with PC3 tumors and appears to be better tolerated than dermaseptin-B2. Biophysical experiments showed that the addition of LHRH to dermaseptin-B2 did not alter its secondary structure or biological activity. The combination of different experimental approaches indicated that this hormonotoxin induces cell death by an apoptotic mechanism instead of necrosis, as observed for dermaseptin-B2. These results could explain the lower toxicity observed for this hormonotoxin compared to dermaseptin-B2 and may represent a promising targeting approach for cancer therapy.
Collapse
|
6
|
Zhou J, Li Y, Huang W, Shi W, Qian H. Source and exploration of the peptides used to construct peptide-drug conjugates. Eur J Med Chem 2021; 224:113712. [PMID: 34303870 DOI: 10.1016/j.ejmech.2021.113712] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 12/16/2022]
Abstract
Peptide-drug conjugates (PDCs) are a class of novel molecules widely designed and synthesized for delivering payload drugs. The peptide part plays a vital role in the whole molecule, because they determine the ability of the molecules to penetrate the membrane and target to the specific targets. Here, we introduce the source of different kinds of cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs) that have been used or could be used in constructing PDCs as well as their latest application in delivering drugs. What's more, the approaches of developing CPPs and CTPs and the techniques to discover novel peptides are focused on and summarized in the review. This review aims to help relevant researchers fast understand the research status of peptides in PDCs and carry forward the process of novel peptides discovery.
Collapse
Affiliation(s)
- Jiaqi Zhou
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yuanyuan Li
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Wenlong Huang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Wei Shi
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Hai Qian
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China.
| |
Collapse
|
7
|
Tzoupis H, Nteli A, Androutsou ME, Tselios T. Gonadotropin-Releasing Hormone and GnRH Receptor: Structure, Function and Drug Development. Curr Med Chem 2021; 27:6136-6158. [PMID: 31309882 DOI: 10.2174/0929867326666190712165444] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND Gonadotropin-Releasing Hormone (GnRH) is a key element in sexual maturation and regulation of the reproductive cycle in the human organism. GnRH interacts with the pituitary cells through the activation of the Gonadotropin Releasing Hormone Receptors (GnRHR). Any impairments/dysfunctions of the GnRH-GnRHR complex lead to the development of various cancer types and disorders. Furthermore, the identification of GnRHR as a potential drug target has led to the development of agonist and antagonist molecules implemented in various treatment protocols. The development of these drugs was based on the information derived from the functional studies of GnRH and GnRHR. OBJECTIVE This review aims at shedding light on the versatile function of GnRH and GnRH receptor and offers an apprehensive summary regarding the development of different agonists, antagonists and non-peptide GnRH analogues. CONCLUSION The information derived from these studies can enhance our understanding of the GnRH-GnRHR versatile nature and offer valuable insight into the design of new more potent molecules.
Collapse
Affiliation(s)
| | - Agathi Nteli
- Department of Chemistry, University of Patras, Rion GR-26504, Greece
| | - Maria-Eleni Androutsou
- Vianex S.A., Tatoiou Str., 18th km Athens-Lamia National Road, Nea Erythrea 14671, Greece
| | - Theodore Tselios
- Department of Chemistry, University of Patras, Rion GR-26504, Greece
| |
Collapse
|
8
|
Chelariu-Raicu A, Nick A, Urban R, Gordinier M, Leuschner C, Bavisotto L, Molin GZD, Whisnant JK, Coleman RL. A multicenter open-label randomized phase II trial of paclitaxel plus EP-100, a novel LHRH receptor-targeted, membrane-disrupting peptide, versus paclitaxel alone for refractory or recurrent ovarian cancer. Gynecol Oncol 2020; 160:418-426. [PMID: 33243443 DOI: 10.1016/j.ygyno.2020.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE This randomized open-label phase II study evaluated the safety and clinical activity of EP-100 plus weekly paclitaxel in patients with recurrent ovarian cancer expressing positive LHRH receptor. METHODS In a limited "run-in" dose escalation phase for EP-100, six patients were treated with ascending dose levels (13 mg/m2, 20 mg/m2, 30 mg/m2). In the randomized phase, patients received weekly paclitaxel (80 mg/m2 intravenously) plus twice weekly EP-100 (30 mg/m2 intravenously; combination arm) or weekly paclitaxel alone (80 mg/m2 intravenously; paclitaxel arm). The primary study endpoint was overall response rate (ORR). RESULTS Forty-four patients were then randomized to either the experimental combination arm (n = 23) or the standard of care paclitaxel monotherapy arm (n = 21). The ORR was 35% (95%CI 16%-57%) for the combination arm and 33% (95% CI 15%-57%) for the paclitaxel arm. An interesting observation from an unplanned analysis was that a subset of patients with target liver lesions showed a greater overall response rate to the combination (69%) compared to paclitaxel alone (16%). The frequency of treatment-related grade 3-4 adverse events was similar between treatment arms: 48% vs 43% for the combination and paclitaxel arms, respectively. CONCLUSIONS ORR in the EP-100 combination arm was similar to that in the group treated with paclitaxel alone; however, a subset of patients with liver metastases appeared to benefit from the combination. The addition of EP-100 did not appear to augment the adverse event profile of paclitaxel and was well tolerated.
Collapse
Affiliation(s)
- Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Alpa Nick
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Renata Urban
- Department of Gynecologic Oncology, University of Washington Medical Center and Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Mary Gordinier
- Department of Gynecologic Oncology, Norton Cancer Institute, Louisville, KY, USA
| | | | | | | | | | - Robert L Coleman
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
9
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
10
|
Roy J, Kaake M, Srinivasarao M, Low PS. Targeted Tubulysin B Hydrazide Conjugate for the Treatment of Luteinizing Hormone-Releasing Hormone Receptor-Positive Cancers. Bioconjug Chem 2018; 29:2208-2214. [DOI: 10.1021/acs.bioconjchem.8b00164] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
11
|
Zhang L, Ren Y, Wang Y, He Y, Feng W, Song C. Pharmacokinetics, distribution and anti-tumor efficacy of liposomal mitoxantrone modified with a luteinizing hormone-releasing hormone receptor-specific peptide. Int J Nanomedicine 2018. [PMID: 29520138 PMCID: PMC5833774 DOI: 10.2147/ijn.s150512] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background A previous study developed a novel luteinizing hormone-releasing hormone (LHRH) receptor-targeted liposome. The aim of this study was to further assess the pharmacokinetics, biodistribution, and anti-tumor efficacy of LHRH receptor-targeted liposomes loaded with the anticancer drug mitoxantrone (MTO). Methods Plasma and tissue distribution profiles of LHRH receptor-targeted MTO-loaded liposomes (LHRH-MTO-LIPs) were quantified in healthy mice or a xenograft tumor nude mouse model of MCF-7 breast cancer, and were compared with non-targeted liposomes and a free-drug solution. Results The LHRH-MTO-LIPs demonstrated a superior pharmacokinetic profile relative to free MTO. The first target site of accumulation is the kidney, followed by the liver, and then the tumor; maximal tumor accumulation occurs at 4 h post-administration. Moreover, the LHRH-MTO-LIPs exhibited enhanced inhibition of MCF-7 breast cancer cell growth in vivo compared with non-targeted MTO-loaded liposomes (MTO-LIPs) and free MTO. Conclusion The novel LHRH receptor-targeted liposome may become a viable platform for the future targeted treatment of cancer.
Collapse
Affiliation(s)
- Linhua Zhang
- Key Laboratory of Biomedical Material of Tianjin, Institute of Biomedical Engineering, Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| | - Yanqing Ren
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province, China
| | - Yong Wang
- Department of Physics and Chemistry, College of Medicine, Hebei University, Baoding City, Hebei Province, China
| | - Yingna He
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province, China
| | - Wei Feng
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Pharmaceutical College, Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province, China
| | - Cunxian Song
- Key Laboratory of Biomedical Material of Tianjin, Institute of Biomedical Engineering, Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
12
|
Abstract
Gonadotropin-releasing hormone (GnRH) is recognized as the central regulator of the functions of the pituitary-gonadal axis. The increasing knowledge on the mechanisms controlling the development and the function of GnRH-producing neurons is leading to a better diagnostic and therapeutic approach for hypogonadotropic hypogonadisms and for alterations of the puberty onset. During female life span, the function of the GnRH pulse generator may be affected by a number of inputs from other neuronal systems, offering alternative strategies for diagnostic and therapeutic interventions. Moreover, the identification of a GnRH/GnRH receptor system in both human ovary and endometrium has widened the spectrum of action of the peptide outside its hypothalamic functions. The pharmacological use of GnRH itself or its synthetic analogs (agonists and antagonists) provides a valid tool to either stimulate or block gonadotropin secretion and to modulate the female fertility in several reproductive disorders and in assisted reproduction technology. The use of GnRH agonists in young female patients undergoing chemotherapy is also considered a promising therapeutic approach to counteract iatrogenic ovarian failure.
Collapse
|
13
|
Dubovy SR, Fernandez MP, Echegaray JJ, Block NL, Unoki N, Perez R, Vidaurre I, Lee RK, Nadji M, Schally AV. Expression of hypothalamic neurohormones and their receptors in the human eye. Oncotarget 2017; 8:66796-66814. [PMID: 28977997 PMCID: PMC5620137 DOI: 10.18632/oncotarget.18358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/20/2017] [Indexed: 12/27/2022] Open
Abstract
Extrapituitary roles for hypothalamic neurohormones have recently become apparent and clinically relevant, based on the use of synthetic peptide analogs for the treatment of multiple conditions including cancers, pulmonary edema and myocardial infarction. In the eye, it has been suggested that some of these hormones and their receptors may be present in the ciliary body, iris, trabecular meshwork and retina, but their physiological role has yet to be elucidated. Our study intends to comprehensively demonstrate the expression of some hypothalamic neuroendocrine hormones and their receptors within different retinal and extraretinal structures of the human eye. Immunofluorescence, Western blot analysis, and RT-PCR were used to evaluate the qualitative and quantitative expression of Luteinizing Hormone Releasing Hormone (LHRH), Growth Hormone Releasing Hormone (GHRH), Thyrotropin Releasing Hormone (TRH), Gastrin Releasing Peptide (GRP) and Somatostatin as well as their respective receptors (LHRH-R, GHRH-R, TRH-R, GRP-R, SST-R1) in cadaveric human eye tissue and in paraffinized human eye tissue sections. The hypothalamic hormones LHRH, GHRH, TRH, GRP and Somatostatin and their respective receptors (LHRH-R, GHRH-R, TRH-R, GRPR/BB2 and SST-R1), were expressed in the conjunctiva, cornea, trabecular meshwork, ciliary body, lens, retina, and optic nerve.
Collapse
Affiliation(s)
- Sander R Dubovy
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Maria P Fernandez
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jose J Echegaray
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Ophthalmology, University of Puerto Rico School of Medicine, San Juan, PR, USA
| | - Norman L Block
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Noriyuki Unoki
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Roberto Perez
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | - Richard K Lee
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Mehrdad Nadji
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Andrew V Schally
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
14
|
Feng Z, Wen H, Ju X, Bi R, Chen X, Yang W, Wu X. Hormone receptor expression profiles differ between primary and recurrent high-grade serous ovarian cancers. Oncotarget 2017; 8:32848-32855. [PMID: 28416763 PMCID: PMC5464832 DOI: 10.18632/oncotarget.15858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/07/2016] [Indexed: 11/25/2022] Open
Abstract
Hormone receptor status assessment is necessary for selecting cancer patients who might potentially benefit from endocrine therapy. To determine whether hormone receptor status changes during tumor progression, we retrospectively examined 107 high-grade serous ovarian cancer (HGSC) patients with paired primary and recurrent tumor specimens. Hormone receptor expression discordance rates between primary and recurrent tumors were as follows: estrogen receptor (ER) 34.9%, progesterone receptor (PR) 12.4%, androgen receptor (AR) 41.7%, follicle stimulating hormone receptor 46.6%, luteinizing hormone receptor 50.5%, and gonadotropin releasing hormone receptor 20.0%. Hormone receptor discordance was not associated with patient survival. The proportion of the PR-ER+AR- subgroup, which exhibited the worst prognosis, was higher in recurrent than primary tumor specimens. Our study demonstrated that paired primary and recurrent HGSC specimens exhibit differing hormone receptor profiles. Thus, to most effectively identify patient-specific therapies, biomarker status re-assessment is required for recurrent patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Female
- Humans
- Middle Aged
- Neoplasm Grading
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Prognosis
- Receptors, Androgen/metabolism
- Receptors, Estrogen/metabolism
- Receptors, FSH/metabolism
- Receptors, LH/metabolism
- Receptors, LHRH/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Receptors, Progesterone/metabolism
- Receptors, Steroid/metabolism
- Tissue Array Analysis/methods
Collapse
Affiliation(s)
- Zheng Feng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hao Wen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xingzhu Ju
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Rui Bi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaojun Chen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Paris JL, de la Torre P, Victoria Cabañas M, Manzano M, Grau M, Flores AI, Vallet-Regí M. Vectorization of ultrasound-responsive nanoparticles in placental mesenchymal stem cells for cancer therapy. NANOSCALE 2017; 9:5528-5537. [PMID: 28402365 DOI: 10.1039/c7nr01070b] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A new platform constituted by engineered responsive nanoparticles transported by human mesenchymal stem cells is here presented as a proof of concept. Ultrasound-responsive mesoporous silica nanoparticles are coated with polyethylenimine to favor their effective uptake by decidua-derived mesenchymal stem cells. The responsive-release ability of the designed nanoparticles is confirmed, both in vial and in vivo. In addition, this capability is maintained inside the cells used as carriers. The migration capacity of the nanoparticle-cell platform towards mammary tumors is assessed in vitro. The efficacy of this platform for anticancer therapy is shown against mammary tumor cells by inducing the release of doxorubicin only when the cell vehicles are exposed to ultrasound.
Collapse
Affiliation(s)
- Juan L Paris
- Dpto. Química Inorgánica y Bioinorgánica, Facultad de Farmacia, UCM, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, 28040-Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
16
|
Deivaraju C, Temple HT, Block N, Robinson P, Schally AV. LHRH receptor expression in sarcomas of bone and soft tissue. Horm Mol Biol Clin Investig 2017; 28:105-111. [PMID: 27639272 DOI: 10.1515/hmbci-2016-0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/05/2016] [Indexed: 01/16/2023]
Abstract
AIM Luteinizing hormone releasing hormone (LHRH) is a neurohormone, secreted by the hypothalamus, which regulates the secretion of gonadotropins, luteinizing hormone (LH) and follicle stimulating hormone (FSH) from the pituitary. LHRH acts by binding to receptors located in the pituitary gland. These receptors (LHRH receptors) have also been found in the cytoplasm of many tumor cells that involve both the reproductive and non-reproductive organs. These receptors have been demonstrated in prostate and breast cancers, endometrial carcinomas, renal cell carcinoma, lymphoma, carcinoma of liver, pancreas and skin. So far, the expression of LHRH receptors on sarcomas (i.e. malignant tumors of mesenchymal origin) has not been studied, except for endometrial sarcomas. It has also been demonstrated that both LHRH agonists and antagonists can down-regulate these receptors and thus inhibit these tumor cells. Another major therapeutic implication is that these receptors can be targeted specifically by peptides conjugated to anti-cancer drugs. The purpose of this study was to determine if LHRH receptors are expressed in primary and/or metastatic sarcomas of human origin. METHODS We looked at LHRH receptor expression in 38 consecutive sarcoma specimens, using immunohistochemistry. The specimens were either from office biopsy or from resected tumor; these were confirmed as sarcomas by histopathological examination. The receptor staining characteristics and the staining intensity were also documented. The pattern of staining was classified either as "focal or diffuse staining of the cytoplasm" and the intensity of staining was graded on a scale from 1+ to 4+. RESULTS Positive receptor staining was seen in 25 of the 38 (66%) specimens. Twelve of the specimens stained diffusely and 13 had focally positive staining. Three tumors had 1+ staining, 10 had 2+ staining, six had 3+ staining, and six tumors had 4+ staining. The tumors included undifferentiated pleomorphic sarcoma, synovial sarcoma, osteosarcoma, myofibroblastic sarcoma, myxofibrosarcoma, liposarcoma, dermatofibrosarcoma protuberans, metastatic chondrosarcoma and chordoma. CONCLUSION Sarcomas express LHRH receptors with a varying incidence and degree. Our study suggests that those sarcomas that are LHRH receptor positive could potentially be treated with targeted chemotherapy.
Collapse
|
17
|
Feng Z, Wen H, Ju X, Bi R, Chen X, Yang W, Wu X. Expression of hypothalamic-pituitary-gonadal axis-related hormone receptors in low-grade serous ovarian cancer (LGSC). J Ovarian Res 2017; 10:7. [PMID: 28122595 PMCID: PMC5264293 DOI: 10.1186/s13048-016-0300-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023] Open
Abstract
Background The aim of our study was to investigate the clinical features and expression levels of hypothalamic-pituitary-gonadal axis-related hormone receptors in low-grade serous ovarian cancer (LGSC). Methods We retrospectively investigated the clinical features of 26 consecutive patients with LGSC who underwent primary staging or debulking surgery between April 2005 and June 2013 in our center; concomitant primary high-grade serous ovarian cancer (HGSC) patients were randomly selected at a 2:1 ratio for comparison. Tissue microarrays were constructed from the LGSC and HGSC specimens, and the expression levels of six hormone receptors in the hypothalamic pituitary-gonadal axis were analyzed by immunohistochemistry. Results The median (range) age of patients with LGSC was 54 (27–77) years. According to the FIGO staging system, the cases were distributed as follows: stage I, 6 (23.1%); stage II, 0 (0%); stage III, 19 (73.1%); and stage IV, 1 (3.8%). The 2-year and 5-year overall survival rates for LGSC were 91.8% and 67.5%, respectively. The expression levels of the hormone receptors were as follows: ER, 80.8%; PR, 34.6%; AR, 53.8%; FSHR, 84.0%; LHR, 65.4%; and GnRHR, 100%. Hormone receptor-positive patients had a better prognosis compared with hormone receptor-negative patients, but the difference was not significant. Conclusions Our study presented a higher overall survival rate and distinctive hormone receptor expression levels of LGSC patients compared with the HGSC cohort. Patients with positive hormone receptor expression tended to have a better prognosis than the corresponding hormone receptor negative patients. Electronic supplementary material The online version of this article (doi:10.1186/s13048-016-0300-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zheng Feng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Hao Wen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China
| | - Xingzhu Ju
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Rui Bi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaojun Chen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
18
|
Preclinical Evaluation of an Epidermal Growth Factor Receptor-Targeted Doxorubicin-Peptide Conjugate: Toxicity, Biodistribution, and Efficacy in Mice. J Pharm Sci 2016; 105:639-649. [PMID: 26869425 DOI: 10.1016/j.xphs.2015.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/30/2015] [Accepted: 10/16/2015] [Indexed: 01/14/2023]
Abstract
Doxorubicin (DOX) is known to induce apoptosis and necrosis in healthy tissue resulting in unwanted toxicities. To improve the ability of DOX to more specifically target tumors and minimize undesirable side effects, conjugation of DOX with epidermal growth factor receptor (EGFR)--binding peptide (DOX-EBP) has been developed to deliver DOX to EGFR-overexpressing neoplastic cells. Here, we investigated whether DOX-EBP was able to reduce toxicity and enhance anticancer efficacy in vivo through receptor-mediated targeted delivery system. Nude mice were treated with DOX or DOX-EBP to estimate general toxicity, normal tissue damage, biodistribution, and antitumor efficacy. In addition, the expression levels of EGFR in tumor tissues and normal organs were investigated by Western blotting, and their mRNA expression was analyzed by reverse transcription PCR. This study demonstrated that DOX-EBP was able to effectively decrease the distribution of DOX in normal tissues without EGFR overexpressing and reduce DOX-induced toxicity. On the other hand, the research also confirmed that DOX-EBP was able to preferentially accumulate DOX in EGFR-overexpressing tumor tissues and showed the enhanced anticancer efficacy over free DOX. DOX-EBP could be used for receptor-targeted chemotherapy with less toxicity and greater efficacy of tumor cells overexpressing EGFR. DOX-EBP conjugate is a good therapeutic agent for cancer treatment.
Collapse
|
19
|
Ye H, Liu X, Sun J, Zhu S, Zhu Y, Chang S. Enhanced therapeutic efficacy of LHRHa-targeted brucea javanica oil liposomes for ovarian cancer. BMC Cancer 2016; 16:831. [PMID: 27793127 PMCID: PMC5086058 DOI: 10.1186/s12885-016-2870-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 10/21/2016] [Indexed: 02/06/2023] Open
Abstract
Background Although brucea javanica oil liposomes (BJOLs) have been used clinically to treat ovarian cancer, its clinical efficacy is often limited by systemic side effects due to non-specific distribution. Luteinizing hormone releasing hormone receptor (LHRHR) is overexpressed in most ovarian cancers but negligibly expressed in most of the other visceral organs. In this study, we aimed to develop a novel LHRHa targeted and BJO-loaded liposomes (LHRHa-BJOLs), and investigate its characteristics, targeting ability and anti-ovarian cancer efficiency both in vitro and in vivo. Methods The LHRHa-BJOLs were prepared by film-dispersion and biotin-streptavidin linkage methods, and characterized in terms of its morphology, particle size, zeta potential, ligand conjugation, encapsulation efficiency and stability. The targeting nature and antitumor effects of the liposomes were evaluated in vitro using cultured human ovarian cancer A2780/DDP cells, and in vivo using ovarian cancer-bearing nude mice. Results The LHRHa-BJOLs were successfully synthesized, with a uniformly spherical shape, appropriate particle size and zeta potential, as well as a high encapsulation efficiency. Compared to non-targeted liposomes and BJO emulsion, the LHRHa-BJOLs could significantly increase specific intracellular uptaking rate, enhance cell inhibitory effect and induce cell apoptosis in A2780/DDP cells in vitro. Meanwhile, LHRHa-BJOLs also had a significantly stronger activity of targeting tumor tissue, inhibiting tumor growth, inducing tumor apoptosis and prolonging survival time in ovarian cancer-bearing mice in vivo. Conclusions Our experiment suggests that LHRHa-BJOLs may be a useful targeted drug for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Hongxia Ye
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Xiaojuan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Shenyin Zhu
- Department of Pharmacy, First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong district, Chongqing, 400010, China
| | - Yi Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China
| | - Shufang Chang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong district, Chongqing, 400010, China.
| |
Collapse
|
20
|
Feng Z, Wen H, Bi R, Ju X, Chen X, Yang W, Wu X. A clinically applicable molecular classification for high-grade serous ovarian cancer based on hormone receptor expression. Sci Rep 2016; 6:25408. [PMID: 27139372 PMCID: PMC4853732 DOI: 10.1038/srep25408] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/18/2016] [Indexed: 11/18/2022] Open
Abstract
To establish an effective hormone receptor-based molecular classification of high-grade serous ovarian cancer (HGSC), we retrospectively examined 875 consecutive HGSC patients who underwent primary surgery at our hospital and constructed tissue microarrays from these specimens. The expression levels of the hormone receptors were as follows: ER 64.4%, PR 12.6%, AR 35.6%, FSHR 54.5%, LHR 34.8%, and GnRHR 88.3%. Based on clustering of their expression patterns, we classified patients into five subgroups with distinctive clinical features (PR+, PR - ER + AR+, PR - ER + AR-, PR - ER - AR+, and PR - ER - AR-). Patients in the PR + group were younger compared to those in the other groups (p < 0.001). More patients were of advanced stage in the PR - ER + AR- group than the other groups (p = 0.020). A greater proportion of patients were sensitive to platinum-based chemotherapy in the PR - ER - AR + group compared with the other groups (p = 0.034). A trend of increasing risk of death was observed among these subgroups (p < 0.001). In the multivariate analysis, patients also had orderly increased hazard ratios for death in the PR + (HR = 2.256, 95% CI, 0.983-5.175), PR - ER + AR + (HR = 2.188, 95% CI, 1.004-4.796), PR - ER - AR- (HR = 2.316, 95% CI, 1.097-5.082) and PR - ER + AR- (HR = 2.928, 95% CI, 1.366-6.276) subgroups compared to the PR - ER - AR+ subgroup. Our classification could help predict patient clinical outcomes, guide individual treatments and stratify patients in future clinical trials.
Collapse
Affiliation(s)
- Zheng Feng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hao Wen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Rui Bi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xingzhu Ju
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaojun Chen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
21
|
Li W, Zhang L, Zhu Y, Zhang J, Shen L, Huang S, Fang S. Improving ovarian cancer imaging with LHRH-NBs: an experimental study. Arch Gynecol Obstet 2016; 294:631-8. [PMID: 27075493 PMCID: PMC4981623 DOI: 10.1007/s00404-016-4092-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/30/2016] [Indexed: 11/24/2022]
Abstract
Purpose Our previous study used freeze-drying and biotin–avidin binding methods and obtained nontargeted nanobubbles (N-NBs) and ovarian cancer-targeting nanobubbles (LHRH-NBs, luteinizing hormone-releasing hormone nanobubbles). Our study also identified the physical and chemical properties of these two contrast agents, and validated the targeting ability and underlying mechanisms of LHRH-NBs in vitro. The present study investigated the imaging of N-NBs and LHRH-NBs in nude mice and their binding with tissues. Methods The nude mice models of xenografts were divided into three groups, N-NB, LHRH-NB, and SonoVue. These contrast agents were injected via the caudal vein to observe the imaging of ovarian cancer. Fluorescence microscope was used to observe the penetration of N-NBs and LHRH-NBs through the vascular endothelial gaps. Immunofluorescence was used to observe the penetration of N-NBs and LHRH-NBs through vascular endothelial gaps and binding to the tumor cells. Results The imaging intensity and duration were not significantly different between N-NBs and LHRH-NBs. The imaging intensity in the N-NB and LHRH-NB groups was not significantly different compared with the SonoVue group; however, the imaging duration in the N-NB and LHRH-NB groups was significantly longer than in the SonoVue group (P < 0.001). Both N-NBs and LHRH-NBs penetrated through the vascular endothelial gaps. After penetrating through the vascular endothelial gapes, LHRH-NBs could target and bind to the tumor cells. Conclusions N-NBs and LHRH-NBs are of good imaging effectiveness and relatively long imaging duration. LHRH-NB is a potent contrast agent for imaging ovarian cancer, while achieving targeted delivery of drugs to the site of ovarian cancer.
Collapse
Affiliation(s)
- Wenjuan Li
- Department of Obstetrics and Gynaecology, Wuxi Maternal and Child Health Hospital, Wuxi, 214002, Jiangshu, People's Republic of China
| | - Lingping Zhang
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, 518133, Guangdong, People's Republic of China
| | - Yuanfang Zhu
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, 518133, Guangdong, People's Republic of China. .,Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Jinyi Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Li Shen
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shuying Huang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shanyu Fang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| |
Collapse
|
22
|
Maggi R, Cariboni AM, Marelli MM, Moretti RM, Andrè V, Marzagalli M, Limonta P. GnRH and GnRH receptors in the pathophysiology of the human female reproductive system. Hum Reprod Update 2015; 22:358-81. [PMID: 26715597 DOI: 10.1093/humupd/dmv059] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Human reproduction depends on an intact hypothalamic-pituitary-gonadal (HPG) axis. Hypothalamic gonadotrophin-releasing hormone (GnRH) has been recognized, since its identification in 1971, as the central regulator of the production and release of the pituitary gonadotrophins that, in turn, regulate the gonadal functions and the production of sex steroids. The characteristic peculiar development, distribution and episodic activity of GnRH-producing neurons have solicited an interdisciplinary interest on the etiopathogenesis of several reproductive diseases. The more recent identification of a GnRH/GnRH receptor (GnRHR) system in both the human endometrium and ovary has widened the spectrum of action of the peptide and of its analogues beyond its hypothalamic function. METHODS An analysis of research and review articles published in international journals until June 2015 has been carried out to comprehensively summarize both the well established and the most recent knowledge on the physiopathology of the GnRH system in the central and peripheral control of female reproductive functions and diseases. RESULTS This review focuses on the role of GnRH neurons in the control of the reproductive axis. New knowledge is accumulating on the genetic programme that drives GnRH neuron development to ameliorate the diagnosis and treatment of GnRH deficiency and consequent delayed or absent puberty. Moreover, a better understanding of the mechanisms controlling the episodic release of GnRH during the onset of puberty and the ovulatory cycle has enabled the pharmacological use of GnRH itself or its synthetic analogues (agonists and antagonists) to either stimulate or to block the gonadotrophin secretion and modulate the functions of the reproductive axis in several reproductive diseases and in assisted reproduction technology. Several inputs from other neuronal populations, as well as metabolic, somatic and age-related signals, may greatly affect the functions of the GnRH pulse generator during the female lifespan; their modulation may offer new possible strategies for diagnostic and therapeutic interventions. A GnRH/GnRHR system is also expressed in female reproductive tissues (e.g. endometrium and ovary), both in normal and pathological conditions. The expression of this system in the human endometrium and ovary supports its physiological regulatory role in the processes of trophoblast invasion of the maternal endometrium and embryo implantation as well as of follicular development and corpus luteum functions. The GnRH/GnRHR system that is expressed in diseased tissues of the female reproductive tract (both benign and malignant) is at present considered an effective molecular target for the development of novel therapeutic approaches for these pathologies. GnRH agonists are also considered as a promising therapeutic approach to counteract ovarian failure in young female patients undergoing chemotherapy. CONCLUSIONS Increasing knowledge about the regulation of GnRH pulsatile release, as well as the therapeutic use of its analogues, offers interesting new perspectives in the diagnosis, treatment and outcome of female reproductive disorders, including tumoral and iatrogenic diseases.
Collapse
Affiliation(s)
- Roberto Maggi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Anna Maria Cariboni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Valentina Andrè
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| |
Collapse
|
23
|
Seitz S, Buchholz S, Schally AV, Weber F, Klinkhammer-Schalke M, Inwald EC, Perez R, Rick FG, Szalontay L, Hohla F, Segerer S, Kwok CW, Ortmann O, Engel JB. Triple negative breast cancers express receptors for LHRH and are potential therapeutic targets for cytotoxic LHRH-analogs, AEZS 108 and AEZS 125. BMC Cancer 2014; 14:847. [PMID: 25410881 PMCID: PMC4289186 DOI: 10.1186/1471-2407-14-847] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 08/25/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is a distinct subtype of breast cancer burdened with a dismal prognosis due to the lack of effective therapeutic agents. Receptors for LHRH (luteinizing hormone-releasing hormone) can be successfully targeted with AEZS-108 [AN-152], an analog of LHRH conjugated to doxorubicin. Our study evaluates the presence of this target LHRH receptor in human specimens of TNBC and investigates the efficacy and toxicity of AEZS-108 in vivo. We also studied in vitro activity of AEZS-125, a new LHRH analog conjugated with the highly potent natural compound, Disorazol Z. METHODS 69 human surgical specimens of TNBC were investigated for LHRH-R expression by immunohistochemistry. Expression of LHRH-R in two TNBC cell lines was evaluated by real time RT-PCR. Cytotoxicity of AEZS-125 was evaluated by Cell Titer Blue cytoxicity assay. LHRH- receptor expression was silenced with an siRNA in both cell lines. For the in vivo experiments an athymic nude mice model xenotransplanted with the cell lines, MDA-MB-231 and HCC 1806, was used. The animals were randomised to three groups receiving solvent only (d 1, 7, 14, i.v.) for control, AEZS-108 (d 1, 7, 14, i.v.) or doxorubicin at an equimolar dose (d 1, 7, 14, i.v.). RESULTS In human clinical specimens of TNBC, expression of the LHRH-receptor was present in 49% (n = 69).HCC 1806 and MDA-MB-231 TNBC cells expressed mRNA for the LHRH-receptor. Silencing of the LHRH-receptor significantly decreased the cytotoxic effect of AEZS-108. MDA-MB-231 and HCC 1806 tumors xenografted into nude mice were significantly inhibited by treatment with AEZS-108; doxorubicin at equimolar doses was ineffective.As compared to AEZS 108, the Disorazol Z - LHRH conjugate, AEZS-125, demonstrated an increased cytotoxicity in vitro in HCC 1806 and MDA-MB-231 TNBC; this was diminished by receptor blockade with synthetic LHRH agonist (triptorelin) pretreatment. CONCLUSION The current study confirms that LHRH-receptors are expressed by a significant proportion of TNBC and can be successfully used as homing sites for cytotoxic analogs of LHRH, such as AEZS-108 and AEZS-125.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jörg Bernhard Engel
- Depertment of Obsteterics and Gynecology, Medical University of Gießen, 35392 Gießen, Germany.
| |
Collapse
|
24
|
Montagnani Marelli M, Manea M, Moretti RM, Marzagalli M, Limonta P. Oxime bond-linked daunorubicin-GnRH-III bioconjugates exert antitumor activity in castration-resistant prostate cancer cells via the type I GnRH receptor. Int J Oncol 2014; 46:243-53. [PMID: 25351635 DOI: 10.3892/ijo.2014.2730] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 08/07/2014] [Indexed: 11/05/2022] Open
Abstract
It is well established that gonadotropin-releasing hormone receptors (GnRH-R) are expressed in different types of cancers, including castration-resistant prostate cancer (CRPC) and mediate the antiproliferative effect of GnRH analogs. Thus, these compounds are employed as targeting moieties to selectively deliver chemotherapeutic agents to cancer cells. GnRH-III, the decapeptide isolated from the sea lamprey brain, has lower potency than GnRH in stimulating gonadotropin secretion, but it exerts antiproliferative effects on many tumors expressing the GnRH-R. GnRH-III-based peptides are considered promising targeting moieties for the preparation of anticancer drug delivery systems. These studies were aimed at i) evaluating the antitumor activity of two cytotoxic oxime bond-linked daunorubicin (Dau)-GnRH-III derivative bioconjugates (Dau-GnRH-III, in which daunorubicin was coupled to the 8Lys in the native form of GnRH-III, and Dau-[4Lys(Ac)]-GnRH-III, in which daunorubicin was attached to the 8Lys of a GnRH-III derivative where 4Ser was replaced by an acetylated lysine) on CRPC cells; and ii) to elucidate the involvement of the classical GnRH-R (type I GnRH-R) in this antitumor activity. Our results demonstrated that both Dau-GnRH-III and Dau-[4Lys(Ac)]-GnRH-III were rapidly internalized into DU145 prostate cancer cells and exerted a significant cytostatic effect. Both bioconjugates increased the levels of the active form of caspase-3, indicating the involvement of apoptosis in their antitumor activity. The antiproliferative effect of both Dau-GnRH-III and Dau-[4Lys(Ac)]-GnRH-III was counteracted by the simultaneous treatment of the cells with Antide, an antagonist of the GnRH-R. Moreover, after silencing the type I GnRH-R the antitumor activity of both bioconjugates was completely abolished. These data demonstrate that in CRPC cells, daunorubicin-GnRH-III derivative bioconjugates: i) inhibit tumor cell proliferation, by triggering the apoptosis process; ii) exert their antitumor effect through the activation of the type I GnRH-R expressed on these cells. Cytotoxic-GnRH-III derivative may represent promising targeted chemotherapeutics for the treatment of CRPC patients.
Collapse
Affiliation(s)
| | - Marilena Manea
- Department of Chemistry, University of Konstanz, D-78457 Konstanz, Germany
| | - Roberta M Moretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, I-20133 Milan, Italy
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, I-20133 Milan, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, University of Milan, I-20133 Milan, Italy
| |
Collapse
|
25
|
Kwok CW, Treeck O, Buchholz S, Seitz S, Ortmann O, Engel JB. Receptors for luteinizing hormone-releasing hormone (GnRH) as therapeutic targets in triple negative breast cancers (TNBC). Target Oncol 2014; 10:365-73. [DOI: 10.1007/s11523-014-0340-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 09/25/2014] [Indexed: 12/25/2022]
|
26
|
Gajadeera C, Willby MJ, Green KD, Shaul P, Fridman M, Garneau-Tsodikova S, Posey JE, Tsodikov OV. Antimycobacterial activity of DNA intercalator inhibitors of Mycobacterium tuberculosis primase DnaG. J Antibiot (Tokyo) 2014; 68:153-7. [PMID: 25248725 DOI: 10.1038/ja.2014.131] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/29/2014] [Accepted: 08/13/2014] [Indexed: 11/09/2022]
Abstract
Owing to the rise in drug resistance in tuberculosis combined with the global spread of its causative pathogen, Mycobacterium tuberculosis (Mtb), innovative anti mycobacterial agents are urgently needed. Recently, we developed a novel primase-pyrophosphatase assay and used it to discover inhibitors of an essential Mtb enzyme, primase DnaG (Mtb DnaG), a promising and unexplored potential target for novel antituberculosis chemotherapeutics. Doxorubicin, an anthracycline antibiotic used as an anticancer drug, was found to be a potent inhibitor of Mtb DnaG. In this study, we investigated both inhibition of Mtb DnaG and the inhibitory activity against in vitro growth of Mtb and M. smegmatis (Msm) by other anthracyclines, daunorubicin and idarubicin, as well as by less cytotoxic DNA intercalators: aloe-emodin, rhein and a mitoxantrone derivative. Generally, low-μM inhibition of Mtb DnaG by the anthracyclines was correlated with their low-μM minimum inhibitory concentrations. Aloe-emodin displayed threefold weaker potency than doxorubicin against Mtb DnaG and similar inhibition of Msm (but not Mtb) in the mid-μM range, whereas rhein (a close analog of aloe-emodin) and a di-glucosylated mitoxantrone derivative did not show significant inhibition of Mtb DnaG or antimycobacterial activity. Taken together, these observations strongly suggest that several clinically used anthracyclines and aloe-emodin target mycobacterial primase, setting the stage for a more extensive exploration of this enzyme as an antibacterial target.
Collapse
Affiliation(s)
- Chathurada Gajadeera
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Melisa J Willby
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Keith D Green
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Pazit Shaul
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Micha Fridman
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | | | - James E Posey
- Division of Tuberculosis Elimination, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
27
|
Zhang J, Liu S, Zhu Y, Zhang L, Li W, Wang F, Huang S. Preparation and characterization of luteinising-hormone releasing hormone nanoliposomal microbubbles specifically targeting ovarian cancer cells in vitro. Mol Med Rep 2014; 10:567-71. [PMID: 24805264 DOI: 10.3892/mmr.2014.2211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 03/25/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to prepare luteinizing-hormone releasing hormone (LHRH) nanoliposomal microbubbles specifically targeting ovarian cancer cells. The lyophilization/sonication method was used to prepare non-targeting nanoliposomal microbubbles (N-N-Mbs). Using the biotin-avidin bridge method, conjugated LHRH antibodies to N-N-Mbs generated LHRH nanoliposomal microbubbles (LHRH-N-Mbs) specifically targeting ovarian cancer cells. The morphology and physicochemical properties of the microbubbles was detected using an optical microscope and zeta detector. The binding affinity between the secondary antibody and LHRH-N-Mbs or N-N-Mbs was determined by flow cytometry. The binding of LHRH-N-Mb to human ovarian cancer cells (OVCAR-3) was detected by light microscopy. The rounded and uniformly distributed N-N-Mbs and LHRH-N-Mbs were successfully generated. The particle size ranged from 295-468 nm with a mean of 360 nm for N-N-Mbs or 369-618 nm with a mean of 508 nm for LHRH-N-Mbs. There was a significant difference in size between the two groups (P<0.05), although the surface potential of the two microbubbles remained the same (-14.6 mV). Following being kept at room temperature for 14 days, no significant difference in the physicochemical properties of the LHRH-N-Mbs was detected compared with that of freshly prepared microbubbles. The secondary antibody binding rate of LHRH-N-Mbs and N-N-Mbs was 75.6 and 0.83%, respectively. Furthermore, the formation of a rosette-like structure surrounding OVCAR-3 cells was observed after the cells were incubated with LHRH-N-Mbs, whereas pre-incubation with LHRH antibody blocked this rosette formation. In conclusion, LHRH-N-Mbs specifically targeting ovarian cancer cells were successfully prepared through biotin-avidin mediation and the lyophilization/sonication method. The key feature of LHRH-N-Mbs is their small size, stability and high efficiency in targeting human OVCAR-3 cells in vitro.
Collapse
Affiliation(s)
- Jinyi Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Sisun Liu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuanfang Zhu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Liping Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenjuan Li
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fen Wang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shuying Huang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
28
|
Tsui KH, Lee WL, Seow KM, Yang LW, Wang SY, Wang PH, Chang CL, Yen MS, Cheng JT, Chen CP. Effect of gonadotropin-releasing hormone agonist on ES-2 ovarian cancer cells. Taiwan J Obstet Gynecol 2014; 53:35-42. [PMID: 24767644 DOI: 10.1016/j.tjog.2013.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2013] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Gonadotropin-releasing hormone (GnRH) receptor is found in the ovarian tissue, including epithelial ovarian cancer (EOC), suggesting that GnRH agonists may have direct action on EOC. MATERIALS AND METHODS Ovarian clear cell cancer (ES-2) cells were treated with low-dose GnRH agonist with/without low-dose paclitaxel (1 μM D-Lys(6) with/without 0.5 μM or 1.0 μM paclitaxel). Growth and behavior of ES-2 cells were evaluated. RESULTS Use of either D-Lys(6) or paclitaxel or a combination of the two did not affect the morphology and growth pattern of ES-2 cells. However, ability of migration and invasion of ES-2 cells was significantly decreased in either use of D-Lys(6) or paclitaxel and more apparent with the combination. Type I GnRH receptor expression of ES-2 was not altered significantly by the combination. CONCLUSION GnRH agonist might modify the ES-2 ovarian cancer cells, and its role might be independent, additional or synergistic, suggesting the potential role of the use of GnRH agonist in the management of clear cell type of the ovarian cancer. However, the results of this study were derived using ES-2 ovarian cancer cells, and might not be valid in other cell types of ovarian cancers.
Collapse
Affiliation(s)
- Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Wen-Ling Lee
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan; Department of Nursing, Oriental Institute of Technology, New Taipei City, Taiwan
| | - Kok-Min Seow
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Shih Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Lin-Wei Yang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Yi Wang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Immunology Center, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| | - Chi-Lun Chang
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Shyen Yen
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jiin-Tsuey Cheng
- Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chih-Ping Chen
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, Taiwan; Department of Obstetrics and Gynecology, Mackay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| |
Collapse
|
29
|
Targeted paclitaxel nanoparticles modified with follicle-stimulating hormone β 81-95 peptide show effective antitumor activity against ovarian carcinoma. Int J Pharm 2013; 453:498-505. [PMID: 23811008 DOI: 10.1016/j.ijpharm.2013.06.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 04/28/2013] [Accepted: 06/12/2013] [Indexed: 02/07/2023]
Abstract
The majority of patients with advanced ovarian cancer will experience a relapse and ultimately die from refractory diseases. Targeted therapy shows promise for these patients. Novel therapeutic strategies should be developed on the basis of the molecular mechanisms involved in ovarian cancer and the steroid hormone environment of ovaries. The ovary is the main target organ of follicle-stimulating hormone (FSH), which bind to its receptor with high affinity. In this study a FSH receptor-targeting ligand, FSH β 81-95 peptide, was used as a targeting moiety to synthesize an FSH receptor-mediated drug delivery system. FSH β 81-95 peptide-conjugated nanoparticles (FSH81-NPs) and paclitaxel-loaded FSH81-NPs (FSH81-NP-PTXs) were synthesized. In vitro studies showed that FSH β 81-95 peptide enabled the specific uptake of cytotoxic drugs and increased the intracellular paclitaxel concentration in FSH receptor-expressing cancer cells, resulting in enhanced cytotoxic effects. In vivo studies showed that FSH81-NP-PTXs possessed higher antitumor efficacy against FSH receptor-expressing tumors without any clinical signs of adverse side effects or body weight loss due to modification with FSH β 81-95 peptide. Therefore, FSH binding peptide-targeted drug delivery system exhibited high potential in the treatment of ovarian cancer, and tumor targeting via reproductive hormone receptors might improve the outcome of diseases.
Collapse
|