1
|
Chapeau D, Beekman S, Piet A, Li L, de Ridder C, Stuurman D, Seimbille Y. eSOMA-DM1, a Maytansinoid-Based Theranostic Small-Molecule Drug Conjugate for Neuroendocrine Tumors. Bioconjug Chem 2024; 35:1823-1834. [PMID: 39395152 DOI: 10.1021/acs.bioconjchem.4c00413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Background: The main challenges of conventional chemotherapy lie in its lack of selectivity and specificity, leading to significant side effects. Using a small-molecule drug conjugate (SMDC) ensures specific delivery of a cytotoxic drug to the tumor site by coupling it to a targeting vector. This promising strategy can be applied to neuroendocrine tumors (NETs) by choosing a targeting vector that binds specifically to somatostatin receptor subtype 2 (SSTR2). Additionally, incorporation of a bifunctional chelate into the molecule enables complexation of both diagnostic and therapeutic radionuclides. Thus, it facilitates monitoring of the distribution of the SMDC in the body and allows for the implementation of combination therapy. In our study, we designed eSOMA-DM1, a SMDC combining the SSTR2-targeted octreotate peptide and the cytotoxic agent DM1 via a chelate-bridged linker (N3-Py-DOTAGA). This approach warrants conjugation of the targeting vector and the drug at opposite sites to avoid undesired steric hindrance effects. Methods: Synthesis of the DM1 moiety (4) involved a three-step synthetic route, followed by the conjugation to the cyclic peptide, N3-Py-DOTAGA-d-Phe-cyclo[Cys-Tyr-d-Trp-Lys-Thr-Cys]-Thr-OH, through a copper-free click reaction, resulting in eSOMA-DM1. Subsequent labeling with [111In]InCl3 gave a high radiochemical yield and purity. In vitro assessments of eSOMA-DM1 binding, uptake, and internalization were conducted in SSTR2-transfected U2OS cells. Ex vivo biodistribution and fluorescence imaging were performed in H69-tumor bearing mice. Results: eSOMA-DM1 exhibited an IC50 value for SSTR2 similar to the gold standard DOTA-TATE. The uptake of [111In]In-eSOMA-DM1 in U2OS.SSTR2 cells was 1.2-fold lower than that of [111In]In-DOTA-TATE. Tumor uptake in H69-xenografted mice was higher for [111In]In-eSOMA-DM1 at all-time points compared to [111In]In-DOTA-TATE. Prolonged blood circulation led to increased accumulation of [111In]In-eSOMA-DM1 in highly vascularized tissues, such as the lungs, skin, and heart. Excretion through the kidneys, liver, and spleen was also observed. Conclusion: eSOMA-DM1 is a SMDC developed for NET showing promising characteristics in vitro. However, the in vivo results obtained with [111In]In-eSOMA-DM1 suggest the need for adjustments to optimize its distribution.
Collapse
Affiliation(s)
- Dylan Chapeau
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Amber Piet
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Le Li
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam 3015 GD, the Netherlands
- Erasmus MC Cancer Institute, Rotterdam 3015 GD, the Netherlands
| |
Collapse
|
2
|
De K, Tanbir SKE, Sinha S, Mukhopadhyay S. Lipid-Based Nanocarrier by Targeting with LHRH Peptide: A Promising Approach for Prostate Cancer Radio-Imaging and Therapy. Mol Pharm 2024; 21:4128-4146. [PMID: 38920398 DOI: 10.1021/acs.molpharmaceut.4c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Prostate cancer is a prevalently detected malignancy with a dismal prognosis. Luteinizing-hormone-releasing-hormone (LHRH) receptors are overexpressed in such cancer cells, to which the LHRH-decapeptide can specifically bind. A lipid-polyethylene glycol-conjugated new LHRH-decapeptide analogue (D-P-HLH) was synthesized and characterized. D-P-HLH-coated and anticancer drug doxorubicin (DX)-loaded solid lipid nanoparticles (F-DX-SLN) were formulated by the cold homogenization technique and characterized by Fourier transform infrared spectroscopy, X-ray diffraction, X-ray photoelectron spectroscopy, differential scanning calorimetry, dynamic light scattering, electron microscopy, entrapment efficiency, and drug-release profile studies. F-DX-SLN allows site-specific DX delivery by reducing the side effects of chemotherapy. Cancer cells could precisely take up F-DX-SLN by targeting specific receptors, boosting the cytotoxicity at the tumor site. The efficacy of F-DX-SLN on PC3/SKBR3 cells by the MTT assay revealed that F-DX-SLN was more cytotoxic than DX and/or DX-SLN. Flow cytometry and confocal microscopic studies further support F-DX-SLNs' increased intracellular absorption capability in targeting LHRH overexpressed cancer cells. F-DX-SLN ensured high apoptotic potential, noticeably larger mitochondrial transmembrane depolarization action, as well as the activation of caspases, a longer half-life, and greater plasma concentration. F-DX-SLN/DX-SLN was radiolabeled with technetium-99m; scintigraphic imaging studies established its tumor selectivity in PC3 tumor-bearing nude mice. The efficacy of the formulations in cancer treatment, in vivo therapeutic efficacy tests, and histopathological studies were also conducted. Results clearly indicate that F-DX-SLN exhibits sustained and superior targeted administration of anticancer drugs, thus opening up the possibility of a drug delivery system with precise control and targeting effects. F-DX-SLN could also provide a nanotheranostic approach with improved efficacy for prostate cancer therapy.
Collapse
Affiliation(s)
- Kakali De
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - S K Eashayan Tanbir
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Samarendu Sinha
- Netaji Subhas Chandra Bose Cancer Hospital, 3081 Nayabad, Kolkata 700094, India
| | - Soma Mukhopadhyay
- Netaji Subhas Chandra Bose Cancer Hospital, 3081 Nayabad, Kolkata 700094, India
| |
Collapse
|
3
|
Resta C, Moustogiannis A, Chatzinikita E, Malligiannis Ntalianis D, Malligiannis Ntalianis K, Philippou A, Koutsilieris M, Vlahos N. Gonadotropin-Releasing Hormone (GnRH)/GnRH Receptors and Their Role in the Treatment of Endometriosis. Cureus 2023; 15:e38136. [PMID: 37122983 PMCID: PMC10145781 DOI: 10.7759/cureus.38136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/02/2023] Open
Abstract
Endometriosis, defined as the development of endometrial tissue outside of the uterine cavity, is a common gynecological disorder. The prevalence of pelvic endometriosis approaches 6%-10% in the general female population, and in women with pain, infertility, or both, the frequency is 35%-50%. The gold standard recommended process for diagnosing endometriosis is laparoscopy, an invasive surgical procedure, with or without histologic verification. The currently available nonsurgical treatments include oral contraceptives (estrogen-progestogen preparations), progestogen preparations (containing progesterone derivatives), androgenic hormones (danazol), and gonadotropin-releasing hormone (GnRH) agonists and antagonists. Two GnRH types have been discovered in mammals, GnRH I and GnRH II. In particular, GnRH I is released by the hypothalamus; however, it can be present in various tissues and organs of the body, including neural tissue, where it exerts neuroendocrine, autocrine, and paracrine actions in the peripheral and central nervous system (CNS). Interestingly, another GnRH isoform, GnRH III, has been identified, which has 60% similarity with GnRH I from which it varies by four amino acids. This peptide has been shown to have a significant role in reproduction, specifically in gametogenesis and steroidogenesis. Further research is needed to identify innovative treatment options for endometriosis, such as the therapeutic exogenous administration of GnRH II or antagonists of the GnRH I receptor. In this review, we examined the role of GnRH in endometriosis, outlining the specific actions of GnRH and GnRH receptors (GnRHRs). The innovative use of GnRH analogs and antagonists in the treatment of endometriosis is also discussed.
Collapse
Affiliation(s)
- Christina Resta
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
- Second Department of Obstetrics and Gynaecology, Aretaieion Hospital, University of Athens, Athens, GRC
| | - Athanasios Moustogiannis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | - Eirini Chatzinikita
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | | | | | - Anastasios Philippou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, GRC
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynaecology, Aretaieion Hospital, University of Athens, Athens, GRC
| |
Collapse
|
4
|
Pryyma A, Matinkhoo K, Bu YJ, Merkens H, Zhang Z, Bénard F, Perrin DM. Synthesis and preliminary evaluation of octreotate conjugates of bioactive synthetic amatoxins for targeting somatostatin receptor (sstr2) expressing cells. RSC Chem Biol 2022; 3:69-78. [PMID: 35128410 PMCID: PMC8729174 DOI: 10.1039/d1cb00036e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022] Open
Abstract
Targeted cancer therapy represents a paradigm-shifting approach that aims to deliver a toxic payload selectively to target-expressing cells thereby sparing normal tissues the off-target effects associated with traditional chemotherapeutics. Since most targeted constructs rely on standard microtubule inhibitors or DNA-reactive molecules as payloads, new toxins that inhibit other intracellular targets are needed to realize the full potential of targeted therapy. Among these new payloads, α-amanitin has gained attraction as a payload in targeted therapy. Here, we conjugate two synthetic amanitins at different sites to demonstrate their utility as payloads in peptide drug conjugates (PDCs). As an exemplary targeting agent, we chose octreotate, a well-studied somatostatin receptor (sstr2) peptide agonist for the conjugation to synthetic amatoxins via three tailor-built linkers. The linker chemistry permitted the evaluation of one non-cleavable and two cleavable self-immolative conjugates. The immolating linkers were chosen to take advantage of either the reducing potential of the intracellular environment or the high levels of lysosomal proteases in tumor cells to trigger toxin release. Cell-based assays on target-positive Ar42J cells revealed target-specific reduction in viability with up to 1000-fold enhancement in bioactivity compared to the untargeted amatoxins. Altogether, this preliminary study enabled the development of a highly modular synthetic platform for the construction of amanitin-based conjugates that can be readily extended to various targeting moieties.
Collapse
Affiliation(s)
- Alla Pryyma
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Kaveh Matinkhoo
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Yong Jia Bu
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Zhengxing Zhang
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - Francois Bénard
- Department of Molecular Oncology, BC Cancer Vancouver BC V5Z 1L3 Canada
| | - David M Perrin
- Department of Chemistry, University of British Columbia 2036 Main Mall Vancouver BC V6T 1Z1 Canada
| |
Collapse
|
5
|
Role of Gonadotropin-Releasing Hormone (GnRH) in Ovarian Cancer. Cells 2021; 10:cells10020437. [PMID: 33670761 PMCID: PMC7922220 DOI: 10.3390/cells10020437] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/28/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus–pituitary–gonadal (HPG) axis is the endocrine regulation system that controls the woman’s cycle. The gonadotropin-releasing hormone (GnRH) plays the central role. In addition to the gonadotrophic cells of the pituitary, GnRH receptors are expressed in other reproductive organs, such as the ovary and in tumors originating from the ovary. In ovarian cancer, GnRH is involved in the regulation of proliferation and metastasis. The effects on ovarian tumors can be indirect or direct. GnRH acts indirectly via the HPG axis and directly via GnRH receptors on the surface of ovarian cancer cells. In this systematic review, we will give an overview of the role of GnRH in ovarian cancer development, progression and therapy.
Collapse
|
6
|
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
|
7
|
Patel TK, Adhikari N, Amin SA, Biswas S, Jha T, Ghosh B. Small molecule drug conjugates (SMDCs): an emerging strategy for anticancer drug design and discovery. NEW J CHEM 2021. [DOI: 10.1039/d0nj04134c] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms of how SMDCs work. Small molecule drugs are conjugated with the targeted ligand using pH sensitive linkers which allow the drug molecule to get released at lower lysosomal pH. It helps to accumulate the chemotherapeutic agents to be localized in the tumor environment upon cleaving of the pH-labile bonds.
Collapse
Affiliation(s)
- Tarun Kumar Patel
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Nilanjan Adhikari
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Sk. Abdul Amin
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| | - Tarun Jha
- Natural Science Laboratory
- Division of Medicinal and Pharmaceutical Chemistry
- Department of Pharmaceutical Technology
- Jadavpur University
- Kolkata 700032
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy
- BITS-Pilani
- Hyderabad
- India
| |
Collapse
|
8
|
Wu HM, Chang HM, Leung PCK. Gonadotropin-releasing hormone analogs: Mechanisms of action and clinical applications in female reproduction. Front Neuroendocrinol 2021; 60:100876. [PMID: 33045257 DOI: 10.1016/j.yfrne.2020.100876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/23/2020] [Accepted: 10/04/2020] [Indexed: 12/15/2022]
Abstract
Extra-hypothalamic GnRH and extra-pituitary GnRH receptors exist in multiple human reproductive tissues, including the ovary, endometrium and myometrium. Recently, new analogs (agonists and antagonists) and modes of GnRH have been developed for clinical application during controlled ovarian hyperstimulation for assisted reproductive technology (ART). Additionally, the analogs and upstream regulators of GnRH suppress gonadotropin secretion and regulate the functions of the reproductive axis. GnRH signaling is primarily involved in the direct control of female reproduction. The cellular mechanisms and action of the GnRH/GnRH receptor system have been clinically applied for the treatment of reproductive disorders and have widely been introduced in ART. New GnRH analogs, such as long-acting GnRH analogs and oral nonpeptide GnRH antagonists, are being continuously developed for clinical application. The identification of the upstream regulators of GnRH, such as kisspeptin and neurokinin B, provides promising potential to develop these upstream regulator-related analogs to control the hypothalamus-pituitary-ovarian axis.
Collapse
Affiliation(s)
- Hsien-Ming Wu
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Medical Center, Chang Gung University School of Medicine, Taoyuan 333, Taiwan, ROC
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6H 3V5, Canada
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia V6H 3V5, Canada.
| |
Collapse
|
9
|
Mehrotra N, Kharbanda S, Singh H. Peptide-based combination nanoformulations for cancer therapy. Nanomedicine (Lond) 2020; 15:2201-2217. [PMID: 32914691 DOI: 10.2217/nnm-2020-0220] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Research in cancer therapy is moving towards the use of biomolecules in combination with conventional approaches for improved disease outcome. Among the biomolecules explored, peptides are strong contenders due to their small size, high specificity, low systemic toxicity and wide inter/intracellular targets. The use of nanoformulations for such combination approaches can lead to further improvement in efficacy by reducing off-target cytotoxicity, increasing circulation time, tumor penetration and accumulation. This review focuses on nanodelivery systems for peptide-based combinations with chemo, immuno, radiation and hormone therapy. It gives an overview of the latest therapeutic research being conducted using combination nanoformulations with anticancer peptides, cell penetrating/tumor targeting peptides, peptide nanocarriers, peptidomimetics, peptide-based hormones and peptide vaccines. The challenges hindering clinical translation are also discussed.
Collapse
Affiliation(s)
- Neha Mehrotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Surender Kharbanda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| |
Collapse
|
10
|
Obayemi JD, Salifu AA, Eluu SC, Uzonwanne VO, Jusu SM, Nwazojie CC, Onyekanne CE, Ojelabi O, Payne L, Moore CM, King JA, Soboyejo WO. LHRH-Conjugated Drugs as Targeted Therapeutic Agents for the Specific Targeting and Localized Treatment of Triple Negative Breast Cancer. Sci Rep 2020; 10:8212. [PMID: 32427904 PMCID: PMC7237454 DOI: 10.1038/s41598-020-64979-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 04/22/2020] [Indexed: 02/07/2023] Open
Abstract
Bulk chemotherapy and drug release strategies for cancer treatment have been associated with lack of specificity and high drug concentrations that often result in toxic side effects. This work presents the results of an experimental study of cancer drugs (prodigiosin or paclitaxel) conjugated to Luteinizing Hormone-Releasing Hormone (LHRH) for the specific targeting and treatment of triple negative breast cancer (TNBC). Injections of LHRH-conjugated drugs (LHRH-prodigiosin or LHRH-paclitaxel) into groups of 4-week-old athymic female nude mice (induced with subcutaneous triple negative xenograft breast tumors) were found to specifically target, eliminate or shrink tumors at early, mid and late stages without any apparent cytotoxicity, as revealed by in vivo toxicity and ex vivo histopathological tests. Our results show that overexpressed LHRH receptors serve as binding sites on the breast cancer cells/tumor and the LHRH-conjugated drugs inhibited the growth of breast cells/tumor in in vitro and in vivo experiments. The inhibitions are attributed to the respective adhesive interactions between LHRH molecular recognition units on the prodigiosin (PGS) and paclitaxel (PTX) drugs and overexpressed LHRH receptors on the breast cancer cells and tumors. The implications of the results are discussed for the development of ligand-conjugated drugs for the specific targeting and treatment of TNBC.
Collapse
Affiliation(s)
- J D Obayemi
- Department of Mechanical Engineering, Higgins Lab, 100 Institute Road, Worcester Polytechnic Institute (WPI), Worcester, MA, 01609, USA.,Department of Biomedical Engineering, Gateway Park Life Sciences Center, 60 Prescott Street, Worcester Polytechnic Institute (WPI), Worcester, MA, 01605, USA
| | - A A Salifu
- Department of Mechanical Engineering, Higgins Lab, 100 Institute Road, Worcester Polytechnic Institute (WPI), Worcester, MA, 01609, USA
| | - S C Eluu
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, 420110, Ifite Awka, Anambra State, Nigeria
| | - V O Uzonwanne
- Department of Mechanical Engineering, Higgins Lab, 100 Institute Road, Worcester Polytechnic Institute (WPI), Worcester, MA, 01609, USA
| | - S M Jusu
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - C C Nwazojie
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - C E Onyekanne
- Department of Material Science, African University of Science and Technology, Km 10 Airport Road, Abuja, Nigeria
| | - O Ojelabi
- RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA, 01605, USA
| | - L Payne
- Department of Psychiatry, Center for Comparative NeuroImaging, University of Massachusetts Medical School, 303 Belmont Street, Worcester, MA, 01604, USA
| | - C M Moore
- Department of Psychiatry, Center for Comparative NeuroImaging, University of Massachusetts Medical School, 303 Belmont Street, Worcester, MA, 01604, USA
| | - J A King
- Department of Psychiatry, Center for Comparative NeuroImaging, University of Massachusetts Medical School, 303 Belmont Street, Worcester, MA, 01604, USA.,Department of Biology & Biotechnology, Gateway Park Life Sciences Center, 60 Prescott Street (Gateway Park I), Worcester Polytechnic Institute (WPI), Worcester, MA, 01605, USA
| | - W O Soboyejo
- Department of Mechanical Engineering, Higgins Lab, 100 Institute Road, Worcester Polytechnic Institute (WPI), Worcester, MA, 01609, USA. .,Department of Biomedical Engineering, Gateway Park Life Sciences Center, 60 Prescott Street, Worcester Polytechnic Institute (WPI), Worcester, MA, 01605, USA.
| |
Collapse
|
11
|
Worm DJ, Hoppenz P, Els-Heindl S, Kellert M, Kuhnert R, Saretz S, Köbberling J, Riedl B, Hey-Hawkins E, Beck-Sickinger AG. Selective Neuropeptide Y Conjugates with Maximized Carborane Loading as Promising Boron Delivery Agents for Boron Neutron Capture Therapy. J Med Chem 2019; 63:2358-2371. [PMID: 31589041 DOI: 10.1021/acs.jmedchem.9b01136] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors like the human Y1 receptor (hY1R) are promising targets in cancer therapy due to their high overexpression on cancer cells and their ability to internalize together with the bound ligand. This mechanism was exploited to shuttle boron atoms into cancer cells for the application of boron neutron capture therapy (BNCT), a noninvasive approach to eliminate cancer cells. A maximized number of carboranes was introduced to the hY1R-preferring ligand [F7,P34]-NPY by solid phase peptide synthesis. Branched conjugates loaded with up to 80 boron atoms per peptide molecule exhibited a maintained receptor activation profile, and the selective uptake into hY1R-expressing cells was demonstrated by internalization studies. In order to ensure appropriate solubility in aqueous solution, we proved the need for eight hydroxyl groups per carborane. Thus, we suggest the utilization of bis-deoxygalactosyl-carborane building blocks in solid phase peptide synthesis to produce selective boron delivery agents for BNCT.
Collapse
Affiliation(s)
- Dennis J Worm
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Paul Hoppenz
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sylvia Els-Heindl
- Institute of Biochemistry, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Martin Kellert
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Robert Kuhnert
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Stefan Saretz
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | | | - Bernd Riedl
- Bayer AG, Aprather Weg 18A, 42113 Wuppertal, Germany
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | | |
Collapse
|
12
|
Peptide Conjugates with Small Molecules Designed to Enhance Efficacy and Safety. Molecules 2019; 24:molecules24101855. [PMID: 31091786 PMCID: PMC6572008 DOI: 10.3390/molecules24101855] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/17/2022] Open
Abstract
Peptides constitute molecular diversity with unique molecular mechanisms of action that are proven indispensable in the management of many human diseases, but of only a mere fraction relative to more traditional small molecule-based medicines. The integration of these two therapeutic modalities offers the potential to enhance and broaden pharmacology while minimizing dose-dependent toxicology. This review summarizes numerous advances in drug design, synthesis and development that provide direction for next-generation research endeavors in this field. Medicinal studies in this area have largely focused upon the application of peptides to selectively enhance small molecule cytotoxicity to more effectively treat multiple oncologic diseases. To a lesser and steadily emerging extent peptides are being therapeutically employed to complement and diversify the pharmacology of small molecule drugs in diseases other than just cancer. No matter the disease, the purpose of the molecular integration remains constant and it is to achieve superior therapeutic outcomes with diminished adverse effects. We review linker technology and conjugation chemistries that have enabled integrated and targeted pharmacology with controlled release. Finally, we offer our perspective on opportunities and obstacles in the field.
Collapse
|
13
|
Ma S, Pradeep S, Villar-Prados A, Wen Y, Bayraktar E, Mangala LS, Kim MS, Wu SY, Hu W, Rodriguez-Aguayo C, Leuschner C, Liang X, Ram PT, Schlacher K, Coleman RL, Sood AK. GnRH-R-Targeted Lytic Peptide Sensitizes BRCA Wild-type Ovarian Cancer to PARP Inhibition. Mol Cancer Ther 2019; 18:969-979. [PMID: 30926640 DOI: 10.1158/1535-7163.mct-18-0770] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/20/2018] [Accepted: 03/15/2019] [Indexed: 12/20/2022]
Abstract
EP-100 is a synthetic lytic peptide that specifically targets the gonadotropin-releasing hormone receptor on cancer cells. To extend the utility of EP-100, we aimed to identify effective combination therapies with EP-100 for ovarian cancer and explore potential mechanisms of this combination. A series of in vitro (MTT assay, immunoblot analysis, reverse-phase protein array, comet assay, and immunofluorescence staining) and in vivo experiments were carried out to determine the biological effects of EP-100 alone and in combination with standard-of-care drugs. EP-100 decreased the viability of ovarian cancer cells and reduced tumor growth in orthotopic mouse models. Of five standard drugs tested (cisplatin, paclitaxel, doxorubicin, topotecan, and olaparib), we found that the combination of EP-100 and olaparib was synergistic in ovarian cancer cell lines. Further experiments revealed that combined treatment of EP-100 and olaparib significantly increased the number of nuclear foci of phosphorylated histone H2AX. In addition, the extent of DNA damage was significantly increased after treatment with EP-100 and olaparib in comet assay. We performed reverse-phase protein array analyses and identified that the PI3K/AKT pathway was inhibited by EP-100, which we validated with in vitro experiments. In vivo experiment using the HeyA8 mouse model demonstrated that mice treated with EP-100 and olaparib had lower tumor weights (0.06 ± 0.13 g) than those treated with a vehicle (1.19 ± 1.09 g), EP-100 alone (0.62 ± 0.78 g), or olaparib alone (0.50 ± 0.63 g). Our findings indicate that combining EP-100 with olaparib is a promising therapeutic strategy for ovarian cancer.
Collapse
Affiliation(s)
- Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alejandro Villar-Prados
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,University of Puerto Rico, School of Medicine, Medical Sciences Campus, San Juan, Puerto Rico
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Seungwook Kim
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Y Wu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Rodriguez-Aguayo
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carola Leuschner
- VP of Research and Development, Esperance Pharmaceuticals, Houston, Texas
| | - Xiaoyan Liang
- Reproductive Medicine Research Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Prahlad T Ram
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Katharina Schlacher
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
14
|
Suo L, Chang X, Xu N, Ji H. The Anti-proliferative Activity of GnRH Through Downregulation of the Akt/ERK Pathways in Pancreatic Cancer. Front Endocrinol (Lausanne) 2019; 10:370. [PMID: 31263453 PMCID: PMC6590102 DOI: 10.3389/fendo.2019.00370] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/24/2019] [Indexed: 11/20/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) has been demonstrated to exert anti-proliferative functions on various tumor cells in endometrial, ovarian, bladder, or prostate cancer as a part of the autocrine system. In addition, the expression levels of GnRH and its receptor had been identified in breast cancer or non-reproductive cancers, such as glioblastoma and pancreatic cancer. Previous studies have reported abnormal GnRH expression in several malignant tumors, suggesting that GnRH and its receptor might be essential for tumourigenesis. In the present study, we attempted to clarify the mechanisms underlying GnRH function in cell proliferation in pancreatic cancer. Our results indicated that GnRH expression might be essential for the malignancy of pancreatic cancer. We then found that GnRH overexpression can induce cell apoptosis through activating the Bcl-2/Bax pathway and autophagy might be involved in the GnRH-mediated apoptosis in Panc1 cells. Further investigation showed that the inhibition of GnRH may promote tumor invasion and migration through upregulation of MMP2 expression in pancreatic cancer cells. Moreover, our results indicated that GnRH can regulate the Akt/ERK1/2 pathways to promote cell proliferation by inhibiting cell apoptosis in Panc1 cells. Therefore, our finding exhibited that the regulation of GnRH expression may be essential for tumourigenesis in pancreatic cancer, and might be a potential target for the treatment of the patients with pancreatic cancer.
Collapse
Affiliation(s)
- Linna Suo
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaocen Chang
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Na Xu
- Natural Sciences Department, LaGuardia Community College, City University of New York, New York City, NY, United States
| | - Hongmei Ji
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
- *Correspondence: Hongmei Ji
| |
Collapse
|
15
|
Oláh G, Dobos N, Vámosi G, Szabó Z, Sipos É, Fodor K, Harda K, Schally AV, Halmos G. Experimental therapy of doxorubicin resistant human uveal melanoma with targeted cytotoxic luteinizing hormone-releasing hormone analog (AN-152). Eur J Pharm Sci 2018; 123:371-376. [PMID: 30076951 DOI: 10.1016/j.ejps.2018.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Cytotoxic analogs of LHRH (luteinizing hormone-releasing hormone) can be successfully used for the treatment of hormone-dependent cancers such as prostatic, ovarian, endometrial, but our knowledge about their effect on hormone-independent cancers such as human uveal melanoma (UM) is limited. Previously, we have demonstrated that 46% of UM express full-length LHRH receptors. This finding has led us to further examine the mechanism of action of LHRH receptor based targeted therapies in this malignancy. AIMS In the present study we investigated the cellular uptake of doxorubicin (DOX) and cytotoxic LHRH analog AN-152 (AEZS-108, zoptarelin doxorubicin) on human UM cell lines (OCM3) and its DOX resistant form OCM3DOX320 by confocal laser scanning microscopy. The LHRH receptor expression was characterized by RT-PCR and immunocytochemistry. RESULTS We were able to establish a new, stable and DOX resistant human UM cell line OCM3DOX320. Our results demonstrated the expression of splice variants and isoforms of receptor for LHRH in OCM3 UM cell line and its doxorubicin resistant form OCM3DOX320. It has been revealed by MTT assay that AN-152 inhibited cell proliferation in a dose dependent manner in OCM3DOX320 cells. Furthermore, receptor-mediated uptake of AN-152 was demonstrated using confocal laser scanning microscopy in both cell line. CONCLUSIONS Our results suggest that the antiproliferative effect of AN-152 can be detected even if only LHRH receptor isoforms are expressed. Our study also demonstrates the LHRH receptor-mediated uptake of AN-152 in DOX resistant OCM3DOX320 cells. Our experiments provide new insights into a potential targeted therapy of UM and give further details about the accumulation of AN-152 in hormone-independent DOX-resistant cells expressing splice variants of the LHRH receptors.
Collapse
Affiliation(s)
- Gábor Oláh
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Dobos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - György Vámosi
- Department of Biophysics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Éva Sipos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klára Fodor
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Kristóf Harda
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA; Department of Pathology, University of Miami, Miami, FL, USA; Department of Medicine, Division of Hematology-Oncology, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Department of Medicine, Division of Endocrinology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA.
| |
Collapse
|
16
|
Sipos E, Dobos N, Rozsa D, Fodor K, Olah G, Szabo Z, Szekvolgyi L, Schally AV, Halmos G. Characterization of luteinizing hormone-releasing hormone receptor type I (LH-RH-I) as a potential molecular target in OCM-1 and OCM-3 human uveal melanoma cell lines. Onco Targets Ther 2018; 11:933-941. [PMID: 29503568 PMCID: PMC5826244 DOI: 10.2147/ott.s148174] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Introduction Uveal melanoma (UM) is the most common primary intraocular malignancy with very poor prognosis. Conventional chemotherapy only rarely prolongs the survival, therefore patients require novel treatment modalities. The discovery of specific receptors for hypothalamic hormones on cancer cells has led to the development of radiolabeled and cytotoxic hormone analogs. Materials and methods In the present study, our aim was to investigate the expression of mRNA for receptors of luteinizing hormone-releasing hormone type I (LH-RH-I) and LH-RH ligand in OCM-1 and OCM-3 human uveal melanoma cell lines. The presence and binding characteristics of LH-RH-I receptor protein was further studied by Western blot, immunocytochemistry and ligand competition assay. The expression of mRNA and protein for LH-RH-I receptors has been also studied using tumor samples originating from nude mice xenografted with OCM-1 or OCM-3 cells. Results The mRNA for LH-RH-I receptor has been detected in OCM-1 and OCM-3 cell lines and was found markedly higher in OCM-3 cells. The mRNA for LH-RH-I receptors was also observed in both UM xenograft models in vivo with higher levels in OCM-3. The presence of LH-RH-I receptor protein was found in both cell lines in vitro by immunocytochemistry and Western blot, and also in tumor tissue samples grown in nude mice by Western blot. Both human uveal melanoma models investigated showed specific high affinity receptors for LH-RH-I using ligand competition assay. The mRNA for LH-RH ligand has also been detected in OCM-1 and OCM-3 cell lines and cancer tissues. Conclusion The demonstration of the expression of LH-RH-I receptors in OCM-1 and OCM-3 human UM cell lines suggests that they could serve as potential molecular target for therapy. Our findings support the development of new therapeutic approaches based on cytotoxic LH-RH analogs or modern powerful antagonistic analogs of LH-RH targeting LH-RH-I receptors in UM.
Collapse
Affiliation(s)
- Eva Sipos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Dobos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - David Rozsa
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klara Fodor
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Gabor Olah
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Szabo
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Lorant Szekvolgyi
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Debrecen, Hungary.,Research Centre for Molecular Medicine; Department of Biochemistry and Molecular Biology, Debrecen, Hungary
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA.,Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Gabor Halmos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary.,Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA
| |
Collapse
|
17
|
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
|
18
|
Tambe P, Kumar P, Karpe YA, Paknikar KM, Gajbhiye V. Triptorelin Tethered Multifunctional PAMAM-Histidine-PEG Nanoconstructs Enable Specific Targeting and Efficient Gene Silencing in LHRH Overexpressing Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:35562-35573. [PMID: 28949503 DOI: 10.1021/acsami.7b11024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Cancer treatment using siRNA based therapies pose various limitations such as off-target effects and degradation due to lack of specific delivery in desired cells. The aim of the present study was to develop multifunctional targeted nanoconstructs, which can efficiently and precisely deliver siRNA and silence the desired gene of interest in various LHRH overexpressing cancer cells. Herein, we report the development of triblock, PAMAM-histidine-PEG dendritic nanoconstructs functionalized with triptorelin (an LHRH analog) for targeted siRNA delivery to LHRH overexpressing breast (MCF-7) and prostate (LNCaP) cancer cells. The nanoconstructs were characterized using 1H NMR and DLS and displayed a very low cationic charge to avoid off-target interactions. The developed nanoconstructs showed negligible cytotoxicity and hemolytic activity with efficient siRNA loading, excellent serum stability, and strongly protected siRNA from degradation. Further, confocal microscopy results confirmed extremely significant (p < 0.001) higher cellular uptake of cy5.5 conjugated targeted nanoparticles (NPs) in both cancer cell lines than nontargeted NPs. Also, targeted NPs specifically delivered cy3-tagged siRNA to MCF-7 cells. Co-localization studies in MCF-7 and LNCaP cells further established that targeted NPs traveled through the endolysosomal pathway and escaped endosomes within 6 h of incubation. Gene silencing studies in luciferase expressing MCF-7 and LNCaP cell lines demonstrated that the targeted NPs exhibited extremely significant (p < 0.001) silencing of luciferase gene. Additionally, receptor blockade studies further confirmed the specificity of targeted NPs and suggested that targeted NPs entered cancer cells via LHRH receptor mediated endocytosis, which was evident through insignificant gene silencing in receptor blocked cells. Thus, the results indicated that PAMAM-histidine-PEG-triptorelin could be a promising approach for siRNA delivery, gene silencing, and tumor therapy in all LHRH overexpressing cancer cells.
Collapse
Affiliation(s)
- Prajakta Tambe
- Nanobioscience, Agharkar Research Institute , Pune 411 004, India
- Savitribai Phule Pune University , Ganeshkhind, Pune 411 007, India
| | - Pramod Kumar
- Nanobioscience, Agharkar Research Institute , Pune 411 004, India
- Savitribai Phule Pune University , Ganeshkhind, Pune 411 007, India
| | - Yogesh A Karpe
- Nanobioscience, Agharkar Research Institute , Pune 411 004, India
- Savitribai Phule Pune University , Ganeshkhind, Pune 411 007, India
| | - Kishore M Paknikar
- Nanobioscience, Agharkar Research Institute , Pune 411 004, India
- Savitribai Phule Pune University , Ganeshkhind, Pune 411 007, India
| | - Virendra Gajbhiye
- Nanobioscience, Agharkar Research Institute , Pune 411 004, India
- Savitribai Phule Pune University , Ganeshkhind, Pune 411 007, India
| |
Collapse
|
19
|
Gründker C, Emons G. The Role of Gonadotropin-Releasing Hormone in Cancer Cell Proliferation and Metastasis. Front Endocrinol (Lausanne) 2017; 8:187. [PMID: 28824547 PMCID: PMC5543040 DOI: 10.3389/fendo.2017.00187] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022] Open
Abstract
In several human malignant tumors of the urogenital tract, including cancers of the endometrium, ovary, urinary bladder, and prostate, it has been possible to identify expression of gonadotropin-releasing hormone (GnRH) and its receptor as part of an autocrine system, which regulates cell proliferation. The expression of GnRH receptor has also been identified in breast cancers and non-reproductive cancers such as pancreatic cancers and glioblastoma. Various investigators have observed dose- and time-dependent growth inhibitory effects of GnRH agonists in cell lines derived from these cancers. GnRH antagonists have also shown marked growth inhibitory effects on most cancer cell lines. This indicates that in the GnRH system in cancer cells, there may not be a dichotomy between GnRH agonists and antagonists. The well-known signaling mechanisms of the GnRH receptor, which are present in pituitary gonadotrophs, are not involved in forwarding the antiproliferative effects of GnRH analogs in cancer cells. Instead, the GnRH receptor activates a phosphotyrosine phosphatase (PTP) and counteracts with the mitogenic signal transduction of growth factor receptors, which results in a reduction of cancer cell proliferation. The PTP activation, which is induced by GnRH, also inhibits G-protein-coupled estrogen receptor 1 (GPER), which is a membrane-bound receptor for estrogens. GPER plays an important role in breast cancers, which do not express the estrogen receptor α (ERα). In metastatic breast, ovarian, and endometrial cancer cells, GnRH reduces cell invasion in vitro, metastasis in vivo, and the increased expression of S100A4 and CYR61. All of these factors play important roles in epithelial-mesenchymal transition. This review will summarize the present state of knowledge about the GnRH receptor and its signaling in human cancers.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| |
Collapse
|
20
|
Cazzamalli S, Corso AD, Neri D. Linker stability influences the anti-tumor activity of acetazolamide-drug conjugates for the therapy of renal cell carcinoma. J Control Release 2016; 246:39-45. [PMID: 27890855 DOI: 10.1016/j.jconrel.2016.11.023] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/26/2016] [Accepted: 11/17/2016] [Indexed: 10/20/2022]
Abstract
Small molecule-drug conjugates (SMDCs) are increasingly being considered as an alternative to antibody-drug conjugates (ADCs) for the selective delivery of anticancer agents to the tumor site, sparing normal tissues. Carbonic anhydrase IX (CAIX) is a membrane-bound enzyme, which is over-expressed in the majority of renal cell carcinomas and which can be efficiently targeted in vivo, using charged derivatives of acetazolamide, a small heteroaromatic sulfonamide. Here, we show that SMDC products, obtained by the coupling of acetazolamide with monomethyl auristatin E (MMAE) using dipeptide linkers, display a potent anti-tumoral activity in mice bearing xenografted SKRC-52 renal cell carcinomas. A comparative evaluation of four dipeptides revealed that SMDCs featuring valine-citrulline and valine-alanine linkers exhibited greater serum stability and superior therapeutic activity, compared to the counterparts with valine-lysine or valine-arginine linkers. The most active products substantially inhibited tumor growth over a prolonged period of time, in a tumor model for which sunitinib and sorafenib do not display therapeutic activity. However, complete tumor eradication was not possible even after ten intravenous injection. Macroscopic near-infrared imaging procedures confirmed that ligands had not lost the ability to selectively localize at the tumor site at the end of therapy and that the neoplastic masses continued to express CAIX. The findings are of mechanistic and of therapeutic significance, since CAIX is a non-internalizing membrane-associated antigen, which can be considered for targeted drug delivery applications in kidney cancer patients.
Collapse
Affiliation(s)
- Samuele Cazzamalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| | - Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich (Switzerland)
| |
Collapse
|
21
|
Gilad Y, Firer M, Gellerman G. Recent Innovations in Peptide Based Targeted Drug Delivery to Cancer Cells. Biomedicines 2016; 4:E11. [PMID: 28536378 PMCID: PMC5344250 DOI: 10.3390/biomedicines4020011] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/16/2016] [Accepted: 05/23/2016] [Indexed: 12/21/2022] Open
Abstract
Targeted delivery of chemotherapeutics and diagnostic agents conjugated to carrier ligands has made significant progress in recent years, both in regards to the structural design of the conjugates and their biological effectiveness. The goal of targeting specific cell surface receptors through structural compatibility has encouraged the use of peptides as highly specific carriers as short peptides are usually non-antigenic, are structurally simple and synthetically diverse. Recent years have seen many developments in the field of peptide based drug conjugates (PDCs), particularly for cancer therapy, as their use aims to bypass off-target side-effects, reducing the morbidity common to conventional chemotherapy. However, no PDCs have as yet obtained regulatory approval. In this review, we describe the evolution of the peptide-based strategy for targeted delivery of chemotherapeutics and discuss recent innovations in the arena that should lead in the near future to their clinical application.
Collapse
Affiliation(s)
- Yosi Gilad
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Michael Firer
- Department of Chemical Engineering and Biotechnology, Ariel University, Ariel 40700, Israel.
| | - Gary Gellerman
- Department of Chemical Sciences, Ariel University, Ariel 40700, Israel.
| |
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
|
Redko B, Ragozin E, Andreii B, Helena T, Amnon A, Talia SZ, Mor OH, Genady K, Gary G. Synthesis, drug release, and biological evaluation of new anticancer drug-bioconjugates containing somatostatin backbone cyclic analog as a targeting moiety. Biopolymers 2015; 104:743-52. [DOI: 10.1002/bip.22694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/12/2015] [Accepted: 05/27/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Boris Redko
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
- Department of Chemistry; The Julius Spokojny Bioorganic Chemistry Laboratory; Bar Ilan University; Ramat Gan 52900 Israel
| | - Elena Ragozin
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| | - Bazylevich Andreii
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| | - Tuchinsky Helena
- Department of Molecular Biology; Ariel University; Ariel 40700 Israel
| | - Albeck Amnon
- Department of Chemistry; The Julius Spokojny Bioorganic Chemistry Laboratory; Bar Ilan University; Ramat Gan 52900 Israel
| | - Shekhter Zahavi Talia
- Department of Molecular Microbiology and Biotechnology; George S. Wise Faculty of Life Sciences; Tel-Aviv University; Tel-Aviv 69978 Israel
| | - Oron-Herman Mor
- The Advanced Technologies Center; Sheba Medical Center; Tel Hashomer 52621 Israel
| | - Kostenich Genady
- The Advanced Technologies Center; Sheba Medical Center; Tel Hashomer 52621 Israel
| | - Gellerman Gary
- Department of Biological Chemistry; Ariel University; Ariel 40700 Israel
| |
Collapse
|
24
|
Popovics P, Schally AV, Szalontay L, Block NL, Rick FG. Targeted cytotoxic analog of luteinizing hormone-releasing hormone (LHRH), AEZS-108 (AN-152), inhibits the growth of DU-145 human castration-resistant prostate cancer in vivo and in vitro through elevating p21 and ROS levels. Oncotarget 2015; 5:4567-78. [PMID: 24994120 PMCID: PMC4147346 DOI: 10.18632/oncotarget.2146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Management of castration-resistant prostate cancer (CRPC) is challenging due to lack of efficacious therapy. Luteinizing hormone-releasing hormone (LHRH) analogs appear to act directly on cells based on the LHRH receptors on human prostate adenocarcinoma cells. We explored anticancer activity of a cytotoxic analog of LHRH, AEZS-108, consisting of LHRH agonist linked to doxorubicin. Nude mice bearing DU-145 tumors were used to compare antitumor effects of AEZS-108 with its individual constituents or their unconjugated combination. The tumor growth inhibition of conjugate was greatest among treatment groups (90.5% inhibition vs. 41% by [D-Lys(6)]LHRH+DOX). The presence of LHRH receptors on DU-145 cells was confirmed by immunocytochemistry. In vitro, AEZS-108 significantly inhibited cell proliferation (61.2% inhibition) and elevated apoptosis rates (by 46%). By the detection of the inherent doxorubicin fluorescence, unconjugated doxorubicin was seen in the nucleus; the conjugate was perinuclear and at cell membrane. Autophagy, visualized by GFP-tagged p62 reporter, was increased by AEZS-108 (7.9-fold vs. 5.3-fold by DOX+[D-Lys(6)]LHRH. AEZS-108 more effectively increased reactive oxygen species (ROS, 2-fold vs. 1.4-fold by DOX+[D-Lys(6)]LHRH) and levels of the apoptotic regulator p21 in vivo and in vitro. We demonstrate robust inhibitory effects of the targeted cytotoxic LHRH analog, AEZS-108, on LHRHR positive castration-resistant prostate cancer cells.
Collapse
Affiliation(s)
- Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Cardiovascular Diseases, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL; Department of Medicine III, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL; Divisions of Hematology/Oncology, University of Miami, Miller School of Medicine, Miami, FL; Endocrinology University of Miami, Miller School of Medicine, Miami, FL
| | - Luca Szalontay
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL
| | - Norman L Block
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL; Divisions of Hematology/Oncology, University of Miami, Miller School of Medicine, Miami, FL
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| |
Collapse
|
25
|
Szabó I, Bősze S, Orbán E, Sipos É, Halmos G, Kovács M, Mező G. Comparative in vitro biological evaluation of daunorubicin containing GnRH-I and GnRH-II conjugates developed for tumor targeting. J Pept Sci 2015; 21:426-35. [PMID: 25908176 DOI: 10.1002/psc.2775] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/22/2022]
Abstract
Hormone based drug targeting is a promising tool for selective tumor therapy. In this study, synthesis and systematic comparative biological evaluation of novel drug containing analogs of gonadotropin-releasing hormone GnRH-I and GnRH-II is reported demonstrating their suitability for tumor targeting. The cytotoxic conjugates were prepared by the attachment of the chemotherapeutical agent daunorubicin (Dau) to GnRH analogs directly or through an enzyme-labile spacer with oxime linkage. All conjugates were found to be proteolytically stable under circumstances applied in biological assays. Both GnRH-I and GnRH-II were able to bind similarly to high-affinity GnRH-I receptors on human pituitary and human prostate cancer cells. The in vitro long-term cytotoxic effect of the conjugates was comparable with that of the free drug in human breast and colon cancer cell lines. Furthermore, a concentration-dependent cellular uptake profile was observed. The in vitro apoptotic effect of the compounds was evaluated by flow cytometry analysis using annexin-V. Our results show that both the GnRH-I and the GnRH-II based analogs might be applied for targeted tumor therapy.
Collapse
Affiliation(s)
- Ildikó Szabó
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös L. University, 1117, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
26
|
Böhme D, Beck-Sickinger AG. Controlling Toxicity of Peptide-Drug Conjugates by Different Chemical Linker Structures. ChemMedChem 2015; 10:804-14. [DOI: 10.1002/cmdc.201402514] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/02/2015] [Indexed: 01/17/2023]
|
27
|
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
|
28
|
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]
|
29
|
He S, Zhou Z, Li L, Yang Q, Yang Y, Guan S, Zhang J, Zhu X, Jin Y, Huang Y. Comparison of active and passive targeting of doxorubicin for somatostatin receptor 2 positive tumor models by octreotide-modified HPMA copolymer-doxorubicin conjugates. Drug Deliv 2014; 23:285-96. [PMID: 24865288 DOI: 10.3109/10717544.2014.911991] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Somatostatin receptor 2 (SSTR2), specifically over-expressed on many tumor cells, is a potential receipt for active targeting in cancer therapy. In the present study, octreotide (Oct), which had high affinity to SSTR2, was attached to N-(2-hydroxypropyl) methacrylamide (HPMA) polymeric system to enhance the antitumor efficiency of the anticancer drug doxorubicin (DOX). Two kinds of cell lines (HepG2 and A549), which overexpress SSTR2, were chosen as cell models. Compared with non-modified conjugates, Oct-modified conjugates exhibited superior cytotoxicity and intracellular uptake on both HepG2 and A549 cell lines. This might be due to the mechanism of receptor-mediated endocytosis. Subsequently, the in vivo biodistribution and antitumor activity evaluations showed that Oct modification significantly improved the tumor accumulation and antitumor efficacy of HPMA copolymer conjugates in SSTR2 over-expressed Kunming mice bearing H22 tumor xenografts. In summary, Oct-modified HPMA polymer-DOX conjugates might be a promising system for the treatment of SSTR2 over-expressed cancers.
Collapse
Affiliation(s)
- Shuang He
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Zhou Zhou
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Lian Li
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Qingqing Yang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Yang Yang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Shan Guan
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Jian Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Xi Zhu
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Yun Jin
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| | - Yuan Huang
- a Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy , Sichuan University , Chengdu , P.R. China
| |
Collapse
|
30
|
Dellinger TH, Monk BJ. Systemic therapy for recurrent endometrial cancer: a review of North American trials. Expert Rev Anticancer Ther 2014; 9:905-16. [DOI: 10.1586/era.09.54] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Targeting triple-negative breast cancer through the somatostatin receptor with the new cytotoxic somatostatin analogue AN-162 [AEZS-124]. Anticancer Drugs 2013; 24:150-7. [PMID: 23080077 DOI: 10.1097/cad.0b013e32835a7e29] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Previously, we have shown that the targeted cytotoxic somatostatin (sst) analogue AN-162 [AZSE-124] inhibits the growth of MDA-MB-231 human breast cancers xenografted into nude mice. In this study, we examined the trafficking of AN-162 into the cell, the expression of the somatostatin receptors (sstr) in specimens of human triple-negative breast cancers (TNBC), and the effect of AN-162 on HCC 1806 human TNBC xenografts. The expression of sstr in TNBC tumor samples was investigated by immunohistochemical staining. The expression of sstr in HCC 1806 was evaluated by reverse transcription PCR. Internalization studies with I-labeled AN-162 were carried out and the autofluorescence sign of doxorubicin moiety in the cell nucleus after incubation with AN-162 was measured using a fluorescence assay. The effects of AN-162 on the growth of HCC 1806 xenografted into nude mice were studied. A fluorescence microscopy cytotoxicity assay in vitro to detect cell death after treatment with AN-162 was also carried out. About 28% of TNBC tumor specimens showed a positive staining for sstr subtype 2a. HCC 1806 expresses all five subtypes of sstr. In the fluorescence cytotoxicity assay, dead HCC 1806 cells were found 24 h after incubation with AN-162. The growth of HCC 1806 tumors in nude mice was significantly inhibited by treatment with AN-162. AN-162 was internalized into the HCC 1806 cells and doxorubicin moiety was detected in the cell nuclei. This study is the first to show that the trafficking of the cytotoxic sst analogue AN-162 into the cell is mediated by sstr. Our work shows that the growth of xenografted HCC 1806 TNBCs can be effectively inhibited in vivo with AN-162. This investigation provides information on the mechanism of action and efficacy of this new targeted cytotoxic sst analogue and identifies in this relation the sstr as a favorable therapeutic target in TNBC.
Collapse
|
32
|
Abstract
Gastrin releasing peptide (GRP) is a regulatory peptide that acts through its receptor (GRPR) to regulate physiological functions in various organs. GRPR is overexpressed in neoplastic cells of most prostate cancers and some renal cell cancers and in the tumoral vessels of urinary tract cancers. Thus, targeting these tumours with specifically designed GRP analogues has potential clinical application. Potent and specific radioactive, cytotoxic or nonradioactive GRP analogues have been designed and tested in various animal tumour models with the aim of receptor targeting for tumour diagnosis or therapy. All three categories of compound were found suitable for tumour targeting in animal models. The cytotoxic and nonradioactive GRP analogues have not yet shown convincing tumour-reducing effects in human trials; however, the first clinical studies of radioactive GRP analogues--both agonists and antagonists--suggest promising opportunities for both diagnostic tumour imaging and radiotherapy of prostate and other GRPR-expressing cancers.
Collapse
|
33
|
Engel J, Emons G, Pinski J, Schally AV. AEZS-108 : a targeted cytotoxic analog of LHRH for the treatment of cancers positive for LHRH receptors. Expert Opin Investig Drugs 2012; 21:891-9. [PMID: 22577891 DOI: 10.1517/13543784.2012.685128] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Receptors for the luteinizing hormone-releasing hormone [LHRH, also known as gonadotropin-releasing hormone (GnRH)] can be regarded as an ideal target for a personalized medicine approach in cancer therapy. LHRH receptors are expressed in about 80% of human endometrial and ovarian cancers, 86% of prostate cancer, and about 50% of breast cancers including triple-negative breast cancer, as well as bladder, colorectal, and pancreatic cancers, sarcomas, lymphomas, melanomas, and renal cell carcinomas. Apart from the pituitary and reproductive organs, other organs and hematopoietic stem cells express LHRH receptors. Thus, a targeted cytotoxic LHRH analog such as AEZS-108 (formerly known as AN-152), in which doxorubin is linked to the LHRH agonist [D-Lys(6)]LHRH, appears to be a suitable drug for targeted chemotherapy of cancers expressing receptors for LHRH, which would be more efficacious and less toxic than standard systemic chemotherapy. AREAS COVERED This review discusses the development of AEZS-108, its targeting mechanism, preclinical studies, and clinical trials in patients with endometrial, ovarian, prostatic, and bladder cancers. We emphasize its development as a personalized medicine approach. The studies reviewed demonstrate the effects of the cytotoxic LHRH analog, AEZS-108, mediated by LHRH receptors, in in vivo models of LHRH-receptor-positive human endometrial, ovarian, breast, prostatic, colorectal, pancreatic, and bladder cancers xenografted into nude mice. Intravenous administration of AEZS 108 inhibits the growth of LHRH-receptor-positive tumors better than equimolar doses of the cytotoxic agent doxorubicin and is far less toxic. AEZS 108 has no antitumor activity in cancers negative to LHRH receptor. This strongly supports the concept of targeting cytotoxic chemotherapy to tumor cells expressing LHRH receptors. Early clinical trials have demonstrated the efficacy of AEZS-108. A Phase I trial assessed the maximum tolerated dose and pharmacokinetics and pharmacodynamics of AEZS-108 given once every 3 weeks in patients with gynecological cancers. Two Phase II studies in heavily pretreated ovarian and recurrent endometrial cancers showed good clinical activity after a maximum of six courses of AEZS-108 as a single agent. Ongoing clinical studies with AEZS-108 in men with castration-resistant prostate cancer and patients with chemotherapy refractory bladder cancer had shown early signs of clinical efficacy. Side effects are moderate and easily manageable. In particular, no pituitary or cardiac toxicity is observed. EXPERT OPINION AEZS-108 is a cytotoxic analog designed for receptor-mediated targeted chemotherapy and consists of an LHRH carrier linked to doxorubicin. Preclinical studies demonstrate that the uptake of AEZS-108 is achieved by receptor-mediated endocytosis. Results of Phase I and II clinical trials in patients with gynecological cancers demonstrated anticancer activity without cardiotoxicity even in highly pretreated patients. Phase I/II studies in castration-resistant prostate cancer and chemotherapy refractory bladder cancer are in progress. Targeted chemotherapy with a cytotoxic analog of LHRH, such as AEZS-108, is therefore being considered for Phase III studies in advanced endometrial cancers positive for LHRH receptor. LHRH receptors are also present in human colon cancers, melanomas, lymphomas, and sarcomas, and treatment of these cancers with AEZS-108 should also be undertaken. Before such treatment with AEZS-108 is begun, the status of tumoral LHRH receptors of patients must be determined.
Collapse
Affiliation(s)
- Joerg Engel
- Medical University of Regensburg, Department of Obstetrics and Gynecology, Landshuter Strasse 65, 93059 Regensburg, Germany
| | | | | | | |
Collapse
|
34
|
Struthers RS. Gonadotropin-Releasing Hormone Targeting for Gonadotroph Ablation: An Approach to Non-surgical Sterilization. Reprod Domest Anim 2012; 47 Suppl 4:233-8. [DOI: 10.1111/j.1439-0531.2012.02081.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
35
|
|
36
|
Engel JB, Schally AV, Buchholz S, Seitz S, Emons G, Ortmann O. Targeted chemotherapy of endometrial, ovarian and breast cancers with cytotoxic analogs of luteinizing hormone-releasing hormone (LHRH). Arch Gynecol Obstet 2012; 286:437-42. [PMID: 22555802 DOI: 10.1007/s00404-012-2335-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 04/12/2012] [Indexed: 10/28/2022]
Abstract
Receptors luteinizing hormone-releasing hormone (LHRH) are expressed in about 80 % of human endometrial and ovarian cancers and account for more than 50 % of breast cancers including triple negative breast cancers. Apart from the pituitary and reproductive organs, no other organs or hematopoietic stem cells express LHRH (GnRH) receptors. Thus, these receptors can be regarded as an ideal target for a personalized medicine approach in cancer therapy. AEZS-108 (formerly known as AN-152) in which doxorubin is linked to the LHRH agonist [D: -Lys(6)]LHRH, appears to be the most advanced compound in late stage clinical development. Results of phase I and phase II clinical trials in patients with gynecological cancers demonstrated anticancer activity without any cardiotoxicity even in highly pretreated patients. AEZS-108 is therefore being considered for phase II trials in triple negative breast cancers and phase III studies in advanced endometrial cancers positive for LHRH-receptor. EP-100 is a membrane-disrupting peptide targeted to LHRH receptors, which is undergoing early clinical studies in ovarian cancer patients.
Collapse
Affiliation(s)
- J B Engel
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93057 Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
BACKGROUND Cancer is responsible for millions of immature deaths every year and is an economical burden on developing countries. One of the major challenges in the present era is to design drugs that can specifically target tumor cells not normal cells. In this context, tumor homing peptides have drawn much attention. These peptides are playing a vital role in delivering drugs in tumor tissues with high specificity. In order to provide service to scientific community, we have developed a database of tumor homing peptides called TumorHoPe. DESCRIPTION TumorHoPe is a manually curated database of experimentally validated tumor homing peptides that specifically recognize tumor cells and tumor associated microenvironment, i.e., angiogenesis. These peptides were collected and compiled from published papers, patents and databases. Current release of TumorHoPe contains 744 peptides. Each entry provides comprehensive information of a peptide that includes its sequence, target tumor, target cell, techniques of identification, peptide receptor, etc. In addition, we have derived various types of information from these peptide sequences that include secondary/tertiary structure, amino acid composition, and physicochemical properties of peptides. Peptides in this database have been found to target different types of tumors that include breast, lung, prostate, melanoma, colon, etc. These peptides have some common motifs including RGD (Arg-Gly-Asp) and NGR (Asn-Gly-Arg) motifs, which specifically recognize tumor angiogenic markers. TumorHoPe has been integrated with many web-based tools like simple/complex search, database browsing and peptide mapping. These tools allow a user to search tumor homing peptides based on their amino acid composition, charge, polarity, hydrophobicity, etc. CONCLUSION TumorHoPe is a unique database of its kind, which provides comprehensive information about experimentally validated tumor homing peptides and their target cells. This database will be very useful in designing peptide-based drugs and drug-delivery system. It is freely available at http://crdd.osdd.net/raghava/tumorhope/.
Collapse
|
38
|
Sun L, Luo J, Mackey LV, Morris LM, Franko-Tobin LG, LePage KT, Coy DH. Investigation of cancer cell lines for peptide receptor-targeted drug development. J Drug Target 2012; 19:719-30. [PMID: 21830941 DOI: 10.3109/1061186x.2011.558089] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Many tumors highly express specific populations of G-protein-coupled receptors (GPCRs) that could be utilized for receptor-targeted therapy. We confirmed significant quantities of mRNAs specific for certain somatostatin (SST), vasoactive intestinal peptide (VIP), and bombesin (BN) receptors in various commercially available tumor cell lines. Very few of the tumor cell lines examined displayed the high receptor-binding affinity despite exhibiting the expression of appropriate mRNAs and proteins of the cognate receptors. However, binding assays establish that some tumor cell lines, such as pancreatic cancer CFPAC-1, prostate cancer DU-145, and pancreatic carcinoid BON, demonstrate high BN receptor binding. BON cells also demonstrate high somatostatin receptor (SSTR) affinity binding. We also found that tumor cell lines, such as BON and host cells expressing SST receptor subtypes 1 or 2 (CHO-R1 or CHO-R2), underwent a decrease in cell surface receptor density in multiple passages. BON and CHO-R2 cells also rapidly internalize a significant proportion of cell surface ligand-receptor complexes. The tumor cells CFPAC-1, DU-145, and BON with high receptor binding could be useful for peptide drug studies. BON cells were further applied to test SST/BN analogs and cytotoxic conjugates. Furthermore, the in vivo antitumor assay showed that the cytotoxic conjugate CPT-SST targeting all SSTR subtypes displayed a potent tumor-suppressive ability to BON tumors expressing multiple SSTR subtypes.
Collapse
Affiliation(s)
- Lichun Sun
- Department of Medicine, Peptide Research Laboratories, Tulane Health Sciences Center , New Orleans, LA 70112-2699, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Schlage P, Mező G, Orbán E, Bősze S, Manea M. Anthracycline-GnRH derivative bioconjugates with different linkages: Synthesis, in vitro drug release and cytostatic effect. J Control Release 2011; 156:170-8. [DOI: 10.1016/j.jconrel.2011.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/30/2011] [Accepted: 08/05/2011] [Indexed: 01/13/2023]
|
40
|
Jabre ND, Korostelova N, Kodanko JJ. A highly divergent approach for synthesis of metal-binding peptide libraries. J Org Chem 2011; 76:2273-6. [PMID: 21381716 DOI: 10.1021/jo102371q] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A highly divergent approach for the synthesis of metal-binding peptides is described. This approach builds pyridine-based chelators in a stepwise fashion on resin and provides rapid access to a diverse array of metal-binding peptides in minimal synthetic steps.
Collapse
Affiliation(s)
- Nitinkumar D Jabre
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
41
|
Dose escalation and pharmacokinetic study of AEZS-108 (AN-152), an LHRH agonist linked to doxorubicin, in women with LHRH receptor-positive tumors. Gynecol Oncol 2010; 119:457-61. [DOI: 10.1016/j.ygyno.2010.08.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/02/2010] [Accepted: 08/04/2010] [Indexed: 11/19/2022]
|
42
|
Zhang J, Jin W, Wang X, Wang J, Zhang X, Zhang Q. A novel octreotide modified lipid vesicle improved the anticancer efficacy of doxorubicin in somatostatin receptor 2 positive tumor models. Mol Pharm 2010; 7:1159-68. [PMID: 20524673 DOI: 10.1021/mp1000235] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Octreotide (Oct) is a potential ligand due to its high affinity to somatostatin receptors (SSTRs), especially subtype 2 (SSTR2), as many tumor cells specifically overexpress SSTR2. In this study, we conjugated Oct to the PEG end of DSPE-PEG and prepared a novel doxorubicin (DOX)-loaded and Oct-modified sterically stabilized liposomes (Oct-SSL-DOX), in order to facilitate intracellular delivery of chemotherapeutic agent to the related tumor cells through active targeting and finally improve its antitumor activity. Three cells were proved to be different in expression level of SSTR2 and were used as model or control. It was demonstrated by fluorescence spectrophotometry, confocal laser scanning microscopy and flow cytometry that active sterically stabilized liposomes (SSL) increased intracellular delivery of DOX in SSTR2-positive cells, through a mechanism of receptor-mediated endocytosis. Compared to SSL, Oct modification on SSL exhibited little effect on the physicochemical properties of SSL. However, it reduced the circulation time of loaded-DOX to some extent in rats, increased cytotoxicity in SSTR2-positive tumor cells, enhanced drug accumulation in tumor tissue and improved anticancer efficacy in SSTR2-overexpressing tumor model. The correlation was found among intracellular uptake, cytotoxicity, drug distribution in tumor and pharmacodynamics of Oct-SSL-DOX, but not the pharmacokinetics based on plasma drug concentration. In summary, octreotide-modified SSL might be a promising system for the treatment of SSTR2-overexpressing cancers.
Collapse
Affiliation(s)
- Junlin Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, P. R. China
| | | | | | | | | | | |
Collapse
|
43
|
Sun L, Morris LM, Luo J, Mackey LV, Leslie JS, Franko-Tobin LG, Fuselier JA, LePage KT, Coy DH. Application of human pancreatic carcinoid BON cells for receptor-targeted drug development. J Drug Target 2010; 19:666-74. [PMID: 21083509 DOI: 10.3109/1061186x.2010.531728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In our previous study, we found that several tumor cell lines displayed high receptor-specific binding affinity, one of which, the human pancreatic carcinoid BON cell line, demonstrates high affinity binding of the bombesin (BN) and somatostatin (SST) receptor-specific ligands. In the present study, BON cells, as a representative model, were further applied to evaluate various peptide analogs and cytotoxic receptor-targeted peptide conjugates. We observed quick ligand-receptor internalization in BON cells as well as high binding affinity. Furthermore, BON cells have high expression of multidrug resistance-associated genes (MDR1) and show camptothecin (CPT) resistance. Various receptor-specific cytotoxic conjugates were synthesized and evaluated in the BON cell model via in vitro and in vivo studies. We found that all the tested conjugates displayed potent antitumor ability in xenografts. Especially, the CPT conjugates, CPT-SST, and CPT-BN, are most likely to increase sensitivity to CPT-resistant BON cells. Our findings suggest that appropriately defined tumor cell lines may provide physiologically relevant cell-based evaluations of novel peptide analogs and receptor-targeted chemotherapeutics.
Collapse
Affiliation(s)
- Lichun Sun
- Department of Medicine, Peptide Research Laboratories, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ming X, Alam MR, Fisher M, Yan Y, Chen X, Juliano RL. Intracellular delivery of an antisense oligonucleotide via endocytosis of a G protein-coupled receptor. Nucleic Acids Res 2010; 38:6567-76. [PMID: 20551131 PMCID: PMC2965246 DOI: 10.1093/nar/gkq534] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gastrin-releasing peptide receptor (GRPR), a member of the G protein-coupled receptor superfamily, has been utilized for receptor-mediated targeting of imaging and therapeutic agents; here we extend its use to oligonucleotide delivery. A splice-shifting antisense oligonucleotide was conjugated to a bombesin (BBN) peptide, and its intracellular delivery was tested in GRPR expressing PC3 cells stably transfected with a luciferase gene interrupted by an abnormally spliced intron. The BBN-conjugate produced significantly higher luciferase expression compared to unmodified oligonucleotide, and this increase was reversed by excess BBN peptide. Kinetic studies revealed a combination of saturable, receptor-mediated endocytosis and non-saturable pinocytosis for uptake of the conjugate. The Km value for saturable uptake was similar to the EC50 value for the pharmacological response, indicating that receptor-mediated endocytosis was a primary contributor to the response. Use of pharmacological and molecular inhibitors of endocytosis showed that the conjugate utilized a clathrin-, actin- and dynamin-dependent pathway to enter PC3 cells. The BBN-conjugate partially localized in endomembrane vesicles that were associated with Rab7 or Rab9, demonstrating that it was transported to late endosomes and the trans-golgi network. These observations suggest that the BBN-oligonucleotide conjugate enters cells via a process of GRPR mediated endocytosis followed by trafficking to deep endomembrane compartments.
Collapse
Affiliation(s)
- Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
45
|
Endokrine Therapie der Zukunft. GYNAKOLOGISCHE ENDOKRINOLOGIE 2010. [DOI: 10.1007/s10304-009-0327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Martino MCD, Hofland LJ, Lamberts SW. Somatostatin and Somatostatin Receptors: from Basic Concepts to Clinical Applications. PROGRESS IN BRAIN RESEARCH 2010; 182:255-80. [DOI: 10.1016/s0079-6123(10)82011-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Preclinical evaluation of properties of a new targeted cytotoxic somatostatin analog, AN-162 (AEZS-124), and its effects on tumor growth inhibition. Anticancer Drugs 2009; 20:553-8. [PMID: 19491659 DOI: 10.1097/cad.0b013e32832d190b] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In view of findings that various tumors express receptors for somatostatin, a new targeted cytotoxic analog of somatostatin, AN-162 (AEZS-124), consisting of doxorubicin linked through glutaric acid to the somatostatin octapeptide RC-121 was developed in our laboratory. We studied the toxicity in vivo and the effect of AN-162 on growth of the MDA-MB-231 estrogen-independent human breast cancer cell line xenografted into nude mice. AN-162 induced significant tumor growth inhibition compared with the control and the group treated with doxorubicin in equimolar doses. We also evaluated the stability of AN-162 in various sera in vitro, as this conjugate is susceptible to hydrolysis by serum carboxylesterase enzymes in the circulation. This study shows for the first time that AN-162 is a safe and effective compound for the treatment of experimental breast cancer. Our findings support the concept of targeted chemotherapy based on cytotoxic peptide analog AN-162 for the treatment of breast cancers and other cancers expressing somatostatin receptors.
Collapse
|
48
|
Current World Literature. Curr Opin Obstet Gynecol 2009; 21:101-9. [DOI: 10.1097/gco.0b013e3283240745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
49
|
Gonzalez N, Moody TW, Igarashi H, Ito T, Jensen RT. Bombesin-related peptides and their receptors: recent advances in their role in physiology and disease states. Curr Opin Endocrinol Diabetes Obes 2008; 15:58-64. [PMID: 18185064 PMCID: PMC2631407 DOI: 10.1097/med.0b013e3282f3709b] [Citation(s) in RCA: 159] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Mammalian bombesin-related peptides, gastrin-releasing peptide and neuromedin B actions are mediated by two receptors (BB1-receptor, BB2-receptor), which are closely related to the orphan receptor BRS-3 (BB3-receptor). The purpose of this review is to highlight advances in the understanding of these peptides in physiology/disease states. RECENT FINDINGS Pharmacologic/receptor-knockout studies show involvement of these receptors in a number of new processes/diseases. Neuromedin B/BB1-receptor is an important physiological regulator of pituitary-thyroid function; in mediating behavior, especially feas/anxiety; in mediating satiety through different cascades than gastrin-releasing peptide/BB2 receptors and for its autocrine tumor-growth effects. Gastrin-releasing peptide/BB2-receptor plays important roles in mediating signals for pruritus, lung development/injury, small intestinal mucosal defense, and central nervous system processes such as learning/memory. The signaling mechanisms of its potent growth effects are being elucidated and their possible therapeutic targets identified. BB3-receptor knockout mice provided insights for their obesity/glucose intolerance and demonstrated that this receptor may be important in the lung response to injury, tumor growth and gastrointestinal motility. Each receptor is frequently overexpressed in human tumors and has potent growth effects. This effect is being explored to develop new antitumor treatments, such as bombesin-receptor ligands conjugated to cytotoxic agents. SUMMARY This receptor family is involved in an increasing number of central nervous system/peripheral processes physiologically and in disease states, and increased understanding of its role may lead to novel treatments.
Collapse
Affiliation(s)
- Nieves Gonzalez
- Digestive Diseases Branch, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
50
|
Bibliography. Current world literature. Growth and development. Curr Opin Endocrinol Diabetes Obes 2008; 15:79-101. [PMID: 18185067 DOI: 10.1097/med.0b013e3282f4f084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|