1
|
Solidoro R, Centonze A, Miciaccia M, Baldelli OM, Armenise D, Ferorelli S, Perrone MG, Scilimati A. Fluorescent imaging probes for in vivo ovarian cancer targeted detection and surgery. Med Res Rev 2024; 44:1800-1866. [PMID: 38367227 DOI: 10.1002/med.22027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/19/2024]
Abstract
Ovarian cancer is the most lethal gynecological cancer, with a survival rate of approximately 40% at five years from the diagno. The first-line treatment consists of cytoreductive surgery combined with chemotherapy (platinum- and taxane-based drugs). To date, the main prognostic factor is related to the complete surgical resection of tumor lesions, including occult micrometastases. The presence of minimal residual diseases not detected by visual inspection and palpation during surgery significantly increases the risk of disease relapse. Intraoperative fluorescence imaging systems have the potential to improve surgical outcomes. Fluorescent tracers administered to the patient may support surgeons for better real-time visualization of tumor lesions during cytoreductive procedures. In the last decade, consistent with the discovery of an increasing number of ovarian cancer-specific targets, a wide range of fluorescent agents were identified to be employed for intraoperatively detecting ovarian cancer. Here, we present a collection of fluorescent probes designed and developed for fluorescence-guided ovarian cancer surgery. Original articles published between 2011 and November 2022 focusing on fluorescent probes, currently under preclinical and clinical investigation, were searched in PubMed. The keywords used were targeted detection, ovarian cancer, fluorescent probe, near-infrared fluorescence, fluorescence-guided surgery, and intraoperative imaging. All identified papers were English-language full-text papers, and probes were classified based on the location of the biological target: intracellular, membrane, and extracellular.
Collapse
Affiliation(s)
- Roberta Solidoro
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Antonella Centonze
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Morena Miciaccia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Olga Maria Baldelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Domenico Armenise
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | - Savina Ferorelli
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| | | | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Bari, Italy
| |
Collapse
|
2
|
Cossu J, Thoreau F, Boturyn D. Multimeric RGD-Based Strategies for Selective Drug Delivery to Tumor Tissues. Pharmaceutics 2023; 15:pharmaceutics15020525. [PMID: 36839846 PMCID: PMC9961187 DOI: 10.3390/pharmaceutics15020525] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
RGD peptides have received a lot of attention over the two last decades, in particular to improve tumor therapy through the targeting of the αVβ3 integrin receptor. This review focuses on the molecular design of multimeric RGD compounds, as well as the design of suitable linkers for drug delivery. Many examples of RGD-drug conjugates have been developed, and we show the importance of RGD constructs to enhance binding affinity to tumor cells, as well as their drug uptake. Further, we also highlight the use of RGD peptides as theranostic systems, promising tools offering dual modality, such as tumor diagnosis and therapy. In conclusion, we address the challenging issues, as well as ongoing and future development, in comparison with large molecules, such as monoclonal antibodies.
Collapse
Affiliation(s)
- Jordan Cossu
- University Grenoble Alpes, CNRS, DCM UMR 5250, F-38000 Grenoble, France
| | - Fabien Thoreau
- University Poitiers, Inst Chim Milieux & Mat Poitiers IC2MP, UMR CNRS 7285, F-86073 Poitiers, France
| | - Didier Boturyn
- University Grenoble Alpes, CNRS, DCM UMR 5250, F-38000 Grenoble, France
- Correspondence:
| |
Collapse
|
3
|
Samec T, Boulos J, Gilmore S, Hazelton A, Alexander-Bryant A. Peptide-based delivery of therapeutics in cancer treatment. Mater Today Bio 2022; 14:100248. [PMID: 35434595 PMCID: PMC9010702 DOI: 10.1016/j.mtbio.2022.100248] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/27/2022] [Indexed: 11/09/2022] Open
Abstract
Current delivery strategies for cancer therapeutics commonly cause significant systemic side effects due to required high doses of therapeutic, inefficient cellular uptake of drug, and poor cell selectivity. Peptide-based delivery systems have shown the ability to alleviate these issues and can significantly enhance therapeutic loading, delivery, and cancer targetability. Peptide systems can be tailor-made for specific cancer applications. This review describes three peptide classes, targeting, cell penetrating, and fusogenic peptides, as stand-alone nanoparticle systems, conjugations to nanoparticle systems, or as the therapeutic modality. Peptide nanoparticle design, characteristics, and applications are discussed as well as peptide applications in the clinical space.
Collapse
Affiliation(s)
- Timothy Samec
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Jessica Boulos
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Serena Gilmore
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Anthony Hazelton
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| | - Angela Alexander-Bryant
- Nanobiotechnology Laboratory, Clemson University, Department of Bioengineering, Clemson, SC, USA
| |
Collapse
|
4
|
Wang L, Chen H, Wang F, Zhang X. The development of peptide-drug conjugates (PDCs) strategies for paclitaxel. Expert Opin Drug Deliv 2022; 19:147-161. [PMID: 35130795 DOI: 10.1080/17425247.2022.2039621] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Paclitaxel is a powerful and effective anti-tumor drug with wide clinical application. However, there are still some limitations, including poor water solubility, low specificity, and susceptibility to drug resistance. The peptide-drug conjugates (PDCs) represent a rising class of therapeutic drugs, which combines small-molecule chemotherapeutic drugs with highly flexible peptides through a cleavable or non-cleavable linker. When this strategy is applied, the therapeutic effects of paclitaxel can be improved. AREAS COVERED In this review, we discuss the application of the PDCs strategy in paclitaxel, including two parts: the tumor targeting peptide-paclitaxel conjugates and the cell penetrating peptide-paclitaxel conjugates. EXPERT OPINION Combining drugs with multifunctional peptides covalently is an effective strategy for delivering paclitaxel to tumors. Depending on different functional peptides, conjugates can increase the water solubility of paclitaxel, tumor permeability of paclitaxel, the accumulation of paclitaxel in tumor tissues, and enhance the antitumor effect of paclitaxel. In addition, due to the change of cell entry mechanism, partial conjugates can restore the therapeutic activity of paclitaxel against resistant tumors. Notably, in order to better translate into the clinical field in the future, more research should be conducted to ensure the safety and effectiveness of peptide-paclitaxel conjugates.
Collapse
Affiliation(s)
- Longkun Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, People's Republic of China
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan 250012, People's Republic of China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, People's Republic of China
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, People's Republic of China
| |
Collapse
|
5
|
Li SS, Zhang CM, Wu JD, Liu C, Liu ZP. A branched small molecule-drug conjugate nanomedicine strategy for the targeted HCC chemotherapy. Eur J Med Chem 2022; 228:114037. [PMID: 34883290 DOI: 10.1016/j.ejmech.2021.114037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Off-target toxicity is one of the main challenges faced by anticancer chemotherapeutics. For tumor targeted and precision chemotherapy, we take the advantages of the ligand directed tumor active targeting of small molecule drug conjugates (SMDCs) and the passive tumor targeting of nanoparticles via the enhanced penetration and retention (EPR) effects, put forward a branched small molecule drug conjugate (BSMDC) nanomedicine design concept. In a proof of concept, we used pentaerythritol as the branched moiety, galactosamine (GalN) as the hepatocellular carcinoma (HCC) directing ligands, PTX as a payload, and a stearoyl moiety as the amphiphilic property adjusting group, designed and synthesized BSMDC 1 and prepared its NPs. In cellular level, the BSMDC 1 NPs targeted asialoglycoprotein receptor (ASGPR)-overexpressing HepG2 cells, were effectively taken up in the cells and released in tumor microenvironments, inhibited the HepG2 cell proliferation, arrested HepG2 cell in G2/M phase and induced tumor cell apoptosis. In HepG2 xenograft nude mice, the BSMDC 1 NPs were high specific to target the tumor and demonstrated a higher antitumor efficiency than BSMDC 1, having no apparent influences on mice body weights and major organs, supporting our BSMDC nanomedicine design concept. Therefore, this new strategy may find applications for cancer targeted and precision chemotherapy.
Collapse
Affiliation(s)
- Sha-Sha Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Cheng-Mei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, PR China.
| |
Collapse
|
6
|
Yu L, Yao L, Yang K, Fei W, Chen Q, Qin L, Liu S, Zou Z, Qin B. Study on the Preparation and Properties of Highly Stable Micelles Sealed by Hydrogen Bonds. POLYMER SCIENCE SERIES A 2021. [DOI: 10.1134/s0965545x21350169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
7
|
Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol Res 2020; 162:105231. [PMID: 33027717 DOI: 10.1016/j.phrs.2020.105231] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/23/2020] [Accepted: 09/30/2020] [Indexed: 01/10/2023]
Abstract
Cancer is the second leading cause of death in the world and its treatment is extremely challenging, mainly due to its complexity. Cell-Penetrating Peptides (CPPs) are peptides that can transport into the cell a wide variety of biologically active conjugates (or cargoes), and are, therefore, promising in the treatment and in the diagnosis of several types of cancer. Some notable examples are TAT and Penetratin, capable of penetrating the central nervous system (CNS) and, therefore, acting in cancers of this system, such as Glioblastoma Multiforme (GBM). These above-mentioned peptides, conjugated with traditional chemotherapeutic such as Doxorubicin (DOX) and Paclitaxel (PTX), have also been shown to induce apoptosis of breast and liver cancer cells, as well as in lung cancer cells, respectively. In other cancers, such as esophageal cancer, the attachment of Magainin 2 (MG2) to Bombesin (MG2B), another CPP, led to pronounced anticancer effects. Other examples are CopA3, that selectively decreased the viability of gastric cancer cells, and the CPP p28. Furthermore, in preclinical tests, the anti-tumor efficacy of this peptide was evaluated on human breast cancer, prostate cancer, ovarian cancer, and melanoma cells in vitro, leading to high expression of p53 and promoting cell cycle arrest. Despite the numerous in vitro and in vivo studies with promising results, and the increasing number of clinical trials using CPPs, few treatments reach the expected clinical efficacy. Usually, their clinical application is limited by its poor aqueous solubility, immunogenicity issues and dose-limiting toxicity. This review describes the most recent advances and innovations in the use of CPPs in several types of cancer, highlighting their crucial importance for various purposes, from therapeutic to diagnosis. Further clinical trials with these peptides are warranted to examine its effects on various types of cancer.
Collapse
|
8
|
Li Z, Zhang Y, Zhu C, Guo T, Xia Q, Hou X, Liu W, Feng N. Folic acid modified lipid-bilayer coated mesoporous silica nanoparticles co-loading paclitaxel and tanshinone IIA for the treatment of acute promyelocytic leukemia. Int J Pharm 2020; 586:119576. [PMID: 32603839 DOI: 10.1016/j.ijpharm.2020.119576] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022]
Abstract
In this work, paclitaxel (Ptx) combined with tanshinone IIA (TanIIA) was found to show synergistic effect on inducing apoptosis of human acute promyelocytic leukemia (APL) cell line NB4, and the anti-tumor effect was strongest when its molar ratio was 1:1. To enhance the efficacy and reduce side effects, an active targeting drug delivery system with mesoporous silica nanoparticles (MSNs) coated with folic acid (FA) modified PEGylated lipid-bilayer (LB) membrane (FA-LB-MSNs) was established for co-loading drugs. The drug loadings of Ptx and TanIIA in FA-LB-MSNs were 5.5% and 1.8%, respectively. Compared with the uncoated MSNs, the FA-LB-MSNs showed a sustained drug release, and Ptx and TanIIA released synchronously from the carriers. By means of biological adhesion between FA and its receptors, the uptake of FA-LB-MSNs by NB4 cells was significantly higher than that of uncoated preparations, and Ptx combined with TanIIA had strong synergistic effect to enhance the apoptosis and differentiation of NB4 cells. The results of pharmacodynamics in vivo showed that the FA-LB-MSNs targeted tumor in nude mice more effectively than the compared formulations without FA modification. The Ptx and TanIIA-loaded FA-LB-MSNs group showed significantly better effects on inducing apoptosis and inhibiting tumor growth than the reference groups, which agreed with the results of anti-tumor experiments in vitro. Furthermore, no toxicity was observed to the heart, liver, spleen, lung and kidney of the tumor-bearing animals, indicating good biocompatibility of the prepared novel nanocarriers. This study confirmed the synergistic therapeutic effect of Ptx and TanIIA on APL, and the superior of FA-LB-MSNs as co-loaded nanocarriers for active targeted therapy of tumors.
Collapse
Affiliation(s)
- Zhe Li
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Chunyun Zhu
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Teng Guo
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Qing Xia
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Xuefeng Hou
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Wei Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China.
| |
Collapse
|
9
|
Shaw SK, Liu W, Gómez Durán CFA, Schreiber CL, Betancourt Mendiola MDL, Zhai C, Roland FM, Padanilam SJ, Smith BD. Non-Covalently Pre-Assembled High-Performance Near-Infrared Fluorescent Molecular Probes for Cancer Imaging. Chemistry 2018; 24:13821-13829. [PMID: 30022552 PMCID: PMC6415912 DOI: 10.1002/chem.201801825] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/05/2018] [Indexed: 12/15/2022]
Abstract
New fluorescent molecular probes, which can selectively target specific cell surface receptors, are needed for microscopy, in vivo imaging, and image guided surgery. The preparation of multivalent probes using standard synthetic chemistry can be a laborious process due to low reaction yields caused by steric effects. In this study, fluorescent molecular probes were prepared by a programmed non-covalent pre-assembly process that used a near-infrared fluorescent squaraine dye to thread a macrocycle bearing a cyclic arginine-glycine-aspartate peptide antagonist (cRGDfK) as a cancer targeting unit. Cell microscopy studies using OVCAR-4 (ovarian cancer) and A549 (lung cancer) cells that express high levels of the integrin αvβ3 or αvβ5 receptors, respectively, revealed a multivalent cell targeting effect. That is, there was comparatively more cell uptake of a pre-assembled probe equipped with two copies of the cRGDfK antagonist than a pre-assembled probe with only one appended cRGDfK antagonist. The remarkably high photostability and low phototoxicity of these near-infrared probes allowed for acquisition of long-term fluorescence movies showing endosome trafficking in living cells. In vivo near-infrared fluorescence imaging experiments compared the biodistribution of a targeted and untargeted probe in a xenograft mouse tumor model. The average tumor-to-muscle ratio for the pre-assembled targeted probe was 3.6 which matches the tumor targeting performance reported for analogous cRGDfK-based probes that were prepared entirely by covalent synthesis. The capability to excite these pre-assembled near-infrared fluorescent probes with blue or deep-red excitation light makes it possible to determine if a target site is located superficially or buried in tissue, a probe performance feature that is likely to be very helpful for eventual applications such as fluorescence guided surgery.
Collapse
Affiliation(s)
- Scott K Shaw
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | - Wenqi Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | | | - Cynthia L Schreiber
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | | | - Canjia Zhai
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | - Felicia M Roland
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | - Simon J Padanilam
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| | - Bradley D Smith
- Department of Chemistry & Biochemistry, University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN, 46545, USA
| |
Collapse
|
10
|
Abstract
There is a growing interest for the discovery of new cancer-targeted delivery systems for drug delivery and diagnosis. A synopsis of the bibliographic data will be presented on bombesin, neurotensin, octreotide, Arg-Gly-Asp, luteinizing hormone-releasing hormone and other peptides. Many of them have reached the clinics for therapeutic or diagnostic purposes, and have been utilized as carriers of known cytotoxic agents such as doxorubicin, paclitaxel, cisplatin, methotrexate or dyes and radioisotopes. In our article, recent advances in the development of peptides as carriers of cytotoxic drugs or radiometals will be analyzed.
Collapse
|
11
|
Raccagni I, Belloli S, Valtorta S, Stefano A, Presotto L, Pascali C, Bogni A, Tortoreto M, Zaffaroni N, Daidone MG, Russo G, Bombardieri E, Moresco RM. [18F]FDG and [18F]FLT PET for the evaluation of response to neo-adjuvant chemotherapy in a model of triple negative breast cancer. PLoS One 2018; 13:e0197754. [PMID: 29791503 PMCID: PMC5965848 DOI: 10.1371/journal.pone.0197754] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
Rationale Pathological response to neo-adjuvant chemotherapy (NAC) represents a commonly used predictor of survival in triple negative breast cancer (TNBC) and the need to identify markers that predict response to NAC is constantly increasing. Aim of this study was to evaluate the potential usefulness of PET imaging with [18F]FDG and [18F]FLT for the discrimination of TNBC responders to Paclitaxel (PTX) therapy compared to the response assessed by an adapted Response Evaluation Criteria In Solid Tumors (RECIST) criteria based on tumor volume (Tumor Volume Response). Methods Nu/nu mice bearing TNBC lesions of different size were evaluated with [18F]FDG and [18F]FLT PET before and after PTX treatment. SUVmax, Metabolic Tumor Volume (MTV) and Total Lesion Glycolysis (TLG) and Proliferation (TLP) were assessed using a graph-based random walk algorithm. Results We found that in our TNBC model the variation of [18F]FDG and [18F]FLT SUVmax similarly defined tumor response to therapy and that SUVmax variation represented the most accurate parameter. Response evaluation using Tumor Volume Response (TVR) showed that the effectiveness of NAC with PTX was completely independent from lesions size at baseline. Conclusions Our study provided interesting results in terms of sensitivity and specificity of PET in TNBC, revealing the similar performances of [18F]FDG and [18F]FLT in the identification of responders to Paclitaxel.
Collapse
Affiliation(s)
- Isabella Raccagni
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Tecnomed, Foundation of the University of Milano-Bicocca, Monza, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Belloli
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Valtorta
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
| | - Luca Presotto
- Nuclear Medicine Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Claudio Pascali
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Anna Bogni
- Nuclear Medicine Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Grazia Daidone
- Biomarkers Unit, Experimental Oncology and Molecular Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
| | | | - Rosa Maria Moresco
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
- Tecnomed, Foundation of the University of Milano-Bicocca, Monza, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Medicine and Surgery Department, University of Milano-Bicocca, Monza, Italy
- * E-mail:
| |
Collapse
|
12
|
Shaw SK, Schreiber CL, Roland FM, Battles PM, Brennan SP, Padanilam SJ, Smith BD. High expression of integrin αvβ3 enables uptake of targeted fluorescent probes into ovarian cancer cells and tumors. Bioorg Med Chem 2018; 26:2085-2091. [PMID: 29548784 DOI: 10.1016/j.bmc.2018.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 02/06/2023]
Abstract
The cell line OVCAR-4 was recently ranked as one of the most representative cell lines for high grade serous ovarian cancer (HGSOC). However, little work has been done to assess the susceptibility of OVCAR-4 cells and tumors to the more common types of molecular targeting. Proteome profiles suggest OVCAR-4 express high levels of integrin αvβ3 receptors. Using flow cytometry with fluorescent antibodies we determined that OVCAR-4 cells have high number of integrin αvβ3 receptors ([9.8 ± 2.5] × 104/cell) compared to the well-characterized cell line U87-MG ([5.2 ± 1.4] × 104/cell). However, OVCAR-4 cells also have roughly three times the surface area of U87-MG cells, so the average αvβ3 receptor density is actually lower (11 ± 3 versus 18 ± 6 receptors/µm2). A series of new fluorescent molecular probes was prepared with structures comprised of a deep-red squaraine fluorophore (∼680 nm emission) covalently attached to zero, one, or two cyclic pentapeptide cRGD sequences for integrin targeting. Microscopy studies showed that uptake of the divalent probe into cultured OVCAR-4 cells was 2.2 ± 0.4 higher than the monovalent probe, which in turn was 2.2 ± 0.4 higher than the untargeted probe. This probe targeting trend was also seen in OVCAR-4 mouse tumor models. The results suggest that clinically relevant OVCAR-4 cells can be targeted using molecular probes based on αvβ3 integrin receptor antagonists such as the cRGD peptide. Furthermore, deep-red fluorescent cRGD-squaraine probes have potential as targeted stains of cancerous tissue associated with HGSOC in surgery and pathology settings.
Collapse
Affiliation(s)
- Scott K Shaw
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Cynthia L Schreiber
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Felicia M Roland
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Paul M Battles
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Seamus P Brennan
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Simon J Padanilam
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States
| | - Bradley D Smith
- University of Notre Dame, 236 Nieuwland Science Hall, Notre Dame, IN 46545, United States.
| |
Collapse
|
13
|
Cazzamalli S, Dal Corso A, Widmayer F, Neri D. Chemically Defined Antibody- and Small Molecule-Drug Conjugates for in Vivo Tumor Targeting Applications: A Comparative Analysis. J Am Chem Soc 2018; 140:1617-1621. [PMID: 29342352 PMCID: PMC5844464 DOI: 10.1021/jacs.7b13361] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We present the first direct comparative evaluation of an antibody-drug conjugate and of a small molecule-drug conjugate for cancer therapy, using chemically defined products which bind with high-affinity to carbonic anhydrase IX, a marker of tumor hypoxia and of renal cell carcinoma.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Carbonic Anhydrase IX/antagonists & inhibitors
- Carbonic Anhydrase IX/metabolism
- Carbonic Anhydrase Inhibitors/chemical synthesis
- Carbonic Anhydrase Inhibitors/chemistry
- Carbonic Anhydrase Inhibitors/pharmacology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Humans
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/pharmacology
- Tumor Hypoxia/drug effects
Collapse
Affiliation(s)
- Samuele Cazzamalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zurich (Switzerland)
| | - Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zurich (Switzerland)
| | - Fontaine Widmayer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zurich (Switzerland)
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zurich (Switzerland)
| |
Collapse
|
14
|
Cao Q, Yan X, Chen K, Huang Q, Melancon MP, Lopez G, Cheng Z, Li C. Macrophages as a potential tumor-microenvironment target for noninvasive imaging of early response to anticancer therapy. Biomaterials 2017; 152:63-76. [PMID: 29111494 DOI: 10.1016/j.biomaterials.2017.10.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/30/2017] [Accepted: 10/19/2017] [Indexed: 10/18/2022]
Abstract
As a result of therapy-induced apoptosis, peripheral blood monocytes are recruited to tumors, where they become tumor-associated macrophages (TAMs). To date, few studies have investigated noninvasive molecular imaging for assessment of macrophage infiltration in response to therapy-induced apoptosis. Here, noninvasive assessment of changes in tumor accumulation of TAMs was proposed as a new way to measure early tumor response to anticancer therapy. Three different nanoparticles, QD710-Dendron quantum dots (QD710-D), Ferumoxytol, and PG-Gd-NIR813, were used for near-infrared fluorescence imaging, T2-weighted magnetic resonance imaging, and dual optical/T1-weighted MR imaging, respectively, in the MDA-MB-435 tumor model. Treatment with Abraxane induced tumor apoptosis and infiltrating macrophages. In spite of markedly different physicochemical properties among the nanoparticles, in vivo imaging revealed increased uptake of all three nanoparticles in Abraxane-treated tumors compared with untreated tumors. Moreover, imaging visualized increased uptake of QD710-D in MDA-MB-435 tumors but not in drug-resistant MDA-MB-435R tumors grown in the mice treated with Abraxane. Our results suggest that infiltration of macrophages due to chemotherapy-induced apoptosis was partially responsible for increased nanoparticle uptake in treated tumors. Noninvasive imaging techniques in conjunction with systemic administration of imageable nanoparticles that are taken up by macrophages are a potentially useful tool for assessing early treatment response.
Collapse
Affiliation(s)
- Qizhen Cao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Xinrui Yan
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Kai Chen
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Qian Huang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marites P Melancon
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gabriel Lopez
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhen Cheng
- Molecular Imaging Program at Stanford, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA, United States.
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Experimental Therapeutics Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, United States.
| |
Collapse
|
15
|
Dal Corso A, Gébleux R, Murer P, Soltermann A, Neri D. A non-internalizing antibody-drug conjugate based on an anthracycline payload displays potent therapeutic activity in vivo. J Control Release 2017; 264:211-218. [PMID: 28867376 DOI: 10.1016/j.jconrel.2017.08.040] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 12/29/2022]
Abstract
Antibody-drug conjugates are generally believed to crucially rely on internalization into cancer cells for therapeutic activity. Here, we show that a non-internalizing antibody-drug conjugate, based on the F16 antibody specific to the alternatively spliced A1 domain of tenascin-C, mediates a potent therapeutic activity when equipped with the anthracycline PNU159682. The peptide linker, connecting the F16 antibody in IgG format at a specific cysteine residue to the drug, was stable in serum but could be efficiently cleaved in the subendothelial extracellular matrix by proteases released by the dying tumor cells. The results indicate that there may be a broader potential applicability of non-internalizing antibody-drug conjugates for cancer therapy than what had previously been assumed.
Collapse
Affiliation(s)
- Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Rémy Gébleux
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Patrizia Murer
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland
| | - Alex Soltermann
- Institute of Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, 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
|
16
|
Zanella S, Angerani S, Pina A, López Rivas P, Giannini C, Panzeri S, Arosio D, Caruso M, Gasparri F, Fraietta I, Albanese C, Marsiglio A, Pignataro L, Belvisi L, Piarulli U, Gennari C. Tumor Targeting with an isoDGR-Drug Conjugate. Chemistry 2017; 23:7910-7914. [PMID: 28449309 PMCID: PMC5488297 DOI: 10.1002/chem.201701844] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Indexed: 11/20/2022]
Abstract
Herein we report the first example of an isoDGR-drug conjugate (2), designed to release paclitaxel selectively within cancer cells expressing integrin αV β3 . Conjugate 2 was synthesized by connecting the isoDGR peptidomimetic 5 with paclitaxel via the lysosomally cleavable Val-Ala dipeptide linker. Conjugate 2 displayed a low nanomolar affinity for the purified integrin αV β3 receptor (IC50 =11.0 nm). The tumor targeting ability of conjugate 2 was assessed in vitro in anti-proliferative assays on two isogenic cancer cell lines characterized by different integrin αV β3 expression: human glioblastoma U87 (αV β3 +) and U87 β3 -KO (αV β3 -). The isoDGR-PTX conjugate 2 displayed a remarkable targeting index (TI=9.9), especially when compared to the strictly related RGD-PTX conjugate 4 (TI=2.4).
Collapse
Affiliation(s)
- Simone Zanella
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Simona Angerani
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Arianna Pina
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Paula López Rivas
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Clelia Giannini
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Silvia Panzeri
- Dipartimento di Scienza e Alta TecnologiaUniversità degli Studi dell'InsubriaVia Valleggio 1122100ComoItaly
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM)CNRVia C. Golgi 1920133MilanoItaly
| | - Michele Caruso
- Nerviano Medical Sciences (NMS)Via Pasteur 1020014NervianoItaly
| | - Fabio Gasparri
- Nerviano Medical Sciences (NMS)Via Pasteur 1020014NervianoItaly
| | - Ivan Fraietta
- Nerviano Medical Sciences (NMS)Via Pasteur 1020014NervianoItaly
| | - Clara Albanese
- Nerviano Medical Sciences (NMS)Via Pasteur 1020014NervianoItaly
| | | | - Luca Pignataro
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Laura Belvisi
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| | - Umberto Piarulli
- Dipartimento di Scienza e Alta TecnologiaUniversità degli Studi dell'InsubriaVia Valleggio 1122100ComoItaly
| | - Cesare Gennari
- Dipartimento di ChimicaUniversità degli Studi di MilanoVia C. Golgi 1920133MilanoItaly
| |
Collapse
|
17
|
Karageorgis A, Claron M, Jugé R, Aspord C, Thoreau F, Leloup C, Kucharczak J, Plumas J, Henry M, Hurbin A, Verdié P, Martinez J, Subra G, Dumy P, Boturyn D, Aouacheria A, Coll JL. Systemic Delivery of Tumor-Targeted Bax-Derived Membrane-Active Peptides for the Treatment of Melanoma Tumors in a Humanized SCID Mouse Model. Mol Ther 2017; 25:534-546. [PMID: 28153100 DOI: 10.1016/j.ymthe.2016.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/09/2016] [Accepted: 11/11/2016] [Indexed: 01/03/2023] Open
Abstract
Melanoma is a highly metastatic and deadly form of cancer. Invasive melanoma cells overexpress integrin αvβ3, which is a well-known target for Arg-Gly-Asp-based (RGD) peptides. We developed a sophisticated method to synthetize milligram amounts of a targeted vector that allows the RGD-mediated targeting, internalization, and release of a mitochondria-disruptive peptide derived from the pro-apoptotic Bax protein. We found that 2.5 μM Bax[109-127] was sufficient to destabilize the mitochondria in ten different tumor cell lines, even in the presence of the anti-apoptotic Bcl2 protein, which is often involved in tumor resistance. This pore-forming peptide displayed antitumor activity when it was covalently linked by a disulfide bridge to the tetrameric RAFT-c[RGD]4-platform and after intravenous injection in a human melanoma tumor model established in humanized immuno-competent mice. In addition to its direct toxic effect, treatment with this combination induced the release of the immuno-stimulating factor monocyte chimoattractant protein 1 (MCP1) in the blood and a decrease in the level of the pro-angiogenic factor FGF2. Our novel multifunctional, apoptosis-inducing agent could be further customized and assayed for potential use in tumor-targeted therapy.
Collapse
Affiliation(s)
- Anastassia Karageorgis
- INSERM U1209, Institut Albert Bonniot, 38706 La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France
| | - Michaël Claron
- Université Grenoble Alpes, 38000 Grenoble, France; CNRS UMR 5250, ICMG FR2607, 38000 Grenoble, France
| | - Romain Jugé
- Molecular Biology of the Cell Laboratory (LBMC), Ecole Normale Supérieure de Lyon, UMR 5239 CNRS - UCBL - ENS Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Caroline Aspord
- Université Grenoble Alpes, 38000 Grenoble, France; EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology and Immunotherapy of Chronic Diseases, 38706 La Tronche, France; EFS Rhone-Alpes, R&D Laboratory, 38701 La Tronche, France
| | - Fabien Thoreau
- INSERM U1209, Institut Albert Bonniot, 38706 La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France; CNRS UMR 5250, ICMG FR2607, 38000 Grenoble, France
| | - Claire Leloup
- Université Grenoble Alpes, 38000 Grenoble, France; EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology and Immunotherapy of Chronic Diseases, 38706 La Tronche, France; EFS Rhone-Alpes, R&D Laboratory, 38701 La Tronche, France
| | - Jérôme Kucharczak
- Molecular Biology of the Cell Laboratory (LBMC), Ecole Normale Supérieure de Lyon, UMR 5239 CNRS - UCBL - ENS Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Joël Plumas
- Université Grenoble Alpes, 38000 Grenoble, France; EMR EFS-UGA-INSERM U1209- CNRS, Immunobiology and Immunotherapy of Chronic Diseases, 38706 La Tronche, France; EFS Rhone-Alpes, R&D Laboratory, 38701 La Tronche, France
| | - Maxime Henry
- INSERM U1209, Institut Albert Bonniot, 38706 La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France
| | - Amandine Hurbin
- INSERM U1209, Institut Albert Bonniot, 38706 La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France
| | - Pascal Verdié
- CNRS UMR 5247, Institut des Biomolécules Max Mousseron IBMM, 34095 Montpellier, France
| | - Jean Martinez
- CNRS UMR 5247, Institut des Biomolécules Max Mousseron IBMM, 34095 Montpellier, France
| | - Gilles Subra
- CNRS UMR 5247, Institut des Biomolécules Max Mousseron IBMM, 34095 Montpellier, France
| | - Pascal Dumy
- CNRS UMR 5250, ICMG FR2607, 38000 Grenoble, France; CNRS UMR 5247, Institut des Biomolécules Max Mousseron IBMM, 34095 Montpellier, France
| | - Didier Boturyn
- Université Grenoble Alpes, 38000 Grenoble, France; CNRS UMR 5250, ICMG FR2607, 38000 Grenoble, France
| | - Abdel Aouacheria
- Molecular Biology of the Cell Laboratory (LBMC), Ecole Normale Supérieure de Lyon, UMR 5239 CNRS - UCBL - ENS Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France; Institut des Sciences de l'Evolution de Montpellier (ISEM), UMR 5554, Université de Montpellier, CNRS, IRD, EPHE, Place Eugène Bataillon, 34095 Montpellier, France.
| | - Jean-Luc Coll
- INSERM U1209, Institut Albert Bonniot, 38706 La Tronche, France; Université Grenoble Alpes, 38000 Grenoble, France.
| |
Collapse
|
18
|
Abstract
Conjugates of cytotoxic agents with RGD peptides (Arg-Gly-Asp) addressed to ανβ3, α5β1 and ανβ6 integrin receptors overexpressed by cancer cells, have recently gained attention as potential selective anticancer chemotherapeutics. In this review, the design and the development of RGD conjugates coupled to different small molecules including known cytotoxic drugs and natural products will be discussed.
Collapse
|
19
|
Schelhaas S, Heinzmann K, Bollineni VR, Kramer GM, Liu Y, Waterton JC, Aboagye EO, Shields AF, Soloviev D, Jacobs AH. Preclinical Applications of 3'-Deoxy-3'-[ 18F]Fluorothymidine in Oncology - A Systematic Review. Theranostics 2017; 7:40-50. [PMID: 28042315 PMCID: PMC5196884 DOI: 10.7150/thno.16676] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/16/2016] [Indexed: 11/05/2022] Open
Abstract
The positron emission tomography (PET) tracer 3'-deoxy-3'-[18F]fluorothymidine ([18F]FLT) has been proposed to measure cell proliferation non-invasively in vivo. Hence, it should provide valuable information for response assessment to tumor therapies. To date, [18F]FLT uptake has found limited use as a response biomarker in clinical trials in part because a better understanding is needed of the determinants of [18F]FLT uptake and therapy-induced changes of its retention in the tumor. In this systematic review of preclinical [18F]FLT studies, comprising 174 reports, we identify the factors governing [18F]FLT uptake in tumors, among which thymidine kinase 1 plays a primary role. The majority of publications (83 %) report that decreased [18F]FLT uptake reflects the effects of anticancer therapies. 144 times [18F]FLT uptake was related to changes in proliferation as determined by ex vivo analyses. Of these approaches, 77 % describe a positive relation, implying a good concordance of tracer accumulation and tumor biology. These preclinical data indicate that [18F]FLT uptake holds promise as an imaging biomarker for response assessment in clinical studies. Understanding of the parameters which influence cellular [18F]FLT uptake and retention as well as the mechanism of changes induced by therapy is essential for successful implementation of this PET tracer. Hence, our systematic review provides the background for the use of [18F]FLT in future clinical studies.
Collapse
Affiliation(s)
- Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany
| | | | - Vikram R Bollineni
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Gerbrand M Kramer
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Yan Liu
- European Organization for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | | | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre, Imperial College London, UK
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Dmitry Soloviev
- Cancer Research UK Cambridge Institute, University of Cambridge, UK
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität (WWU) Münster, Münster, Germany.; Department of Geriatric Medicine, Johanniter Hospital, Bonn, 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: 50] [Impact Index Per Article: 6.3] [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
|
Cazzamalli S, Dal Corso A, Neri D. Acetazolamide Serves as Selective Delivery Vehicle for Dipeptide-Linked Drugs to Renal Cell Carcinoma. Mol Cancer Ther 2016; 15:2926-2935. [PMID: 27609641 DOI: 10.1158/1535-7163.mct-16-0283] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/27/2016] [Accepted: 08/28/2016] [Indexed: 01/12/2023]
Abstract
In most cases, cytotoxic drugs do not preferentially accumulate at the tumor site, causing unwanted toxicities and preventing dose escalation to therapeutically active regimens. Here, we show that acetazolamide derivatives, which bind to carbonic anhydrase IX (CAIX) on the surface of kidney cancer cells, selectively deliver payloads at the site of disease, sparing normal organs. Biodistribution studies, performed in tumor-bearing mice with acetazolamide derivatives bearing a technetium-99m chelator complex or a red fluorophore as payload, revealed a preferential tumor accumulation of the compound at doses up to 560 nmol/kg. The percentage of injected dose per gram in the tumor was dose-dependent and revealed optimal tumor:organ ratios at 140 nmol/kg, with a tumor:blood ratio of 80:1 at 6 hours. Acetazolamide, coupled to potent cytotoxic drugs via a dipeptide linker, exhibited a potent antitumor activity in nude mice bearing SKRC-52 renal cell carcinomas, whereas drug derivatives devoid of the acetazolamide moiety did not exhibit any detectable anticancer activity at the same doses. The observation of tumor regression with a noninternalizing ligand and with different cytotoxic moieties (MMAE and PNU-159682) indicates a general mechanism of action, based on the selective accumulation of the product on tumor cells, followed by the extracellular proteolytic release of the cytotoxic payload at the neoplastic site and the subsequent drug internalization into tumor cells. Acetazolamide-based drug conjugates may represent a promising class of targeted agents for the treatment of metastatic kidney cancer, as the majority of human clear cell renal cell carcinomas are strongly positive for CAIX. Mol Cancer Ther; 15(12); 2926-35. ©2016 AACR.
Collapse
Affiliation(s)
- Samuele Cazzamalli
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland)
| | - Alberto Dal Corso
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland)
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich (Switzerland).
| |
Collapse
|
22
|
Bianchi A, Arosio D, Perego P, De Cesare M, Carenini N, Zaffaroni N, De Matteo M, Manzoni L. Design, synthesis and biological evaluation of novel dimeric and tetrameric cRGD-paclitaxel conjugates for integrin-assisted drug delivery. Org Biomol Chem 2016; 13:7530-41. [PMID: 26074454 DOI: 10.1039/c5ob00497g] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Integrins are associated with tumour cell survival and progression, and their expression has been shown to be increased in tumours. Thus, four novel conjugates of the tripeptide integrin ligand Arg-Gly-Asp (RGD) and the cytotoxic agent paclitaxel (cRGD-PTX) were prepared to investigate the potential of the multivalent presentation of the RGD moiety in improving the antitumor efficacy of PTX by tumour targeting. PTX was conjugated to two or four integrin recognizing ligands. The influence of multivalent presentation on in vitro αvβ3-receptor affinity was confirmed. For all the conjugates compared to the previously synthesized monovalent counterparts, an enhancement of the binding strength was observed; this behaviour was more pronounced when considering the tetravalent presented RGD-conjugate. Cell growth inhibition assays on a panel of human tumour cell lines showed remarkable cytotoxic activity for all conjugates with IC50 values in a nanomolar range. Among the four conjugates, the bivalent derivative 3b was selected for in vivo studies in an ovarian carcinoma cell model xenografted in immunodeficient mice. A marked antitumor activity was observed, similar to that of PTX, but with a much more favourable toxicity profile. Overall, the novel cRGD-PTX conjugates disclosed here represent promising candidates for further advancement in the domain of targeted anti-tumour therapy.
Collapse
Affiliation(s)
- A Bianchi
- Centro Interdipartimentale Studi Biomolecolari e Applicazioni Industriali, Università degli Studi di Milano, Via Fantoli 16/15, I-20138 Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kue CS, Kamkaew A, Burgess K, Kiew LV, Chung LY, Lee HB. Small Molecules for Active Targeting in Cancer. Med Res Rev 2016; 36:494-575. [PMID: 26992114 DOI: 10.1002/med.21387] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/29/2022]
Abstract
For the purpose of this review, active targeting in cancer research encompasses strategies wherein a ligand for a cell surface receptor expressed on tumor cells is used to deliver a cytotoxic or imaging cargo. This area of research is more than two decades old, but in those 20 and more years, how many receptors have been studied extensively? What kinds of the ligands are used for active targeting? Are they mostly naturally occurring molecules such as folic acid, or synthetic substances developed in campaigns for medicinal chemistry efforts? This review outlines the most important receptor or ligand combinations that have been used in active targeting to answer these questions, and therefore to address the most important one of all: is research in active targeting affording diminishing returns, or is this an area for which the potential far exceeds progress made so far?
Collapse
Affiliation(s)
- Chin S Kue
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Anyanee Kamkaew
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Kevin Burgess
- Department of Chemistry, Texas A & M University, Box 30012, College Station, TX, 77842
| | - Lik V Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Lip Y Chung
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hong B Lee
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|
24
|
Arosio D, Casagrande C. Advancement in integrin facilitated drug delivery. Adv Drug Deliv Rev 2016; 97:111-43. [PMID: 26686830 DOI: 10.1016/j.addr.2015.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
Abstract
The research of integrin-targeted anticancer agents has recorded important advancements in ingenious design of delivery systems, based either on the prodrug approach, or on nanoparticle carriers, but for now, none of these has reached a clinical stage of development. Past work in this area has been extensively reviewed by us and others. Thus, the purpose and scope of the present review is to survey the advancement reported in the last 3years, with focus on innovative delivery systems that appear to afford openings for future developments. These systems exploit the labelling with conventional and novel integrin ligands for targeting the interface of cancer cells and of endothelial cells involved in cancer angiogenesis, with the proteins of the extracellular matrix, in the circulation, in tissues, and in tumour stroma, as the site of progression and metastatic evolution of the disease. Furthermore, these systems implement the expertise in the development of nanomedicines to the purpose of achieving preferential biodistribution and uptake in cancer tissues, internalisation in cancer cells, and release of the transported drugs at intracellular sites. The assessment of the value of controlling these factors, and their combination, for future developments requires support of biological testing in appropriate mechanistic models, but also imperatively demand confirmation in therapeutically relevant in vivo models for biodistribution, efficacy, and lack of off-target effects. Thus, among many studies, we have tried to point out the results supported by relevant in vivo studies, and we have emphasised in specific sections those addressing the medical needs of drug delivery to brain tumours, as well as the delivery of oligonucleotides modulating gene-dependent pathological mechanism. The latter could constitute the basis of a promising third branch in the therapeutic armamentarium against cancer, in addition to antibody-based agents and to cytotoxic agents.
Collapse
Affiliation(s)
- Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM), CNR, Via C. Golgi 19, I-20133 Milan, Italy.
| | - Cesare Casagrande
- Università degli Studi di Milano, Dipartimento di Chimica, Via C. Golgi 19, I-20133 Milan, Italy.
| |
Collapse
|
25
|
Wu CH, Liu IJ, Lu RM, Wu HC. Advancement and applications of peptide phage display technology in biomedical science. J Biomed Sci 2016; 23:8. [PMID: 26786672 PMCID: PMC4717660 DOI: 10.1186/s12929-016-0223-x] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
Combinatorial phage library is a powerful research tool for high-throughput screening of protein interactions. Of all available molecular display techniques, phage display has proven to be the most popular approach. Screening phage-displayed random peptide libraries is an effective means of identifying peptides that can bind target molecules and regulate their function. Phage-displayed peptide libraries can be used for (i) B-cell and T-cell epitope mapping, (ii) selection of bioactive peptides bound to receptors or proteins, disease-specific antigen mimics, peptides bound to non-protein targets, cell-specific peptides, or organ-specific peptides, and (iii) development of peptide-mediated drug delivery systems and other applications. Targeting peptides identified using phage display technology may be useful for basic research and translational medicine. In this review article, we summarize the latest technological advancements in the application of phage-displayed peptide libraries to applied biomedical sciences.
Collapse
Affiliation(s)
- Chien-Hsun Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - I-Ju Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
26
|
Dal Corso A, Caruso M, Belvisi L, Arosio D, Piarulli U, Albanese C, Gasparri F, Marsiglio A, Sola F, Troiani S, Valsasina B, Pignataro L, Donati D, Gennari C. Synthesis and Biological Evaluation of RGD Peptidomimetic-Paclitaxel Conjugates Bearing Lysosomally Cleavable Linkers. Chemistry 2015; 21:6921-9. [DOI: 10.1002/chem.201500158] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Indexed: 11/06/2022]
|
27
|
Protein– and Peptide–Drug Conjugates. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 98:1-55. [DOI: 10.1016/bs.apcsb.2014.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Abstract
For nearly two decades now, the RGD (Arg-Gly-Asp)-binding αvβ3-integrin has been a focus of anti-angiogenic drug design. These inhibitors are well-tolerated, but have shown only limited success in patients. Over the years, studies in β3-integrin-knockout mice have shed some light on possible explanations for disappointing clinical outcomes. However, studying angiogenesis in β3-integrin-knockout mice is a blunt tool to investigate β3-integrin's role in pathological angiogenesis. Since establishing our laboratory at University of East Anglia (UEA), we have adopted more refined models of genetically manipulating the expression of the β3-integrin subunit. The present review will highlight some of our findings from these models and describe how data from them have forced us to rethink how targeting αvβ3-integrin expression affects tumour angiogenesis and cancer progression. Revisiting the fundamental biology behind how this integrin regulates tumour growth and angiogenesis, we believe, is the key not only to understanding how angiogenesis is normally co-ordinated, but also in success with drugs directed against it.
Collapse
|
29
|
Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014; 351:13-22. [PMID: 24836189 DOI: 10.1016/j.canlet.2014.05.002] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/31/2014] [Accepted: 05/01/2014] [Indexed: 01/01/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer therapies mainly focus on mass cell killing without high specificity and often cause severe side effects and toxicities. Peptides are a novel class of anticancer agents that could specifically target cancer cells with lower toxicity to normal tissues, which will offer new opportunities for cancer prevention and treatment. Anticancer peptides face several therapeutic challenges. In this review, we present the sources and mechanisms of anticancer peptides and further discuss modification strategies to improve the anticancer effects of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanfeng Gao
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanming Qi
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China.
| | - Lixiang Chen
- School of Life Science, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yuanfang Ma
- College of Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Yanzhang Li
- College of Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
30
|
Liu J, Wang H, Wang Y, Yin Y, Du Z, Liu Z, Yang J, Hu S, Wang C, Chen Y. The stem cell adjuvant with Exendin-4 repairs the heart after myocardial infarction via STAT3 activation. J Cell Mol Med 2014; 18:1381-91. [PMID: 24779911 PMCID: PMC4124022 DOI: 10.1111/jcmm.12272] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 01/30/2014] [Indexed: 01/03/2023] Open
Abstract
The poor survival of cells in ischaemic myocardium is a major obstacle for stem cell therapy. Exendin-4 holds the potential of cardioprotective effect based on its pleiotropic activity. This study investigated whether Exendin-4 in conjunction with adipose-derived stem cells (ADSCs) could improve the stem cell survival and contribute to myocardial repairs after infarction. Myocardial infarction (MI) was induced by the left anterior descending artery ligation in adult male Sprague-Dawley rats. ADSCs carrying double-fusion reporter gene [firefly luciferase and monomeric red fluorescent protein (fluc-mRFP)] were quickly injected into border zone of MI in rats treated with or without Exendin-4. Exendin-4 enhanced the survival of transplanted ADSCs, as demonstrated by the longitudinal in vivo bioluminescence imaging. Moreover, ADSCs adjuvant with Exendin-4 decreased oxidative stress, apoptosis and fibrosis. They also improved myocardial viability and cardiac function and increased the differentiation rates of ADSCs into cardiomyocytes and vascular smooth muscle cells in vivo. Then, ADSCs were exposed to hydrogen peroxide/serum deprivation (H2O2/SD) to mimic the ischaemic environment in vitro. Results showed that Exendin-4 decreased the apoptosis and enhanced the paracrine effect of ADSCs. In addition, Exendin-4 activated signal transducers and activators of transcription 3 (STAT3) through the phosphorylation of Akt and ERK1/2. Furthermore, Exendin-4 increased the anti-apoptotic protein Bcl-2, but decreased the pro-apoptotic protein Bax of ADSCs. In conclusion, Exendin-4 could improve the survival and therapeutic efficacy of transplanted ADSCs through STAT3 activation via the phosphorylation of Akt and ERK1/2. This study suggests the potential application of Exendin-4 for stem cell–based heart regeneration.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Cardiology, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China; Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gill JH, Loadman PM, Shnyder SD, Cooper P, Atkinson JM, Ribeiro Morais G, Patterson LH, Falconer RA. Tumor-targeted prodrug ICT2588 demonstrates therapeutic activity against solid tumors and reduced potential for cardiovascular toxicity. Mol Pharm 2014; 11:1294-300. [PMID: 24641451 DOI: 10.1021/mp400760b] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Development of therapeutic strategies for tumor-selective delivery of therapeutics through exploitation of the proteolytic tumor phenotype has significant scope for improvement of cancer treatment. ICT2588 is a peptide-conjugated prodrug of the vascular disrupting agent (VDA) azademethylcolchicine developed to be selectively hydrolyzed by matrix metalloproteinase-14 (MMP-14) within the tumor. In this report, we extend our previous proof-of-concept studies and demonstrate the therapeutic potential of this agent against models of human colorectal, lung, breast, and prostate cancer. In all tumor types, ICT2588 was superior to azademethylcolchicine and was greater or comparable to standard clinically used agents for the respective tumor type. Prodrug activation in clinical human lung tumor homogenates relative to stability in human plasma and liver was observed, supporting clinical translation potential. A major limiting factor to the clinical value of VDAs is their inherent cardiovascular toxicity. No increase in plasma von Willebrand factor (vWF) levels, an indicator of systemic vascular dysfunction and acute cardiovascular toxicity, was detected with ICT2588, thereby supporting the tumor-selective activation and reduced potential of ICT2588 to cause cardiovascular toxicity. Our findings reinforce the improved therapeutic index and tumor-selective approach offered by ICT2588 and this nanotherapeutic approach.
Collapse
Affiliation(s)
- Jason H Gill
- Institute of Cancer Therapeutics, School of Life Sciences, University of Bradford , Bradford, Yorkshire BD7 1DP, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
UNLABELLED We recently demonstrated that a soluble protein, Gas6, can facilitate viral entry by bridging viral envelope phosphatidylserine to Axl, a receptor tyrosine kinase expressed on target cells. The interaction between phosphatidylserine, Gas6, and Axl was originally shown to be a molecular mechanism through which phagocytes recognize phosphatidylserine exposed on dead cells. Since our initial report, several groups have confirmed that Axl/Gas6, as well as other phosphatidylserine receptors, facilitate entry of dengue, West Nile, and Ebola viruses. Virus binding by viral envelope phosphatidylserine is now a viral entry mechanism generalized to many families of viruses. In addition to Axl/Gas6, various molecules are known to recognize phosphatidylserine; however, the effects of these molecules on virus binding and entry have not been comprehensively evaluated and compared. In this study, we examined most of the known human phosphatidylserine-recognizing molecules, including MFG-E8, TIM-1, -3, and -4, CD300a, BAI1, and stabilin-1 and -2, for their abilities to facilitate virus binding and infection. Using pseudotyped lentiviral vectors, we found that a soluble phosphatidylserine-binding protein, MFG-E8, enhances transduction. Cell surface receptors TIM-1 and -4 also enhance virus binding/transduction. The extent of enhancement by these molecules varies, depending on the type of pseudotyping envelope proteins. Mutated MFG-E8, which binds viral envelope phosphatidylserine without bridging virus to cells, but, surprisingly, not annexin V, which has been used to block phagocytosis of dead cells by concealing phosphatidylserine, efficiently blocks these phosphatidylserine-dependent viral entry mechanisms. These results provide insight into understanding the role of viral envelope phosphatidylserine in viral infection. IMPORTANCE Envelope phosphatidylserine has previously been shown to be important for replication of various envelope viruses, but details of this mechanism(s) were unclear. We were the first to report that a bifunctional serum protein, Gas6, bridges envelope phosphatidylserine to a cell surface receptor, Axl. Recent studies demonstrated that many envelope viruses, including vaccinia, dengue, West Nile, and Ebola viruses, utilize Axl/Gas6 to facilitate their entry, suggesting that the phosphatidylserine-mediated viral entry mechanism can be shared by various enveloped viruses. In addition to Axl/Gas6, various molecules are known to recognize phosphatidylserine; however, the effects of these molecules on virus binding and entry have not been comprehensively evaluated and compared. In this study, we examined most human phosphatidylserine-recognizing molecules for their abilities to facilitate viral infection. The results provide insights into the role(s) of envelope phosphatidylserine in viral infection, which can be applicable to the development of novel antiviral reagents that block phosphatidylserine-mediated viral entry.
Collapse
|
33
|
Krall N, Scheuermann J, Neri D. Small Targeted Cytotoxics: Current State and Promises from DNA-Encoded Chemical Libraries. Angew Chem Int Ed Engl 2013; 52:1384-402. [DOI: 10.1002/anie.201204631] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Indexed: 01/06/2023]
|
34
|
Krall N, Scheuermann J, Neri D. Entwicklung zielgerichteter niedermolekularer zytotoxischer Wirkstoffverbindungen mit DNA-codierten chemischen Bibliotheken. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201204631] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
35
|
Colombo R, Mingozzi M, Belvisi L, Arosio D, Piarulli U, Carenini N, Perego P, Zaffaroni N, De Cesare M, Castiglioni V, Scanziani E, Gennari C. Synthesis and biological evaluation (in vitro and in vivo) of cyclic arginine-glycine-aspartate (RGD) peptidomimetic-paclitaxel conjugates targeting integrin αVβ3. J Med Chem 2012; 55:10460-74. [PMID: 23140358 DOI: 10.1021/jm301058f] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A small library of integrin ligand-paclitaxel conjugates 10-13 was synthesized with the aim of using the tumor-homing cyclo[DKP-RGD] peptidomimetics for site-directed delivery of the cytotoxic drug. All the paclitaxel-RGD constructs 10-13 inhibited biotinylated vitronectin binding to the purified αVβ3 integrin receptor at low nanomolar concentration and showed in vitro cytotoxic activity against a panel of human tumor cell lines similar to that of paclitaxel. Among the cell lines, the cisplatin-resistant IGROV-1/Pt1 cells expressed high levels of integrin αVβ3, making them attractive to be tested in in vivo models. cyclo[DKP-f3-RGD]-PTX 11 displayed sufficient stability in physiological solution and in both human and murine plasma to be a good candidate for in vivo testing. In tumor-targeting experiments against the IGROV-1/Pt1 human ovarian carcinoma xenotransplanted in nude mice, compound 11 exhibited a superior activity compared with paclitaxel, despite the lower (about half) molar dosage used.
Collapse
Affiliation(s)
- Raffaele Colombo
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, I-20133, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Adulnirath A, Chung SW, Park J, Hwang SR, Kim JY, Yang VC, Kim SY, Moon HT, Byun Y. Cyclic RGDyk-conjugated LMWH-taurocholate derivative as a targeting angiogenesis inhibitor. J Control Release 2012; 164:8-16. [PMID: 23063549 DOI: 10.1016/j.jconrel.2012.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 08/17/2012] [Accepted: 10/02/2012] [Indexed: 01/05/2023]
Abstract
LMWH-taurocholate derivative (LHT7) has been reported as a novel angiogenesis inhibitor, due to its ability to bind to several kinds of growth factors, which play critical roles in tumor angiogenesis. In this study, we have highlighted the enhanced antiangiogenic activity of LHT7, by using cyclic RGDyk (cRGD), a targeting moiety that was chemically conjugated to LHT7 via amide bond. The SPR study revealed that cRGD-LHT7 bound to α(v)β(3) integrin as strongly as cRGD, and it bound to VEGF as strongly as LHT7. Importantly, in vitro anti-angiogenesis studies revealed that cRGD-LHT7 had a significant inhibition effect on HUVEC tubular formation. Finally, cRGD-LHT7 showed a greater inhibitory efficiency on the tumor growth in the U87MG xenograft model than the original LHT7, which was owed to its ability to target the tumor cells. All of these findings demonstrated that cRGD-LHT7 targeted α(v)β(3) integrin-positive cancer cells and endothelial cells, resulting in a greater anti-angiogenesis effect on the solid tumors.
Collapse
MESH Headings
- Angiogenesis Inhibitors/chemical synthesis
- Angiogenesis Inhibitors/chemistry
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/therapeutic use
- Animals
- Cell Line, Tumor
- Cell Survival/drug effects
- Heparin, Low-Molecular-Weight/analogs & derivatives
- Heparin, Low-Molecular-Weight/chemical synthesis
- Heparin, Low-Molecular-Weight/chemistry
- Heparin, Low-Molecular-Weight/pharmacology
- Heparin, Low-Molecular-Weight/therapeutic use
- Human Umbilical Vein Endothelial Cells
- Humans
- Integrin alphaVbeta3/metabolism
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Microscopy, Confocal
- Microtubules/drug effects
- Microtubules/ultrastructure
- Molecular Structure
- Peptides, Cyclic/chemistry
- Protein Binding
- Taurocholic Acid/analogs & derivatives
- Taurocholic Acid/chemical synthesis
- Taurocholic Acid/chemistry
- Taurocholic Acid/pharmacology
- Taurocholic Acid/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Asarasin Adulnirath
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Pilkington-Miksa M, Arosio D, Battistini L, Belvisi L, De Matteo M, Vasile F, Burreddu P, Carta P, Rassu G, Perego P, Carenini N, Zunino F, De Cesare M, Castiglioni V, Scanziani E, Scolastico C, Casiraghi G, Zanardi F, Manzoni L. Design, Synthesis, and Biological Evaluation of Novel cRGD–Paclitaxel Conjugates for Integrin-Assisted Drug Delivery. Bioconjug Chem 2012; 23:1610-22. [DOI: 10.1021/bc300164t] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Michael Pilkington-Miksa
- Centro Interdipartimentale
Studi Biomolecolari e Applicazioni Industriali, Università degli Studi di Milano, Via Fantoli
16/15, I-20138 Milano, Italy
| | - Daniela Arosio
- Istituto di Scienze
e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via Golgi 19, I-20133 Milano, Italy
| | - Lucia Battistini
- Dipartimento Farmaceutico, Università degli Studi di Parma, Parco Area
delle Scienze 27A, I-43124 Parma, Italy
| | - Laura Belvisi
- Dipartimento di Chimica
Organica e Industriale, Università degli Studi di Milano, Via Venezian 21, I-20133 Milano,
Italy
| | - Marilenia De Matteo
- Centro Interdipartimentale
Studi Biomolecolari e Applicazioni Industriali, Università degli Studi di Milano, Via Fantoli
16/15, I-20138 Milano, Italy
| | - Francesca Vasile
- Centro Interdipartimentale
Studi Biomolecolari e Applicazioni Industriali, Università degli Studi di Milano, Via Fantoli
16/15, I-20138 Milano, Italy
| | - Paola Burreddu
- Istituto
di Chimica
Biomolecolare, Consiglio Nazionale delle Ricerche, Traversa La Crucca 3, I-07100 Li Punti, Sassari,
Italy
| | - Paola Carta
- Porto Conte Ricerche Srl, I-07041 Tramariglio Alghero, Sassari, Italy
| | - Gloria Rassu
- Istituto
di Chimica
Biomolecolare, Consiglio Nazionale delle Ricerche, Traversa La Crucca 3, I-07100 Li Punti, Sassari,
Italy
| | - Paola Perego
- Dipartimento di Oncologia Sperimentale
e Medicina Molecolare, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, I-20133 Milano, Italy
| | - Nives Carenini
- Dipartimento di Oncologia Sperimentale
e Medicina Molecolare, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, I-20133 Milano, Italy
| | - Franco Zunino
- Dipartimento di Oncologia Sperimentale
e Medicina Molecolare, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, I-20133 Milano, Italy
| | - Michelandrea De Cesare
- Dipartimento di Oncologia Sperimentale
e Medicina Molecolare, Fondazione IRCCS Istituto Nazionale Tumori, Via Amadeo 42, I-20133 Milano, Italy
| | - Vittoria Castiglioni
- Dipartimento di
Patologia Animale, Igiene e Sanità Pubblica Veterinaria
(DIPAV), Facoltà di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, I-20133 Milano,
Italy
- Mouse and Animal Pathology
Laboratory, Fondazione Filarete, Viale
Ortles 22/4, I-20139 Milano,
Italy
| | - Eugenio Scanziani
- Dipartimento di
Patologia Animale, Igiene e Sanità Pubblica Veterinaria
(DIPAV), Facoltà di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, I-20133 Milano,
Italy
- Mouse and Animal Pathology
Laboratory, Fondazione Filarete, Viale
Ortles 22/4, I-20139 Milano,
Italy
| | - Carlo Scolastico
- Centro Interdipartimentale
Studi Biomolecolari e Applicazioni Industriali, Università degli Studi di Milano, Via Fantoli
16/15, I-20138 Milano, Italy
| | - Giovanni Casiraghi
- Dipartimento Farmaceutico, Università degli Studi di Parma, Parco Area
delle Scienze 27A, I-43124 Parma, Italy
| | - Franca Zanardi
- Dipartimento Farmaceutico, Università degli Studi di Parma, Parco Area
delle Scienze 27A, I-43124 Parma, Italy
| | - Leonardo Manzoni
- Istituto di Scienze
e Tecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via Golgi 19, I-20133 Milano, Italy
| |
Collapse
|
38
|
Lee MH, Kim JY, Han JH, Bhuniya S, Sessler JL, Kang C, Kim JS. Direct Fluorescence Monitoring of the Delivery and Cellular Uptake of a Cancer-Targeted RGD Peptide-Appended Naphthalimide Theragnostic Prodrug. J Am Chem Soc 2012; 134:12668-74. [DOI: 10.1021/ja303998y] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Min Hee Lee
- Department of Chemistry, Korea University, Seoul, 136-701, Korea
| | - Jin Young Kim
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | - Ji Hye Han
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | | | - Jonathan L. Sessler
- Department
of Chemistry and Biochemistry, The University of Texas at Austin, Austin, Texas 78712-0165,
United States
- Department
of Chemistry, Yonsei University, 262 Seonsanno
Sinchon-dong, Seodaemun-gu,
Seoul 120-749, Korea
| | - Chulhun Kang
- The School of East-West Medical
Science, Kyung Hee University, Yongin,
446-701, Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 136-701, Korea
| |
Collapse
|
39
|
Coughlin A, West J. Gold nanoshells for imaging and photothermal ablation of cancer. Nanomedicine (Lond) 2012. [DOI: 10.1533/9780857096449.2.326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
40
|
Animal tumor models for PET in drug development. Ann Nucl Med 2011; 25:717-31. [DOI: 10.1007/s12149-011-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
41
|
Abstract
Optical imaging has emerged as a powerful modality for studying molecular recognitions and molecular imaging in a noninvasive, sensitive, and real-time way. Some advantages of optical imaging include cost-effectiveness, convenience, and non-ionization safety as well as complementation with other imaging modalities such as positron emission tomography (PET), single-photon emission computed tomography (SPECT), and magnetic resonance imaging (MRI). Over the past decade, considerable advances have been made in tumor optical imaging by targeting integrin receptors in preclinical studies. This review has emphasized the construction and evaluation of diverse integrin targeting agents for optical imaging of tumors in mouse models. They mainly include some near-infrared fluorescent dye-RGD peptide conjugates, their multivalent analogs, and nanoparticle conjugates for targeting integrin αvβ3. Some compounds targeting other integrin subtypes such as α4β1 and α3 for tumor optical imaging have also been included. Both in vitro and in vivo studies have revealed some promising integrin-targeting optical agents which have further enhanced our understanding of integrin expression and targeting in cancer biology as well as related anticancer drug discovery. Especially, some integrin-targeted multifunctional optical agents including nanoparticle-based optical agents can multiplex optical imaging with other imaging modalities and targeted therapy, serving as an attractive type of theranostics for simultaneous imaging and targeted therapy. Continued efforts to discover and develop novel, innovative integrin-based optical agents with improved targeting specificity and imaging sensitivity hold great promises for improving cancer early detection, diagnosis, and targeted therapy in clinic.
Collapse
|
42
|
Zhang Q, Bindokas V, Shen J, Fan H, Hoffman RM, Xing HR. Time-course imaging of therapeutic functional tumor vascular normalization by antiangiogenic agents. Mol Cancer Ther 2011; 10:1173-84. [PMID: 21586628 DOI: 10.1158/1535-7163.mct-11-0008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We describe here new technology that enables noninvasive imaging of therapeutic functional normalization of tumor blood vessels by antiangiogenic agents. Noninvasive variable-magnification in vivo-fluorescence imaging as well as fluorescence tomography was used to visualize functional vessel normalization. Changes in the same vessel before and after drug treatment were imaged with high resolution in real time. Differences in vascular responses to the mTOR inhibitor rapamycin and to an anti-VEGF antibody were functionally imaged. Tumor vessel normalization was shown by significantly reduced leakiness and subsequent improved tumor delivery of Paclitaxel-BODPY as well as by normalized morphology. The tumor vascular pool agent, AngioSense(750), was retained only in tumors after either anti-VEGF antibody or rapamycin treatment, as visualized by noninvasive fluorescence tomography. The antiangiogenic therapy normalized vessels, which significantly enhanced the antitumor efficacy of paclitaxel because of increased drug penetration throughout the tumor. The optical imaging technology described here is thus a powerful, noninvasive, time-course imaging tool of functional tumor vessel normalization and its therapeutic consequences.
Collapse
Affiliation(s)
- Qingbei Zhang
- Department of Pathology, The University of Chicago, 5841 South Maryland Avenue, AMB N339, MC1089, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
43
|
Zhu S, Qian L, Hong M, Zhang L, Pei Y, Jiang Y. RGD-modified PEG-PAMAM-DOX conjugate: in vitro and in vivo targeting to both tumor neovascular endothelial cells and tumor cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H84-9. [PMID: 21360776 DOI: 10.1002/adma.201003944] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Indexed: 05/25/2023]
Affiliation(s)
- Saijie Zhu
- Department of Pharmaceutics, Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA School of Pharmacy, Fudan University, Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
44
|
Kimura S, Masunaga SI, Harada T, Kawamura Y, Ueda S, Okuda K, Nagasawa H. Synthesis and evaluation of cyclic RGD-boron cluster conjugates to develop tumor-selective boron carriers for boron neutron capture therapy. Bioorg Med Chem 2011; 19:1721-8. [DOI: 10.1016/j.bmc.2011.01.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 01/11/2011] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
|
45
|
Abstract
Targeted delivery of chemotherapeutics is defined in the sense, that is, to maximize the therapeutic index of a chemotherapeutic agent by strictly localizing its pharmacological activity to the site or tissue of action. Integrins are a family of heterodimeric transmembrane glycoproteins involved in a wide range of cell-to-extracellular matrix (ECM) and cell-to-cell interactions. As cell surface receptors, integrins readily interact with extracellular ligands and play a vital role in angiogenesis, leukocytes function and tumor development, which sets up integrins as an excellent target for chemotherapy treatment. The peptide ligands containing the arginine-glycine-aspartic acid (RGD), which displays a strong binding affinity and selectivity to integrins, particularly to integrin αvβ3, have been developed to conjugate with various conventional chemotherapeutic agents, such as small molecules, peptides and proteins, and nanoparticle-carried drugs for integtrin targeted therapeutic studies. This review highlights the recent advances in integrin targeted delivery of chemotherapeutic agents with emphasis on target of integrin αvβ3, and describes the considerations for the design of the diverse RGD peptide-chemotherapeutics conjugates and their major applications.
Collapse
|
46
|
Sun X, Yan Y, Liu S, Cao Q, Yang M, Neamati N, Shen B, Niu G, Chen X. 18F-FPPRGD2 and 18F-FDG PET of response to Abraxane therapy. J Nucl Med 2010; 52:140-6. [PMID: 21149494 DOI: 10.2967/jnumed.110.080606] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Abraxane (nanoparticle albumin-bound paclitaxel) is an anticancer drug approved by the Food and Drug Administration. However, the mechanism of action of Abraxane is complex, and no established biomarker is available to accurately monitor its treatment outcomes. The aim of this study was to investigate whether the integrin-specific PET tracer 18F-FPPRGD2 (investigational new drug 104150) can be used to monitor early response of tumors to Abraxane therapy. METHODS Orthotopic MDA-MB-435 breast cancer mice were treated with Abraxane (25 mg/kg every other day, 3 doses) or phosphate-buffered saline. Tumor volume was monitored by caliper measurement. PET scans were obtained before and at different times after the start of treatment (days 0, 3, 7, 14, and 21) using 18F-FPPRGD2 and 18F-FDG. The tumoricidal effect was also assessed ex vivo by immunohistochemistry. RESULTS Abraxane treatment inhibited the tumor growth, and a significant difference in tumor volume could be seen at day 5 after the initiation of treatment. The tumor uptake of 18F-FPPRGD2 in the Abraxane-treated group was significantly lower on days 3 and 7 than at baseline but returned to the baseline level at days 14 and 21, indicative of relapse of the tumors after the treatment was halted. Immunohistologic staining confirmed that the change of 18F-FPPRGD2 uptake correlated with the variation of integrin level in the tumor vasculature induced by Abraxane treatment. No significant change of tumor (rather than vascular) integrin expression was observed throughout the study. No significant decrease of 18F-FDG uptake was found between the treated and the control tumors on days 3, 14, and 21, although an increase in 18F-FDG tumor uptake of treated mice, as compared with the control mice, was found on day 7. The increase of 18F-FDG on day 7 was related to the inflammatory response during therapy. CONCLUSION Abraxane-mediated downregulation of integrin αvβ3 expression on tumor endothelial cells can be quantitatively visualized by PET. The change of integrin expression precedes that of tumor size. Consequently, 18F-FPPRGD2 PET is superior to 18F-FDG PET in monitoring early response to treatment, favoring its potential clinical translation.
Collapse
Affiliation(s)
- Xilin Sun
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892-2281, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Leamon CP, Reddy JA, Klein PJ, Vlahov IR, Dorton R, Bloomfield A, Nelson M, Westrick E, Parker N, Bruna K, Vetzel M, Gehrke M, Nicoson JS, Messmann RA, LoRusso PM, Sausville EA. Reducing Undesirable Hepatic Clearance of a Tumor-Targeted Vinca Alkaloid via Novel Saccharopeptidic Modifications. J Pharmacol Exp Ther 2010; 336:336-43. [DOI: 10.1124/jpet.110.175109] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
48
|
Abstract
IMPORTANCE OF THE FIELD Recently, there has been substantial progress in the development of integrin targeted pharmaceuticals and drug delivery systems. Integrin is an important member in the cell adhesion molecule family, which is involved in regulation of complex biological conditions, from keeping normal physiological activities to causing cellular dysfunction in diseased cells. Hence, it is timely to summarize the recent developments in integrin targeted drug and gene delivery systems to understand better their advantages and limitations. AREAS COVERED IN THIS REVIEW In this review, advances in the discovery and clinical trials of these integrin antagonists against different integrin subunits are summarized and discussed. Besides using integrin inhibitor as a single therapeutic agent, integrin antagonists that were conjugated to cytotoxic drugs by synthetic chemistry or coupled to biomacromolecules by either DNA recombination technology or fusion protein technology for integrin targeted therapy have been explored. Furthermore, nanoparticles with integrin targeting ligands for both drug and gene delivery, typically for antiangiogenesis and anticancer therapy, are highlighted and evaluated. WHAT THE READER WILL GAIN This review sheds light on the future development of integrin targeted drug and/or gene delivery systems. TAKE HOME MESSAGE Although thus far there are still limitations, integrin targeted delivery systems have already shown their potential as important pharmaceuticals in the near future.
Collapse
Affiliation(s)
- Zhe Wang
- National University of Singapore, Department of Pharmacy, 18 Science Drive 4, Singapore 117543, Singapore
| | | | | |
Collapse
|
49
|
Xiong XB, Ma Z, Lai R, Lavasanifar A. The therapeutic response to multifunctional polymeric nano-conjugates in the targeted cellular and subcellular delivery of doxorubicin. Biomaterials 2010; 31:757-68. [DOI: 10.1016/j.biomaterials.2009.09.080] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 09/21/2009] [Indexed: 10/20/2022]
|
50
|
Liu R, Wolinsky JB, Walpole J, Southard E, Chirieac LR, Grinstaff MW, Colson YL. Prevention of local tumor recurrence following surgery using low-dose chemotherapeutic polymer films. Ann Surg Oncol 2009; 17:1203-13. [PMID: 19957041 DOI: 10.1245/s10434-009-0856-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Indexed: 01/16/2023]
Abstract
AIM To evaluate the efficacy of a polymer film designed for prolonged paclitaxel release at surgical margins to prevent local recurrence in non-small-cell lung cancer (NSCLC) following complete surgical resection in a murine model. METHODS Poly(glycerol monostearate co-epsilon-caprolactone) polymer films were prepared with or without 10% (w/w) paclitaxel and characterized for prolonged tumor cytotoxicity in vitro against several NSCLC cell lines including LLC, NCI-H460, and NCI-H292. Films were implanted following complete LLC tumor resection and assessed in vivo for prevention of local tumor recurrence, impact on wound healing, and extent of local drug delivery. Plasma and local tissue concentrations of paclitaxel were compared following systemic administration and film implantation. RESULTS The flexible polymeric films eluted paclitaxel over several weeks and remained cytotoxic to LLC, NCI-H460, and NCI-H292 cells in vitro for 50 days, while unloaded films did not impair tumor cell growth. Implanted paclitaxel films prevented local tumor recurrence in vivo in 83.3% of animals, compared with unloaded films (12.5%), systemic (22.2%) or locally administered paclitaxel (0%) (P < 0.005). Although minimal paclitaxel remained in either plasma or tissue 10 days after systemic injection, local paclitaxel concentration at the site of surgical resection was significantly greater (3,000-fold) at 10 days when paclitaxel was locally delivered via films (P = 0.024). CONCLUSIONS Local application of paclitaxel-loaded polymer films following surgical resection can prevent local tumor recurrence without impairing wound healing.
Collapse
Affiliation(s)
- Rong Liu
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|