1
|
Mellid-Carballal R, Gutierrez-Gutierrez S, Rivas C, Garcia-Fuentes M. Viral protein-based nanoparticles (part 2): Pharmaceutical applications. Eur J Pharm Sci 2023; 189:106558. [PMID: 37567394 DOI: 10.1016/j.ejps.2023.106558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/10/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Viral protein nanoparticles (ViP NPs) such as virus-like particles and virosomes are structures halfway between viruses and synthetic nanoparticles. The biological nature of ViP NPs endows them with the biocompatibility, biodegradability, and functional properties that many synthetic nanoparticles lack. At the same time, the absence of a viral genome avoids the safety concerns of viruses. Such characteristics of ViP NPs offer a myriad of opportunities for theirapplication at several points across disease development: from prophylaxis to diagnosis and treatment. ViP NPs present remarkable immunostimulant properties, and thus the vaccination field has benefited the most from these platforms capable of overcoming the limitations of both traditional and subunit vaccines. This was reflected in the marketing authorization of several VLP- and virosome-based vaccines. Besides, ViP NPs inherit the ability of viruses to deliver their cargo to target cells. Because of that, ViP NPs are promising candidates as vectors for drug and gene delivery, and for diagnostic applications. In this review, we analyze the pharmaceutical applications of ViP NPs, describing the products that are commercially available or under clinical evaluation, but also the advances that scientists are making toward the implementation of ViP NPs in other areas of major pharmaceutical interest.
Collapse
Affiliation(s)
- Rocio Mellid-Carballal
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Sara Gutierrez-Gutierrez
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain
| | - Carmen Rivas
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain; Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CNB)-CSIC, Spain
| | - Marcos Garcia-Fuentes
- CiMUS Research Center, Universidad de Santiago de Compostela, Spain; Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidad de Santiago de Compostela, Spain; Health Research Institute of Santiago de Compostela (IDIS), Universidad de Santiago de Compostela, Spain.
| |
Collapse
|
2
|
Nanoparticles for Topical Application in the Treatment of Skin Dysfunctions-An Overview of Dermo-Cosmetic and Dermatological Products. Int J Mol Sci 2022; 23:ijms232415980. [PMID: 36555619 PMCID: PMC9780930 DOI: 10.3390/ijms232415980] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Nanomaterials (NM) arouse interest in various fields of science and industry due to their composition-tunable properties and the ease of modification. They appear currently as components of many consumer products such as sunscreen, dressings, sports clothes, surface-cleaning agents, computer devices, paints, as well as pharmaceutical and cosmetics formulations. The use of NPs in products for topical applications improves the permeation/penetration of the bioactive compounds into deeper layers of the skin, providing a depot effect with sustained drug release and specific cellular and subcellular targeting. Nanocarriers provide advances in dermatology and systemic treatments. Examples are a non-invasive method of vaccination, advanced diagnostic techniques, and transdermal drug delivery. The mechanism of action of NPs, efficiency of skin penetration, and potential threat to human health are still open and not fully explained. This review gives a brief outline of the latest nanotechnology achievements in products used in topical applications to prevent and treat skin diseases. We highlighted aspects such as the penetration of NPs through the skin (influence of physical-chemical properties of NPs, the experimental models for skin penetration, methods applied to improve the penetration of NPs through the skin, and methods applied to investigate the skin penetration by NPs). The review summarizes various therapies using NPs to diagnose and treat skin diseases (melanoma, acne, alopecia, vitiligo, psoriasis) and anti-aging and UV-protectant nano-cosmetics.
Collapse
|
3
|
Oey O, Sunjaya AP. Applications of nanoparticles in cardiovascular imaging and therapeutics. Asian Cardiovasc Thorac Ann 2022; 30:653-660. [PMID: 35259973 DOI: 10.1177/02184923221087003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cardiovascular disease (CVD) is a major health problem afflicting millions of people worldwide. Early detection methods are lacking, and current therapies have significant limitations. Recently, there has been a surge in the number of studies investigating the utilisation of nanoparticles in cardiovascular imaging and therapy. With respect to cardiovascular imaging, previous studies have looked at the role of nanoparticles in thrombus formation, angiogenesis, blood pool and stem cell imaging. Whereas, with respect to therapy, nanoparticles have been studied for delivering drugs and nucleic acids, specifically to the site of interest; in the context of cardiac regeneration; and its potential in refining current therapy guidelines for CVD management. This review aims to extensively summarise the studies that have been conducted investigating the role of nanoparticles in different aspects of cardiovascular imaging and therapy.
Collapse
Affiliation(s)
- Oliver Oey
- 94920St John of God Midland Hospital, Perth, Australia.,85075Faculty of Medicine, University of Western Australia, Perth, Australia
| | - Anthony Paulo Sunjaya
- 98994Faculty of Medicine, University of New South Wales, Sydney, Australia.,98994The George Institute for Global Health, Sydney, Australia
| |
Collapse
|
4
|
Amin M, Mansourian M, Burgers PC, Amin B, Jaafari MR, ten Hagen TLM. Increased Targeting Area in Tumors by Dual-Ligand Modification of Liposomes with RGD and TAT Peptides. Pharmaceutics 2022; 14:pharmaceutics14020458. [PMID: 35214190 PMCID: PMC8878433 DOI: 10.3390/pharmaceutics14020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Modification with polyethylene glycol (PEGylation) and the use of rigid phospholipids drastically improve the pharmacokinetics of chemotherapeutics and result in more manageable or reduced side-effects. A major drawback is retarded cellular delivery of content, which, along with tumor heterogeneity, are the two main obstacles against tumor targeting. To enhance cellular delivery and reach a bigger area of a tumor, we designed liposomes decorated with two ligands: one for targeting tumor vasculature via a cyclic-pentapeptide containing arginine-glycine-aspartic acid (RGD), which impacts tumor independent of passive accumulation inside tumors, and one for extravascular targeting of tumor cells via a cell-penetrating peptide derived from human immunodeficiency virus type 1 transactivator of transcription (TAT). Liposomes with different ligand combinations were prepared and compared with respect to performance in targeting. Intravital imaging illustrates the heterogeneous behavior of RGD-liposomes in both intravascular and extravascular distribution, whereas TAT-liposomes exhibit a predictable extravascular localization but no intravascular targeting. Dual-ligand modification results in enhanced vascular targeting and a predictable extravascular behavior that improves the therapeutic efficacy of doxorubicin-loaded liposomes but also an augmented clearance rate of liposomes. However, the dual-modified liposome could be a great candidate for targeted delivery of non-toxic payloads or contrast agents for therapeutic or diagnostic purposes. Here we show that the combination of vascular-specific and tumor cell-specific ligands in a liposomal system is beneficial in bypassing the heterogeneous expression of tumor-specific markers.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| | - Mercedeh Mansourian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran; (M.M.); (M.R.J.)
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Peter C. Burgers
- Laboratory of Neuro-Oncology, Department of Neurology, Erasmus MC University Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands;
| | - Bahareh Amin
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar 9613873136, Iran;
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9196773117, Iran; (M.M.); (M.R.J.)
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| | - Timo L. M. ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands;
| |
Collapse
|
5
|
Kalinina A, Golubeva I, Kudryavtsev I, Khromova N, Antoshina E, Trukhanova L, Gorkova T, Kazansky D, Khromykh L. Cyclophilin A is a factor of antitumor defense in the early stages of tumor development. Int Immunopharmacol 2021; 94:107470. [PMID: 33640856 DOI: 10.1016/j.intimp.2021.107470] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/14/2021] [Accepted: 02/02/2021] [Indexed: 01/12/2023]
Abstract
Cyclophilin A (CypA) is a pro-inflammatory factor with multiple immunomodulating effects. Here, we investigated the effects of recombinant human CypA (rhCypA) as a factor of antitumor host defense. Our results demonstrated that rhCypA dramatically inhibited the growth of murine transplantable tumors (mammary adenocarcinoma Ca755, melanoma B16, Lewis lung carcinoma (LLC), and cervical cancer CC-5). In the B16 model, rhCypA effects were observed only when tumor cells were transplanted at the significantly reduced injection dose, indicating that antitumor properties of rhCypA are more effective at the initial stages of cancer development. Antitumor effect of rhCypA in the CC-5 model was comparable to the action of 5-fluorouracil (5FU), and rhCypA administration prevented 5FU - induced leukopenia in the blood of tumor-bearing mice. In the LLC model, rhCypA injection before but not after tumor resection significantly suppressed the formation of post-surgical metastases. RhCypA exhibited no direct cytotoxic effects in vitro on human leukemia cells (K-562, HL-60, KG-1), indicating that rhCypA antitumor action could be mediated by its immunomodulating activity. In the B16 model, rhCypA had no impact on tumor angiogenesis and gene expression of several MMPs, endogenous CypA, and CD147, which play a crucial role in cancer progression. However, in this model, rhCypA stimulated gene expression of MMPs 8, 9, and 12 that could contribute to malignancy growth inhibition. Here, our findings pointed out CypA as one of the factors of antitumor host defense that can effectively control the initial stages of tumor and metastases formation by regulating the action of MMPs and changing the tumor microenvironment.
Collapse
Affiliation(s)
- Anastasiia Kalinina
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Irina Golubeva
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Igor Kudryavtsev
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Natalia Khromova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Elena Antoshina
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Lubov Trukhanova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Tatyana Gorkova
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Dmitry Kazansky
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation
| | - Ludmila Khromykh
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology" of the Ministry of Health of the Russian Federation, Kashirskoe sh. 24, Moscow 115478, Russian Federation.
| |
Collapse
|
6
|
Abstract
Melanoma is an aggressive form of skin cancer with a very high mortality rate. Early diagnosis of the disease, the utilization of more potent pharmacological agents, and more effective drug delivery systems are essential to achieve an optimal treatment plan. The applications of nanotechnology to improve therapeutic efficacy and early diagnosis for melanoma treatment have received great interest among researchers and clinicians. In this review, we summarize the recent progress of utilizing various nanomaterials for theranostics of melanoma. The key importance of using nanomaterials for theranostics of melanoma is to improve efficacy and reduce side effects, ensuring safe implementation in clinical use. As opposed to conventional in vitro diagnostic methods, in vivo medical imaging technologies have the advantages of being a type of non-invasive, real-time monitoring. Several common nanoparticles, including ultrasmall superparamagnetic iron oxide nanoparticles, silica nanoparticles, and carbon-based nanoparticles, have been applied to deliver chemotherapeutic agents for the theranostics of melanoma. The application of nanomaterials for theranostics in molecular imaging (MRI, PET, US, OI, etc.) plays an important role in targeting drug delivery of melanoma, by monitoring the distribution site of the molecular imaging probe and the therapeutic drug in the body in real-time. Hence, it is worthwhile to anticipate the approval of these nanomaterials for theranostics in molecular imaging by the US Food and Drug Administration in clinical trials.
Collapse
|
7
|
Li Y, Chen R, Li Z, Cheng H, Li X, Li T, Zhu C. miR-204 Negatively Regulates Cell Growth And Metastasis By Targeting ROBO4 In Human Bladder Cancer. Onco Targets Ther 2019; 12:8515-8524. [PMID: 31802889 PMCID: PMC6801631 DOI: 10.2147/ott.s205023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 09/20/2019] [Indexed: 01/16/2023] Open
Abstract
Background MicroRNAs (miRNAs) are well characterized for their important roles in human cancers by influencing various aspects of malignancy. Till now, the function and mechanism of miR-204, a tumor suppressor in several cancers, remain unclear in bladder cancer (BC). Here, we intend to explore its roles in BC progression. Methods qRT-PCR was applied to determine miR-204 and ROBO4 expression in BC tissues and cell lines. miR-204 expression with clinicopathological features was analyzed. The impacts of miR-204 on BC cell growth and metastasis in vitro were evaluated by both loss-of-function and gain-of-function assays (CCK-8, crystal violet staining, wound healing and transwell assays). Furthermore, qRT-PCR, Western blot and luciferase reporter assays were used to validate the targeting of ROBO4 by miR-204. Finally, linear regression was performed to analyze the correlation of miR-204 and ROBO4 in BC tissues. Results Expression of miR-204 was markedly decreased in BC tissues and cell lines were compared with respective controls. Low miR-204 expression was associated with positive advanced T stage and lymph node metastasis. Cellular function studies revealed that miR-204 inhibited BC cell growth, migration and invasion. Mechanistic exploration found that miR-204 directly targeted ROBO4. Rescue assays indicated that ROBO4 restoration could reverse the antitumor effects of miR-204 in BC. Finally, ROBO4 was significantly correlated with miR-204 levels inversely. Conclusion miR-204 might serve as a tumor suppressor in BC by targeting ROBO4.
Collapse
Affiliation(s)
- Yang Li
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| | - Rong Chen
- Department of Urology, Zhuji People's Hospital of Zhejiang Province, Zhuji 311800, China
| | - Zun Li
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| | - Hepeng Cheng
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| | - Xiaodong Li
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| | - Tieqiang Li
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| | - Chaoyang Zhu
- Department of Urology, Henan University Huaihe Hospital, Kaifeng 475000, People's Republic of China
| |
Collapse
|
8
|
Gao X, Guo L, Li J, Thu HE, Hussain Z. Nanomedicines guided nanoimaging probes and nanotherapeutics for early detection of lung cancer and abolishing pulmonary metastasis: Critical appraisal of newer developments and challenges to clinical transition. J Control Release 2018; 292:29-57. [PMID: 30359665 DOI: 10.1016/j.jconrel.2018.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer (LC) is the second most prevalent type of cancer and primary cause of mortality among both men and women, worldwide. The most commonly employed diagnostic modalities for LC include chest X-ray (CXR), magnetic-resonance-imaging (MRI), computed tomography (CT-scan), and fused-positron-emitting-tomography-CT (PET-CT). Owing to several limitations associated with the use of conventional diagnostic tools such as radiation burden to the patient, misleading diagnosis ("missed lung cancer"), false staging and low sensitivity and resolution, contemporary diagnostic regimen needed to be employed for screening of LC. In recent decades, nanotechnology-guided interventions have been transpired as emerging nanoimaging probes for detection of LC at advanced stages, while producing signal amplification, better resolution for surface and deep tissue imaging, and enhanced translocation and biodistribution of imaging probes within the cancerous tissues. Besides enormous potential of nanoimaging probes, nanotechnology-based advancements have also been evidenced for superior efficacy for treatment of LC and abolishing pulmonary metastasis (PM). The success of nanotherapeutics is due to their ability to maximise translocation and biodistribution of anti-neoplastic agents into the tumor tissues, improve pharmacokinetic profiles of anti-metastatic agents, optimise target-specific drug delivery, and control release kinetics of encapsulated moieties in target tissues. This review aims to overview and critically discuss the superiority of nanoimaging probes and nanotherapeutics over conventional regimen for early detection of LC and abolishing PM. Current challenges to clinical transition of nanoimaging probes and therapeutic viability of nanotherapeutics for treatment for LC and PM have also been pondered.
Collapse
Affiliation(s)
- Xiaoling Gao
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Lihua Guo
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jianqiang Li
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Hnin Ei Thu
- Department of Pharmacology and Dental Therapeutics, Faculty of Dentistry, Lincoln University College, Jalan Stadium, SS 7/15, Kelana Jaya, 47301 Petaling Jaya, Selangor, Malaysia
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
9
|
Atukorale PU, Covarrubias G, Bauer L, Karathanasis E. Vascular targeting of nanoparticles for molecular imaging of diseased endothelium. Adv Drug Deliv Rev 2017; 113:141-156. [PMID: 27639317 DOI: 10.1016/j.addr.2016.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/08/2023]
Abstract
This review seeks to highlight the enormous potential of targeted nanoparticles for molecular imaging applications. Being the closest point-of-contact, circulating nanoparticles can gain direct access to targetable molecular markers of disease that appear on the endothelium. Further, nanoparticles are ideally suitable to vascular targeting due to geometrically enhanced multivalent attachment on the vascular target. This natural synergy between nanoparticles, vascular targeting and molecular imaging can provide new avenues for diagnosis and prognosis of disease with quantitative precision. In addition to the obvious applications of targeting molecular signatures of vascular diseases (e.g., atherosclerosis), deep-tissue diseases often manifest themselves by continuously altering and remodeling their neighboring blood vessels (e.g., cancer). Thus, the remodeled endothelium provides a wide range of targets for nanoparticles and molecular imaging. To demonstrate the potential of molecular imaging, we present a variety of nanoparticles designed for molecular imaging of cancer or atherosclerosis using different imaging modalities.
Collapse
|
10
|
Ma X, Phi Van V, Kimm MA, Prakash J, Kessler H, Kosanke K, Feuchtinger A, Aichler M, Gupta A, Rummeny EJ, Eisenblätter M, Siveke J, Walch AK, Braren R, Ntziachristos V, Wildgruber M. Integrin-Targeted Hybrid Fluorescence Molecular Tomography/X-ray Computed Tomography for Imaging Tumor Progression and Early Response in Non-Small Cell Lung Cancer. Neoplasia 2017; 19:8-16. [PMID: 27940248 PMCID: PMC5157790 DOI: 10.1016/j.neo.2016.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 11/05/2022]
Abstract
Integrins play an important role in tumor progression, invasion and metastasis. Therefore we aimed to evaluate a preclinical imaging approach applying ανβ3 integrin targeted hybrid Fluorescence Molecular Tomography/X-ray Computed Tomography (FMT-XCT) for monitoring tumor progression as well as early therapy response in a syngeneic murine Non-Small Cell Lung Cancer (NSCLC) model. Lewis Lung Carcinomas were grown orthotopically in C57BL/6 J mice and imaged in-vivo using a ανβ3 targeted near-infrared fluorescence (NIRF) probe. ανβ3-targeted FMT-XCT was able to track tumor progression. Cilengitide was able to substantially block the binding of the NIRF probe and suppress the imaging signal. Additionally mice were treated with an established chemotherapy regimen of Cisplatin and Bevacizumab or with a novel MEK inhibitor (Refametinib) for 2 weeks. While μCT revealed only a moderate slowdown of tumor growth, ανβ3 dependent signal decreased significantly compared to non-treated mice already at one week post treatment. ανβ3 targeted imaging might therefore become a promising tool for assessment of early therapy response in the future.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Carcinoma, Lewis Lung/diagnosis
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/diagnosis
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Cell Line, Tumor
- Disease Models, Animal
- Disease Progression
- Fluorescence
- Gene Expression
- Humans
- Integrin alphaVbeta3/genetics
- Integrin alphaVbeta3/metabolism
- Integrins/genetics
- Integrins/metabolism
- Lung Neoplasms/diagnosis
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Molecular Imaging
- Protein Kinase Inhibitors/pharmacology
- Tomography
- Tomography, X-Ray Computed
- Treatment Outcome
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xiaopeng Ma
- Institute for Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Valerie Phi Van
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Melanie A Kimm
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Jaya Prakash
- Institute for Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Horst Kessler
- Chemistry Department and TUM Institute for Advanced Study, Lichtenbergstrasse 2a, D-85748, Garching, Germany
| | - Katja Kosanke
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Aayush Gupta
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Ernst J Rummeny
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Michel Eisenblätter
- Department of Clinical Radiology, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany
| | - Jens Siveke
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK), partner site Essen, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Axel K Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Rickmer Braren
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, D-85764 Oberschleissheim, Germany
| | - Moritz Wildgruber
- Department of Radiology, Klinikum Rechts der Isar, Technische Universität München, Ismaningerstrasse 22, D-81675, München, Germany; Department of Clinical Radiology, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, D-48149, Münster, Germany.
| |
Collapse
|
11
|
Jallouk AP, Palekar RU, Marsh JN, Pan H, Pham CTN, Schlesinger PH, Wickline SA. Delivery of a Protease-Activated Cytolytic Peptide Prodrug by Perfluorocarbon Nanoparticles. Bioconjug Chem 2015; 26:1640-50. [PMID: 26083278 DOI: 10.1021/acs.bioconjchem.5b00246] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Melittin is a cytolytic peptide derived from bee venom that inserts into lipid membranes and oligomerizes to form membrane pores. Although this peptide is an attractive candidate for treatment of cancers and infectious processes, its nonspecific cytotoxicity and hemolytic activity have limited its therapeutic applications. Several groups have reported the development of cytolytic peptide prodrugs that only exhibit cytotoxicity following activation by site-specific proteases. However, systemic administration of these constructs has proven difficult because of their poor pharmacokinetic properties. Here, we present a platform for the design of protease-activated melittin derivatives that may be used in conjunction with a perfluorocarbon nanoparticle delivery system. Although native melittin was substantially hemolytic (HD50: 1.9 μM) and cytotoxic (IC50: 2.4 μM), the prodrug exhibited 2 orders of magnitude less hemolytic activity (HD50: > 100 μM) and cytotoxicity (IC50: > 100 μM). Incubation with matrix metalloproteinase-9 (MMP-9) led to cleavage of the prodrug at the expected site and restoration of hemolytic activity (HD50: 3.4 μM) and cytotoxicity (IC50: 8.1 μM). Incubation of the prodrug with perfluorocarbon nanoparticles led to stable loading of 10,250 peptides per nanoparticle. Nanoparticle-bound prodrug was also cleaved and activated by MMP-9, albeit at a fourfold slower rate. Intravenous administration of prodrug-loaded nanoparticles in a mouse model of melanoma significantly decreased tumor growth rate (p = 0.01). Because MMPs and other proteases play a key role in cancer invasion and metastasis, this platform holds promise for the development of personalized cancer therapies directed toward a patient's individual protease expression profile.
Collapse
Affiliation(s)
- Andrew P Jallouk
- †Consortium for Translational Research in Advanced Imaging and Nanomedicine, Division of Cardiology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63108, United States
| | - Rohun U Palekar
- †Consortium for Translational Research in Advanced Imaging and Nanomedicine, Division of Cardiology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63108, United States
| | - Jon N Marsh
- †Consortium for Translational Research in Advanced Imaging and Nanomedicine, Division of Cardiology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63108, United States
| | - Hua Pan
- †Consortium for Translational Research in Advanced Imaging and Nanomedicine, Division of Cardiology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63108, United States
| | | | | | - Samuel A Wickline
- †Consortium for Translational Research in Advanced Imaging and Nanomedicine, Division of Cardiology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63108, United States
| |
Collapse
|
12
|
Abstract
Nanotechnology has the potential to increase the selectivity and potency of chemical, physical, and biological approaches for eliciting cancer cell death while minimizing collateral toxicity to nonmalignant cells. Materials on the nanoscale are increasingly being targeted to cancer cells with great specificity through both active and passive targeting. In this review, we summarize recent literature that has broken new ground in the use of nanotechnology for cancer treatment with an emphasis on targeted drug delivery.
Collapse
Affiliation(s)
- William H. Gmeiner
- Corresponding author: William H. Gmeiner, Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 21757, USA,
| | | |
Collapse
|
13
|
Ermolayev V, Mohajerani P, Ale A, Sarantopoulos A, Aichler M, Kayser G, Walch A, Ntziachristos V. Early recognition of lung cancer by integrin targeted imaging in K-ras mouse model. Int J Cancer 2015; 137:1107-18. [DOI: 10.1002/ijc.29372] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 11/03/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Vladimir Ermolayev
- Institute for Biological and Medical Imaging; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Pouyan Mohajerani
- Institute for Biological and Medical Imaging; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Angelique Ale
- Institute for Biological and Medical Imaging; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Athanasios Sarantopoulos
- Institute for Biological and Medical Imaging; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology-Institute of Pathology; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Gian Kayser
- Institute of Pathology; Universitätsklinkum Freiburg; Freiburg im Breisgau Germany
| | - Axel Walch
- Research Unit Analytical Pathology-Institute of Pathology; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging; Helmholtz Zentrum Munich, Ingolstaedter Landstrasse 1 D-85764 Neuherberg Germany
- Techniche Universitaet Muenchen; Chair for Biological Imaging; Arcisstrasse 21, D-80333 Munich
| |
Collapse
|
14
|
Zhuang X, Ahmed F, Zhang Y, Ferguson HJ, Steele JC, Steven NM, Nagy Z, Heath VL, Toellner KM, Bicknell R. Robo4 vaccines induce antibodies that retard tumor growth. Angiogenesis 2014; 18:83-95. [PMID: 25348086 DOI: 10.1007/s10456-014-9448-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 10/13/2014] [Indexed: 01/24/2023]
Abstract
Tumor endothelial specific expression of Robo4 in adults identifies this plasma membrane protein as an anti-cancer target for immunotherapeutic approaches, such as vaccination. In this report, we describe how vaccination against Robo4 inhibits angiogenesis and tumor growth. To break tolerance to the auto-antigen Robo4, mice were immunised with the extracellular domain of mouse Robo4, fused to the Fc domain of human immunoglobulin within an adjuvant. Vaccinated mice show a strong antibody response to Robo4, with no objectively detectable adverse effects on health. Robo4 vaccinated mice showed impaired fibrovascular invasion and angiogenesis in a rodent sponge implantation assay, as well as a reduced growth of implanted syngeneic Lewis lung carcinoma. The anti-tumor effect of Robo4 vaccination was present in CD8 deficient mice but absent in B cell or IgG1 knockout mice, suggesting antibody dependent cell mediated cytotoxicity as the anti-vascular/anti-tumor mechanism. Finally, we show that an adjuvant free soluble Robo4-carrier conjugate can retard tumor growth in carrier primed mice. These results point to appropriate Robo4 conjugates as potential anti-angiogenic vaccines for cancer patients.
Collapse
Affiliation(s)
- Xiaodong Zhuang
- Institute for Biomedical Research, Schools of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Toy R, Bauer L, Hoimes C, Ghaghada KB, Karathanasis E. Targeted nanotechnology for cancer imaging. Adv Drug Deliv Rev 2014; 76:79-97. [PMID: 25116445 PMCID: PMC4169743 DOI: 10.1016/j.addr.2014.08.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 07/26/2014] [Accepted: 08/04/2014] [Indexed: 02/02/2023]
Abstract
Targeted nanoparticle imaging agents provide many benefits and new opportunities to facilitate accurate diagnosis of cancer and significantly impact patient outcome. Due to the highly engineerable nature of nanotechnology, targeted nanoparticles exhibit significant advantages including increased contrast sensitivity, binding avidity and targeting specificity. Considering the various nanoparticle designs and their adjustable ability to target a specific site and generate detectable signals, nanoparticles can be optimally designed in terms of biophysical interactions (i.e., intravascular and interstitial transport) and biochemical interactions (i.e., targeting avidity towards cancer-related biomarkers) for site-specific detection of very distinct microenvironments. This review seeks to illustrate that the design of a nanoparticle dictates its in vivo journey and targeting of hard-to-reach cancer sites, facilitating early and accurate diagnosis and interrogation of the most aggressive forms of cancer. We will report various targeted nanoparticles for cancer imaging using X-ray computed tomography, ultrasound, magnetic resonance imaging, nuclear imaging and optical imaging. Finally, to realize the full potential of targeted nanotechnology for cancer imaging, we will describe the challenges and opportunities for the clinical translation and widespread adaptation of targeted nanoparticles imaging agents.
Collapse
Affiliation(s)
- Randall Toy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lisa Bauer
- Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Physics, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Christopher Hoimes
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; University Hospitals Case Medical Center, Cleveland, OH 44106, USA
| | - Ketan B Ghaghada
- Edward B. Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX 77030, USA; Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; Case Center for Imaging Research, Case Western Reserve University, Cleveland, OH 44106, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Radiology, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
16
|
Winter P. Molecular Imaging at Nanoscale with Magnetic Resonance Imaging. Nanomedicine (Lond) 2014. [DOI: 10.1201/b17246-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
17
|
Winter PM. Perfluorocarbon nanoparticles: evolution of a multimodality and multifunctional imaging agent. SCIENTIFICA 2014; 2014:746574. [PMID: 25024867 PMCID: PMC4082945 DOI: 10.1155/2014/746574] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 05/20/2014] [Indexed: 06/03/2023]
Abstract
Perfluorocarbon nanoparticles offer a biologically inert, highly stable, and nontoxic platform that can be specifically designed to accomplish a range of molecular imaging and drug delivery functions in vivo. The particle surface can be decorated with targeting ligands to direct the agent to a variety of biomarkers that are associated with diseases such as cancer, cardiovascular disease, obesity, and thrombosis. The surface can also carry a high payload of imaging agents, ranging from paramagnetic metals for MRI, radionuclides for nuclear imaging, iodine for CT, and florescent tags for histology, allowing high sensitivity mapping of cellular receptors that may be expressed at very low levels in the body. In addition to these diagnostic imaging applications, the particles can be engineered to carry highly potent drugs and specifically deposit them into cell populations that display biosignatures of a variety of diseases. The highly flexible and robust nature of this combined molecular imaging and drug delivery vehicle has been exploited in a variety of animal models to demonstrate its potential impact on the care and treatment of patients suffering from some of the most debilitating diseases.
Collapse
Affiliation(s)
- Patrick M. Winter
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| |
Collapse
|
18
|
Winter PM, Pearce J, Chu Z, McPherson CM, Takigiku R, Lee JH, Qi X. Imaging of brain tumors with paramagnetic vesicles targeted to phosphatidylserine. J Magn Reson Imaging 2014; 41:1079-87. [PMID: 24797437 DOI: 10.1002/jmri.24654] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/14/2014] [Accepted: 04/17/2014] [Indexed: 01/06/2023] Open
Abstract
PURPOSE To investigate paramagnetic saposin C and dioleylphosphatidylserine (SapC-DOPS) vesicles as a targeted contrast agent for imaging phosphatidylserine (PS) expressed by glioblastoma multiforme (GBM) tumors. MATERIALS AND METHODS Gd-DTPA-BSA/SapC-DOPS vesicles were formulated, and the vesicle diameter and relaxivity were measured. Targeting of Gd-DTPA-BSA/SapC-DOPS vesicles to tumor cells in vitro and in vivo was compared with nontargeted paramagnetic vesicles (lacking SapC). Mice with GBM brain tumors were imaged at 3, 10, 20, and 24 h postinjection to measure the relaxation rate (R1) in the tumor and the normal brain. RESULTS The mean diameter of vesicles was 175 nm, and the relaxivity at 7 Tesla was 3.32 (s*mM)(-1) relative to the gadolinium concentration. Gd-DTPA-BSA/SapC-DOPS vesicles targeted cultured cancer cells, leading to an increased R1 and gadolinium level in the cells. In vivo, Gd-DTPA-BSA/SapC-DOPS vesicles produced a 9% increase in the R1 of GBM brain tumors in mice 10 h postinjection, but only minimal changes (1.2% increase) in the normal brain. Nontargeted paramagnetic vesicles yielded minimal change in the tumor R1 at 10 h postinjection (1.3%). CONCLUSION These experiments demonstrate that Gd-DTPA-BSA/SapC-DOPS vesicles can selectively target implanted brain tumors in vivo, providing noninvasive mapping of the cancer biomarker PS.
Collapse
Affiliation(s)
- Patrick M Winter
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Role of ROBO4 signalling in developmental and pathological angiogenesis. BIOMED RESEARCH INTERNATIONAL 2014; 2014:683025. [PMID: 24689049 PMCID: PMC3933320 DOI: 10.1155/2014/683025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 11/29/2013] [Accepted: 12/12/2013] [Indexed: 01/13/2023]
Abstract
Transmembrane roundabout receptor family members (ROBO1-ROBO4) principally orchestrate the neuronal guidance mechanism of the nervous system. Secreted glycoprotein SLITs are the most appreciated ligands for ROBOs. Recently identified ROBO4 is the key mediator of SLIT-ROBO mediated developmental and pathological angiogenesis. Although SLIT2 has been shown to interact with ROBO4 as ligand, it remains an open question whether this protein is the physiologic partner of ROBO4. The purpose of this review is to summarise how reliable SLIT2 as ligand for ROBO4 is, if not what the other possible mechanisms demonstrated till date for ROBO4 mediated developmental and pathological angiogenesis are. We conclude that ROBO4 is expressed specially in vascular endothelial cells and maintains the vascular integrity via either SLIT2 dependent or SLIT2 independent manner. On the contrary, it promotes the pathological angiogenesis by involving different signalling arm(s)/unknown ligand(s). This review explores the interactions SLIT2/ROBO1, SLIT2/ROBO1-ROBO4, ROBO1/ROBO4, and ROBO4/UNC5B which can be promising and potential therapeutic targets for developmental angiogenesis defects and pathological angiogenesis. Finally we have reviewed the ROBO4 signalling pathways and made an effort to elaborate the insight of this signalling as therapeutic target of pathological angiogenesis.
Collapse
|
20
|
Hussain T, Nguyen QT. Molecular imaging for cancer diagnosis and surgery. Adv Drug Deliv Rev 2014; 66:90-100. [PMID: 24064465 DOI: 10.1016/j.addr.2013.09.007] [Citation(s) in RCA: 216] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/07/2013] [Accepted: 09/13/2013] [Indexed: 12/11/2022]
Abstract
Novel molecular imaging techniques have the potential to significantly enhance the diagnostic and therapeutic approaches for cancer treatment. For solid tumors in particular, novel molecular enhancers for imaging modalities such as US, CT, MRI and PET may facilitate earlier and more accurate diagnosis and staging which are prerequisites for successful surgical therapy. Enzymatically activatable "smart" molecular MRI probes seem particularly promising because of their potential to image tumors before and after surgical removal without re-administration of the probe to evaluate completeness of surgical resection. Furthermore, the use of "smart" MR probes as part of screening programs may enable detection of small tumors throughout the body in at-risk patient populations. Dual labeling of molecular MR probes with fluorescent dyes can add real time intraoperative guidance facilitating complete tumor resection and preservation of important structures. A truly theranostic approach with the further addition of therapeutic agents to the molecular probe for adjuvant therapy is conceivable for the future.
Collapse
|
21
|
Teng FF, Meng X, Sun XD, Yu JM. New strategy for monitoring targeted therapy: molecular imaging. Int J Nanomedicine 2013; 8:3703-13. [PMID: 24124361 PMCID: PMC3794840 DOI: 10.2147/ijn.s51264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Targeted therapy is becoming an increasingly important component in the treatment of cancer. How to accurately monitor targeted therapy has been crucial in clinical practice. The traditional approach to monitor treatment through imaging has relied on assessing the change of tumor size by refined World Health Organization criteria, or more recently, by the Response Evaluation Criteria in Solid Tumors. However, these criteria, which are based on the change of tumor size, show some limitations for evaluating targeted therapy. Currently, genetic alterations are identified with prognostic as well as predictive potential concerning the use of molecularly targeted drugs. Conversely, considering the limitations of invasiveness and the issue of expression heterogeneity, molecular imaging is better able to assay in vivo biologic processes noninvasively and quantitatively, and has been a particularly attractive tool for monitoring treatment in clinical cancer practice. This review focuses on the applications of different kinds of molecular imaging including positron emission tomography-, magnetic resonance imaging-, ultrasonography-, and computed tomography-based imaging strategies on monitoring targeted therapy. In addition, the key challenges of molecular imaging are addressed to successfully translate these promising techniques in the future.
Collapse
Affiliation(s)
- Fei-Fei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, People's Republic of China
| | | | | | | |
Collapse
|
22
|
Xue S, Qiao J, Pu F, Cameron M, Yang JJ. Design of a novel class of protein-based magnetic resonance imaging contrast agents for the molecular imaging of cancer biomarkers. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2013; 5:163-79. [PMID: 23335551 DOI: 10.1002/wnan.1205] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Magnetic resonance imaging (MRI) of disease biomarkers, especially cancer biomarkers, could potentially improve our understanding of the disease and drug activity during preclinical and clinical drug treatment and patient stratification. MRI contrast agents with high relaxivity and targeting capability to tumor biomarkers are highly required. Extensive work has been done to develop MRI contrast agents. However, only a few limited literatures report that protein residues can function as ligands to bind Gd(3+) with high binding affinity, selectivity, and relaxivity. In this paper, we focus on reporting our current progress on designing a novel class of protein-based Gd(3+) MRI contrast agents (ProCAs) equipped with several desirable capabilities for in vivo application of MRI of tumor biomarkers. We will first discuss our strategy for improving the relaxivity by a novel protein-based design. We then discuss the effect of increased relaxivity of ProCAs on improving the detection limits for MRI contrast agent, especially for in vivo application. We will further report our efforts to improve in vivo imaging capability and our achievement in molecular imaging of cancer biomarkers with potential preclinical and clinical applications.
Collapse
Affiliation(s)
- Shenghui Xue
- Departments of Chemistry and Biology, Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
23
|
Yan C, Wu Y, Feng J, Chen W, Liu X, Hao P, Yang R, Zhang J, Lin B, Xu Y, Liu R. Anti-αvβ3 antibody guided three-step pretargeting approach using magnetoliposomes for molecular magnetic resonance imaging of breast cancer angiogenesis. Int J Nanomedicine 2013; 8:245-55. [PMID: 23345972 PMCID: PMC3548418 DOI: 10.2147/ijn.s38678] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Pretargeting of biomarkers with nanoparticles in molecular imaging is promising to improve diagnostic specificity and realize signal amplification, but data regarding its targeting potential in magnetic resonance (MR) imaging are limited. The purpose of this study was to evaluate the tumor angiogenesis targeting efficacy of the anti-αvβ3 antibody guided three-step pretargeting approach with magnetoliposomes. METHODS Polyethylene glycol-modified and superparamagnetic iron oxide-encapsulated magnetoliposomes with and without biotin were synthesized and characterized. The cytotoxicity of both probes was evaluated using the methyl thiazdyl tetrazolium assay, and their cellular uptake by mouse macrophage was visualized using Prussian blue staining. Three-step pretargeting MR imaging was performed on MDA-MB-435S breast cancer-bearing mice by intravenous administration of biotinylated anti-αvβ3 monoclonal antibodies (first step), followed by avidin and streptavidin (second step), and by biotinylated magnetoliposomes or magnetoliposomes in the targeted or nontargeted group, respectively (third step). The specificity of αvβ3 targeting was assessed by histologic examinations. RESULTS The developed magnetoliposomes were superparamagnetic and biocompatible as confirmed by cell toxicity assay. The liposomal bilayer and polyethylene glycol modification protected Fe(3)O(4) cores from uptake by macrophage cells. MR imaging by three-step pretargeting resulted in a greater signal enhancement along the tumor periphery, occupying 7.0% of the tumor area, compared with 2.0% enhancement of the nontargeted group (P < 0.05). Histologic analysis demonstrated the targeted magnetoliposomes colocalized with neovasculature, which was responsible for the MR signal decrease. CONCLUSION These results indicate that our strategy for MR imaging of αvβ3-integrin is an effective means for sensitive detection of tumor angiogenesis, and may provide a targetable nanodelivery system for anticancer drugs.
Collapse
Affiliation(s)
- Chenggong Yan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kircher MF, Willmann JK. Molecular body imaging: MR imaging, CT, and US. Part II. Applications. Radiology 2012; 264:349-68. [PMID: 22821695 DOI: 10.1148/radiol.12111703] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Molecular imaging is expected to have a major impact on the early diagnosis of diseases and disease monitoring in the next decade. Traditionally, nuclear imaging techniques have been the mainstay of molecular imaging in the clinical arena. However, with continued development of molecularly targeted contrast agents for nonnuclear imaging techniques such as magnetic resonance (MR), computed tomography (CT), and ultrasonography (US), the spectrum of clinical molecular imaging applications is expanding. In the second part of this review series, an overview of applications of molecular MR imaging-, CT-, and US-based imaging strategies that show promise for clinical translation is presented, and key challenges that need to be addressed to successfully translate these promising techniques in the future are discussed. © RSNA, 2012.
Collapse
Affiliation(s)
- Moritz F Kircher
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
25
|
Zhou HF, Yan H, Senpan A, Wickline SA, Pan D, Lanza GM, Pham CTN. Suppression of inflammation in a mouse model of rheumatoid arthritis using targeted lipase-labile fumagillin prodrug nanoparticles. Biomaterials 2012; 33:8632-40. [PMID: 22922023 DOI: 10.1016/j.biomaterials.2012.08.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 08/01/2012] [Indexed: 12/22/2022]
Abstract
Nanoparticle-based therapeutics are emerging technologies that have the potential to greatly impact the treatment of many human diseases. However, drug instability and premature release from the nanoparticles during circulation currently preclude clinical translation. Herein, we use a lipase-labile (Sn 2) fumagillin prodrug platform coupled with a unique lipid surface-to-surface targeted delivery mechanism, termed contact-facilitated drug delivery, to counter the premature drug release and overcome the inherent photo-instability of fumagillin, an established anti-angiogenic agent. We show that α(v)β(3)-integrin targeted fumagillin prodrug nanoparticles, administered at 0.3 mg of fumagillin prodrug/kg of body weight suppress the clinical disease indices of KRN serum-mediated arthritis in a dose-dependent manner when compared to treatment with the control nanoparticles with no drug. This study demonstrates the effectiveness of this lipase-labile prodrug nanocarrier in a relevant preclinical model that approximates human rheumatoid arthritis. The lipase-labile prodrug paradigm offers a translatable approach that is broadly applicable to many targeted nanosystems and increases the translational potential of this platform for many diseases.
Collapse
Affiliation(s)
- Hui-Fang Zhou
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, Box 8045, St. Louis, MO 63110, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Chen WT, Shih TTF, Chen RC, Tu SY, Wen-Yuen H, Yang PC. Integrin α vβ 3–Targeted Dynamic Contrast–Enhanced Magnetic Resonance Imaging Using a Gadolinium-Loaded Polyethylene Gycol–Dendrimer–Cyclic RGD Conjugate to Evaluate Tumor Angiogenesis and to Assess Early Antiangiogenic Treatment Response in a Mouse Xenograft Tumor Model. Mol Imaging 2012; 11:7290.2011.00050. [DOI: 10.2310/7290.2011.00050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
The purpose of this study was to validate an integrin αvβ3–targeted magnetic resonance contrast agent, PEG-G3-(Gd-DTPA)6-(cRGD-DTPA)2, for its ability to detect tumor angiogenesis and assess early response to antiangiogenic therapy using dynamic contrast–enhanced (DCE) magnetic resonance imaging (MRI). Integrin αvβ3–positive U87 cells and control groups were incubated with fluorescein-labeled cRGD-conjugated dendrimer, and the cellular attachment of the dendrimer was observed. DCE MRI was performed on mice bearing KB xenograft tumors using either PEG-G3-(Gd-DTPA)6-(cRGD-DTPA)2 or PEG-G3-(Gd-DTPA)6-(cRAD-DTPA)2. DCE MRI was also performed 2 hours after anti–integrin αvβ3 monoclonal antibody treatment and after bevacizumab treatment on days 3 and 6t. Using DCE MRI, the 30-minute contrast washout percentage was significantly lower in the cRGD-conjugate injection groups. The enhancement patterns were different between the two contrast injection groups. In the antiangiogenic therapy groups, a rapid increase in 30-minute contrast washout percentage was observed in both the LM609 and bevacizumab treatment groups, and this occurred before there was an observable decrease in tumor size. The integrin αvβ3 targeting ability of PEG-G3-(Gd-DTPA)6-(cRGD-DTPA)2 in vitro and in vivo was demonstrated. The 30-minute contrast washout percentage is a useful parameter for examining tumor angiogenesis and for the early assessment of antiangiogenic treatment response.
Collapse
Affiliation(s)
- Wei-Tsung Chen
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tiffany Ting Fang Shih
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ran-Chou Chen
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shin-Yang Tu
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsieh Wen-Yuen
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Pang-Chyr Yang
- From the Department of Radiology, Taipei City Hospital, Taipei; Departments of Radiology, Medical Imaging, and Medicine, School of Medicine, National Taiwan University, Taipei; Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan; Union Chemical Laboratory, Industrial Technology Research Institute, Hsinchu, Taiwan; and Department of Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
27
|
Vascular density and endothelial cell expression of integrin alpha v beta 3 and E-selectin in murine tumours. Tumour Biol 2012; 33:1709-17. [PMID: 22669616 DOI: 10.1007/s13277-012-0428-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 05/23/2012] [Indexed: 01/17/2023] Open
Abstract
The endothelial cell adhesion molecules, including the integrin alpha v beta 3 (αvβ3) and E-selectin, are involved in the process of angiogenesis required for tumour growth, cell migration and metastasis. The purpose of this study was to assess and compare widely used tumour models to select the ones most suitable for angiogenesis research. Fifteen murine tumours were selected including melanoma (B16), colon (C26, C38, C51), mammary (MA13, MA16, MA16/Adr, MA17, MA17/Adr, MA25, MA44), pancreatic (PO2, PO3), Glasgow osteogenic sarcoma (GOS) and Lewis lung carcinoma (LLC). The tumour vascular density, assessed using the platelet endothelial cell adhesion molecule 1 (PECAM-1; CD31) immunostaining, revealed that B16 melanoma was poorly vascularized (<5 %), whereas the colon and mammary tumours were well vascularized (5-15 %). The most vascularized tumours (>15 %) were the pancreatic tumours (PO2 and PO3), the sarcoma (GOS) and the lung tumour (LLC). The integrin αvβ3 and E-selectin, evaluated by immunohistology, showed that 7/15 tumours expressed the αvβ3 integrin which was homogeneously distributed on all tumour sections (B16, C26, MA17/Adr, MA25, MA44, PO2, LLC). E-selectin was expressed in 4/15 tumours and its expression was restricted to the tumour periphery. Only 2/15 tumours (B16 and C26) were shown to express both integrin αvβ3 and E-selectin. In conclusion, these data not only contribute to a better understanding of the tumour biology of murine tumours but can also guide the choice of appropriate models for antiangiogenic therapy, for selective drug delivery to tumours and the validation of tumour imaging modalities targeting these endothelial cell adhesion molecules.
Collapse
|
28
|
Magnetic Nanoparticles in the Imaging of Tumor Angiogenesis. APPLIED SCIENCES-BASEL 2012. [DOI: 10.3390/app2020525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Abstract
What are nanoparticles and why are they important in dermatology? These questions are addressed by highlighting recent developments in the nanotechnology field that have increased the potential for intentional and unintentional nanoparticle skin exposure. The role of environmental factors in the interaction of nanoparticles with skin and the potential mechanisms by which nanoparticles may influence skin response to environmental factors are discussed. Trends emerging from recent literature suggest that the positive benefit of engineered nanoparticles for use in cosmetics and as tools for understanding skin biology and curing skin disease outweigh potential toxicity concerns. Discoveries reported in this journal are highlighted. This review begins with a general introduction to the field of nanotechnology and nanomedicine. This is followed by a discussion of the current state of understanding of nanoparticle skin penetration and their use in three therapeutic applications. Challenges that must be overcome to derive clinical benefit from the application of nanotechnology to skin are discussed last, providing perspective on the significant opportunity that exists for future studies in investigative dermatology.
Collapse
Affiliation(s)
- Lisa A DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14642, USA.
| |
Collapse
|