51
|
Desa DE, Bhanote M, Hill RL, Majeski JB, Buscaglia B, D’Aguiar M, Strawderman R, Hicks DG, Turner BM, Brown EB. Second-harmonic generation directionality is associated with neoadjuvant chemotherapy response in breast cancer core needle biopsies. JOURNAL OF BIOMEDICAL OPTICS 2019; 24:1-9. [PMID: 31456385 PMCID: PMC6983524 DOI: 10.1117/1.jbo.24.8.086503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/05/2019] [Indexed: 05/30/2023]
Abstract
Neoadjuvant chemotherapy (NACT) is routinely administered to subsets of breast cancer patients, including triple negative (TN) or human epidermal growth factor receptor 2-positive (HER2+) cancers. After NACT and subsequent surgical resection, 5% to 30% of patients have no residual invasive carcinoma, termed pathological complete response. Unfortunately, many patients experience little-to-no response after NACT and unnecessarily suffer its side effects. Methods are needed to predict an individual patient’s response to NACT. Core needle biopsies, taken before NACT, consist of tumor cells and the surrounding extracellular matrix. We performed second-harmonic generation (SHG) imaging of fibrillar collagen in core needle biopsy sections as a possible predictor of response to NACT. The ratio of forward-to-backward scattering (F/B) SHG was assessed in the “tumor bulk” and “tumor–host interface” in HER2+ and TN core needle biopsy sections. Patient response was classified post-treatment using the Residual Cancer Burden (RCB) score. In HER2+ biopsies, RCB class was associated with F/B derived from the tumor–stromal interface, but not tumor bulk. F/B was not associated with RCB class in TN biopsies. These findings suggest that F/B from needle biopsy sections may be a useful predictor of which patients will respond favorably to NACT, with the potential to help reduce overtreatment.
Collapse
Affiliation(s)
- Danielle E. Desa
- University of Rochester, Hajim School of Engineering and Applied Sciences, Department of Biomedical Engineering, Rochester, New York, United States
| | - Monisha Bhanote
- University of Rochester Medical Center, School of Medicine and Dentistry, Department of Pathology and Laboratory Medicine, Rochester, New York, United States
| | - Robert L. Hill
- Harmonigenic Corporation, Rochester, New York, United States
| | - Joseph B. Majeski
- University of Rochester, Hajim School of Engineering and Applied Sciences, Department of Biomedical Engineering, Rochester, New York, United States
| | - Brandon Buscaglia
- Rochester Institute of Technology, Kate Gleason College of Engineering, Department of Biomedical Engineering, Rochester, New York, United States
| | - Marcus D’Aguiar
- Rochester Institute of Technology, Kate Gleason College of Engineering, Department of Biomedical Engineering, Rochester, New York, United States
| | - Robert Strawderman
- University of Rochester Medical Center, School of Medicine and Dentistry, Department of Biostatistics and Computational Biology, Rochester, New York, United States
| | - David G. Hicks
- University of Rochester Medical Center, School of Medicine and Dentistry, Department of Pathology and Laboratory Medicine, Rochester, New York, United States
| | - Bradley M. Turner
- University of Rochester Medical Center, School of Medicine and Dentistry, Department of Pathology and Laboratory Medicine, Rochester, New York, United States
| | - Edward B. Brown
- University of Rochester, Hajim School of Engineering and Applied Sciences, Department of Biomedical Engineering, Rochester, New York, United States
| |
Collapse
|
52
|
Ciocan-Cartita CA, Jurj A, Buse M, Gulei D, Braicu C, Raduly L, Cojocneanu R, Pruteanu LL, Iuga CA, Coza O, Berindan-Neagoe I. The Relevance of Mass Spectrometry Analysis for Personalized Medicine through Its Successful Application in Cancer "Omics". Int J Mol Sci 2019; 20:ijms20102576. [PMID: 31130665 PMCID: PMC6567119 DOI: 10.3390/ijms20102576] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 01/06/2023] Open
Abstract
Mass spectrometry (MS) is an essential analytical technology on which the emerging omics domains; such as genomics; transcriptomics; proteomics and metabolomics; are based. This quantifiable technique allows for the identification of thousands of proteins from cell culture; bodily fluids or tissue using either global or targeted strategies; or detection of biologically active metabolites in ultra amounts. The routine performance of MS technology in the oncological field provides a better understanding of human diseases in terms of pathophysiology; prevention; diagnosis and treatment; as well as development of new biomarkers; drugs targets and therapies. In this review; we argue that the recent; successful advances in MS technologies towards cancer omics studies provides a strong rationale for its implementation in biomedicine as a whole.
Collapse
Affiliation(s)
- Cristina Alexandra Ciocan-Cartita
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
| | - Ancuța Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy.
| | - Mihail Buse
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
| | - Diana Gulei
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy.
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy.
| | - Roxana Cojocneanu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy.
| | - Lavinia Lorena Pruteanu
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
| | - Cristina Adela Iuga
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 6 Louis Pasteur Street, 400349 Cluj-Napoca.
| | - Ovidiu Coza
- Department of Oncology, "Iuliu Hațieganu" University of Medicine and Pharmacy, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania.
- Department of Radiotherapy with High Energies and Brachytherapy, Oncology Institute "Prof. Dr. Ion Chiricuta", 34-36 Republicii Street, 400015 Cluj-Napoca.
| | - Ioana Berindan-Neagoe
- MEDFUTURE -Research Center for Advanced Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy, 4-6 Louis Pasteur Street, 400349 Cluj-Napoca, Romania.
- Research Center for Functional Genomics, Biomedicine and Translational Medicine," Iuliu Hațieganu" University of Medicine and Pharmacy.
- Department of Functional Genomics and Experimental Pathology, Ion Chiricuțǎ Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca.
| |
Collapse
|
53
|
Barrios-Gumiel A, Sepúlveda-Crespo D, Jiménez JL, Gómez R, Muñoz-Fernández MÁ, de la Mata FJ. Dendronized magnetic nanoparticles for HIV-1 capture and rapid diagnostic. Colloids Surf B Biointerfaces 2019; 181:360-368. [PMID: 31158698 DOI: 10.1016/j.colsurfb.2019.05.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 01/29/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) remains a global public health problem. Detection and reduction of the rates of late diagnosis of HIV-1 infection are one of the main challenges in combating the HIV-1 epidemic. Magnetic nanoparticles (MNPs) have several characteristics that make them susceptible to capture HIV-1 of a wide range of biological samples reducing waiting times between the acquisition of HIV-1 infection and its detection by current techniques. Carbosilane dendrons decorated with peripheral carboxyl groups and alcoxysilane function at the focal point have been used to stabilize MNPs by co-precipitation method in one step. The characterization of these systems and of their carboxylate analogues was performed by Fourier transform infrared spectroscopy (FTIR), transmission electron microscopy (TEM), dynamic light scattering (DLS), ζ potential and thermal gravimetric analysis (TGA). The ability of carboxyl and carboxylate MNPs to capture R5-HIV-1 and X4-HIV-1 strains was evaluated to achieve a rapid and easy diagnostic method in order to reduce or eliminate the risk of HIV-1 transmission.
Collapse
Affiliation(s)
- Andrea Barrios-Gumiel
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain
| | - Daniel Sepúlveda-Crespo
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain
| | - José Luis Jiménez
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain; Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, Madrid, Spain; IiSGM, Madrid, Spain
| | - Rafael Gómez
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain
| | - María Ángeles Muñoz-Fernández
- Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Sección Inmunología. Laboratorio InmunoBiología Molecular, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Spanish HIV HGM BioBank, Madrid, Spain; Plataforma de Laboratorio, Hospital General Universitario Gregorio Marañón, Madrid, Spain; IiSGM, Madrid, Spain.
| | - F Javier de la Mata
- Dpto. de Química Orgánica y Química Inorgánica, Universidad de Alcalá (UAH), Campus Universitario, E-28871 Alcalá de Henares (Madrid), Spain; Instituto de Investigación Química "Andrés M. del Río" (IQAR), Universidad de Alcalá (UAH), Spain; Networking Research Center for Bioengineering, Biomaterials and Nanomedicine, (CIBER-BBN), Spain; Instituto Ramón y Cajal de Investigación Sanitaria, IRYCIS, Spain.
| |
Collapse
|
54
|
Chen R, Chen Q, Qin H, Xing D. A photoacoustic shockwave triggers the size shrinkage of nanoparticles to obviously improve tumor penetration and therapeutic efficacy. NANOSCALE 2019; 11:1423-1436. [PMID: 30608103 DOI: 10.1039/c8nr08271e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Drug delivery to a tumor site with an insufficient microvascular network remains a challenge due to the size preference for transport in terms of circulation and distribution. In this work, an integrated nano-therapeutic parcel disintegrable by a photoacoustic shockwave was developed. Nano-therapeutic particles with red absorbance are packaged into a larger parcel to generate a longer circulation half-life and improved accumulation in tumor tissue. Pulse-laser irradiation is absorbed by the nanoparticles and it generates a photoacoustic shockwave. This triggers a liquid-gas phase transition of the nano-parcel, leading to the high-efficiency release of smaller nanoparticles, thus achieving excellent therapeutic diffusion with improved uniformity. This results in a highly effective therapeutic effect, as demonstrated with both in vitro and in vivo tumor models. Compared to previously reported work, this approach has the distinctive advantage of precisely controllable therapeutic release that is independent of the physiological environment in the tumor and it is less limited than a UV-based release mechanism. In addition, the concept of photoacoustic shockwave-based nanoparticle release can be extended over a wide wavelength range, including microwaves, to match specific needs and achieve optimal therapeutic depth. The results demonstrate that the proposed strategy holds great potential for improved tumor therapy efficacy.
Collapse
Affiliation(s)
- Rong Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, South China Normal University, Guangzhou 510631, China.
| | | | | | | |
Collapse
|
55
|
Hsu YH, Liu WW, Wu TH, Lee CJT, Chen YH, Li PC. Study of diffusive- and convective-transport mediated microtumor growth in a controlled microchamber. Biomed Microdevices 2019; 21:7. [PMID: 30607550 DOI: 10.1007/s10544-018-0356-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this paper, we report on using mass transport to control nutrition supply of colorectal cancer cells for developing a microtumor in a confined microchamber. To mimic the spatial heterogeneity of a tumor, two microfluidic configurations based on resistive circuits are designed. One has a convection-dominated microchamber to simulate the tumor region proximal to leaky blood vessels. The other has a diffusion-dominated microchamber to mimic the tumor core that lacks blood vessels and nutrient supply. Thus, the time for nutrition to fill the microchamber can vary from tens of minutes to several hours. Results show that cells cultured under a diffusive supply of nutrition have a high glycolytic rate and a nearly constant oxygen consumption rate. In contrast, cells cultured under convective supply of nutrition have a gradual increase of oxygen consumption rate with a low glycolytic rate. This suggests that cancer cells have distinct reactions under different mass transport and nutrition supply. Using these two microfluidic platforms to create different rate of nutrition supply, it is found that a continuous microtumor that almost fills the mm-size microchamber can be developed under a low-nutrient supply environment, but not for the convective condition. It also is demonstrated that microchannels can simulate the delivery of anti-cancer drugs to the microtumor under controlled mass-transport. This method provides a means to develop a larger scale microtumor in a lab-on-a-Chip system for post development and stimulations, and microchannels can be applied to control the physical and chemical environment for anti-cancer drug screening.
Collapse
Affiliation(s)
- Yu-Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China.
| | - Wei-Wen Liu
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China
| | - Tung-Han Wu
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China
| | - Carina Jean-Tien Lee
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China
| | - Yu-Hsi Chen
- Institute of Applied Mechanics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China
| | - Pai-Chi Li
- Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, No. 1, Sec.4, Roosevelt Rd., Taipei, 10617, Taiwan, Republic of China
| |
Collapse
|
56
|
Zhao N, Yan L, Zhao X, Chen X, Li A, Zheng D, Zhou X, Dai X, Xu FJ. Versatile Types of Organic/Inorganic Nanohybrids: From Strategic Design to Biomedical Applications. Chem Rev 2018; 119:1666-1762. [DOI: 10.1021/acs.chemrev.8b00401] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liemei Yan
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyi Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xinyan Chen
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Aihua Li
- College of Materials Science and Engineering, Institute for Graphene Applied Technology Innovation, Laboratory of Fiber Materials and Modern Textiles, Growing Base for State Key Laboratory, Collaborative Innovation Center for Marine Biomass Fibers Materials and Textiles of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Di Zheng
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xin Zhou
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoguang Dai
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
57
|
Teijeiro-Valiño C, Novoa-Carballal R, Borrajo E, Vidal A, Alonso-Nocelo M, de la Fuente Freire M, Lopez-Casas PP, Hidalgo M, Csaba N, Alonso MJ. A multifunctional drug nanocarrier for efficient anticancer therapy. J Control Release 2018; 294:154-164. [PMID: 30529724 DOI: 10.1016/j.jconrel.2018.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/28/2018] [Accepted: 12/02/2018] [Indexed: 12/25/2022]
Abstract
So far, the success of anticancer nanomedicines has been moderate due to their lack of adequate targeting properties and/or to their difficulties for penetrating tumors. Here we report a multifunctional drug nanocarrier consisting of hyaluronic acid nanocapsules conjugated with the tumor homing peptide tLyp1, which exhibits both, dual targeting properties (to the tumor and to the lymphatics), and enhanced tumor penetration. Data from a 3D co-culture in vitro model showed the capacity of these nanocapsules to interact with the NRP1 receptors over-expressed in cancer cells. The targeting capacity of the nanocapsules was evidenced in orthotopic lung cancer-bearing mice, using docetaxel as a standard drug. The results showed a dramatic accumulation of docetaxel in the tumor (37-fold the one achieved with Taxotere®). This biodistribution profile correlated with the high efficacy shown in terms of tumor growth regression and drastic reduction of metastasis in the lymphatics. When efficacy was validated in a pancreatic patient-derived tumor, the nanocapsule's activity was comparable to that of a dose ten times higher of Abraxane®. Multi-functionality was found to be the key to the success of this new therapy.
Collapse
Affiliation(s)
- Carmen Teijeiro-Valiño
- Department of Pharmacy and Pharmaceutical Technology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ramon Novoa-Carballal
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, Portugal
| | - Erea Borrajo
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Anxo Vidal
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marta Alonso-Nocelo
- Nano-Oncology Unit. Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, CIBERONC, Santiago de Compostela, Spain
| | - María de la Fuente Freire
- Nano-Oncology Unit. Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, CIBERONC, Santiago de Compostela, Spain
| | - Pedro P Lopez-Casas
- Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Manuel Hidalgo
- Spanish National Cancer Research Centre (CNIO), Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Noémi Csaba
- Department of Pharmacy and Pharmaceutical Technology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - María José Alonso
- Department of Pharmacy and Pharmaceutical Technology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
58
|
Chen H, Gu Z, An H, Chen C, Chen J, Cui R, Chen S, Chen W, Chen X, Chen X, Chen Z, Ding B, Dong Q, Fan Q, Fu T, Hou D, Jiang Q, Ke H, Jiang X, Liu G, Li S, Li T, Liu Z, Nie G, Ovais M, Pang D, Qiu N, Shen Y, Tian H, Wang C, Wang H, Wang Z, Xu H, Xu JF, Yang X, Zhu S, Zheng X, Zhang X, Zhao Y, Tan W, Zhang X, Zhao Y. Precise nanomedicine for intelligent therapy of cancer. Sci China Chem 2018. [DOI: 10.1007/s11426-018-9397-5] [Citation(s) in RCA: 293] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
59
|
Deonarain MP. Miniaturised 'antibody'-drug conjugates for solid tumours? DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:47-53. [PMID: 30553520 DOI: 10.1016/j.ddtec.2018.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 06/09/2023]
Abstract
With Antibody-Drug Conjugate strategies firmly focussed on the precise conjugation to the large protein Immunoglobulin-G format, it is easy to miss the more recent technological innovations in small-format drug conjugates. Here, the targeting ligand can be at 50-95% reduced in size, or even smaller if peptidic in nature. Antibody domains or alternative binding scaffolds, chemically-modified with ultra-potent cytotoxic payloads offer an alternative approach for oncology therapeutics, promising a wider therapeutic window by virtue of superior solid tumour penetration properties and more rapid system clearance. Many of the traditional ADC concepts still apply, but as these miniaturised ADCs enter the clinic over the next 2-3 years, we will learn whether these new features translate to patient benefits.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd, Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2FX, UK; Dept. of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK.
| |
Collapse
|
60
|
Regional Infusion of Chimeric Antigen Receptor T Cells to Overcome Barriers for Solid Tumor Immunotherapy. J Vasc Interv Radiol 2018; 29:1017-1021.e1. [PMID: 29935783 DOI: 10.1016/j.jvir.2018.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/22/2018] [Accepted: 03/02/2018] [Indexed: 11/24/2022] Open
|
61
|
Jo Y, Choi N, Kim K, Koo HJ, Choi J, Kim HN. Chemoresistance of Cancer Cells: Requirements of Tumor Microenvironment-mimicking In Vitro Models in Anti-Cancer Drug Development. Am J Cancer Res 2018; 8:5259-5275. [PMID: 30555545 PMCID: PMC6276092 DOI: 10.7150/thno.29098] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/04/2018] [Indexed: 01/09/2023] Open
Abstract
For decades, scientists have been using two-dimensional cell culture platforms for high-throughput drug screening of anticancer drugs. Growing evidence indicates that the results of anti-cancer drug screening vary with the cell culture microenvironment, and this variation has been proposed as a reason for the high failure rate of clinical trials. Since the culture condition-dependent drug sensitivity of anti-cancer drugs may negatively impact the identification of clinically effective drug candidates, more reliable in vitro cancer platforms are urgently needed. In this review article, we provide an overview of how cell culture conditions can alter drug efficacy and highlight the importance of developing more reliable cancer drug testing platforms for use in the drug discovery process. The environmental factors that can alter drug delivery and efficacy are reviewed. Based on these observations of chemoresistant tumor physiology, we summarize the recent advances in the fabrication of in vitro cancer models and the model-dependent cytotoxicity of anti-cancer drugs, with a particular focus on engineered environmental factors in these platforms. It is believed that more physiologically relevant cancer models can revolutionize the drug discovery process.
Collapse
|
62
|
Amin M, Pourshohod A, Kheirollah A, Afrakhteh M, Gholami-Borujeni F, Zeinali M, Jamalan M. Specific delivery of idarubicin to HER2-positive breast cancerous cell line by trastuzumab-conjugated liposomes. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
63
|
Kremheller J, Vuong AT, Yoshihara L, Wall WA, Schrefler BA. A monolithic multiphase porous medium framework for (a-)vascular tumor growth. COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 2018; 340:657-683. [PMID: 33132456 PMCID: PMC7598028 DOI: 10.1016/j.cma.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
We present a dynamic vascular tumor model combining a multiphase porous medium framework for avascular tumor growth in a consistent Arbitrary Lagrangian Eulerian formulation and a novel approach to incorporate angiogenesis. The multiphase model is based on Thermodynamically Constrained Averaging Theory and comprises the extracellular matrix as a porous solid phase and three fluid phases: (living and necrotic) tumor cells, host cells and the interstitial fluid. Angiogenesis is modeled by treating the neovasculature as a proper additional phase with volume fraction or blood vessel density. This allows us to define consistent inter-phase exchange terms between the neovasculature and the interstitial fluid. As a consequence, transcapillary leakage and lymphatic drainage can be modeled. By including these important processes we are able to reproduce the increased interstitial pressure in tumors which is a crucial factor in drug delivery and, thus, therapeutic outcome. Different coupling schemes to solve the resulting five-phase problem are realized and compared with respect to robustness and computational efficiency. We find that a fully monolithic approach is superior to both the standard partitioned and a hybrid monolithic-partitioned scheme for a wide range of parameters. The flexible implementation of the novel model makes further extensions (e.g., inclusion of additional phases and species) straightforward.
Collapse
Affiliation(s)
- Johannes Kremheller
- Institute for Computational Mechanics, Technische Universität München, Boltzmannstrasse 15, D-85748 Garching b. München, Germany
| | - Anh-Tu Vuong
- Institute for Computational Mechanics, Technische Universität München, Boltzmannstrasse 15, D-85748 Garching b. München, Germany
| | - Lena Yoshihara
- Institute for Computational Mechanics, Technische Universität München, Boltzmannstrasse 15, D-85748 Garching b. München, Germany
| | - Wolfgang A. Wall
- Institute for Computational Mechanics, Technische Universität München, Boltzmannstrasse 15, D-85748 Garching b. München, Germany
| | - Bernhard A. Schrefler
- Institute for Advanced Study, Technische Universität München, Lichtenbergstrasse 2a, D-85748 Garching b. München, Germany
- Department of Civil, Environmental and Architectural Engineering, University of Padova, Italy
| |
Collapse
|
64
|
Choi E, Yoo W, Park JH, Kim S. Simultaneous Delivery of Electrostatically Complexed Multiple Gene-Targeting siRNAs and an Anticancer Drug for Synergistically Enhanced Treatment of Prostate Cancer. Mol Pharm 2018; 15:3777-3785. [PMID: 30028622 DOI: 10.1021/acs.molpharmaceut.8b00227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Simultaneous silencing of multiple apoptosis-related genes is an attractive approach to treat cancer. In this article, we present a multiple gene-targeting siRNA/drug delivery system for prostate cancer treatment with a high efficiency. Bcl-2, survivin, and androgen receptor genes involved in the cell apoptosis pathways were chosen as silencing targets with three different siRNAs. The colloidal nanocomplex delivery system (<10 nm in size) was formulated electrostatically between anionic siRNAs and a cationic drug (BZT), followed by encapsulation with the Pluronic F-68 polymer. The formulated nanocomplex system exhibited sufficient stability against nuclease-induced degradation, leading to successful intracellular delivery for the desired therapeutic performance. Silencing of targeted genes and apoptosis induction were evaluated in vitro on human prostate LNCaP-LN3 cancer cells by using various biological analysis tools (e.g., real-time PCR, MTT cell viability test, and flow cytometry). It was demonstrated that when the total loaded siRNA amounts were kept the same in the nanocomplexes, the simultaneous silencing of triple genes with co-loaded siRNAs (i.e., Bcl-2, survivin, and AR-targeting siRNAs) enhanced BZT-induced apoptosis of cancer cells more efficiently than the silencing of each single gene alone, offering a novel way of improving the efficacy of gene therapeutics including anticancer drug.
Collapse
Affiliation(s)
- Eunshil Choi
- Center for Theragnosis , Korea Institute of Science and Technology (KIST) , Seoul 136-791 , Korea
| | - Wonjae Yoo
- Center for Theragnosis , Korea Institute of Science and Technology (KIST) , Seoul 136-791 , Korea.,School of Chemical Engineering, College of Engineering , Sungkyunkwan Univeristy , Suwon 440-746 , Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering , Sungkyunkwan Univeristy , Suwon 440-746 , Korea
| | - Sehoon Kim
- Center for Theragnosis , Korea Institute of Science and Technology (KIST) , Seoul 136-791 , Korea.,Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 136-791 , Korea
| |
Collapse
|
65
|
Engler FA, Polli JR, Li T, An B, Otteneder M, Qu J, Balthasar JP. "Catch-and-Release" Anti-Carcinoembryonic Antigen Monoclonal Antibody Leads to Greater Plasma and Tumor Exposure in a Mouse Model of Colorectal Cancer. J Pharmacol Exp Ther 2018; 366:205-219. [PMID: 29735609 PMCID: PMC6040839 DOI: 10.1124/jpet.117.246900] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 05/01/2018] [Indexed: 11/22/2022] Open
Abstract
In this study, we examined the effects of target expression, neonatal Fc receptor (FcRn) expression in tumors, and pH-dependent target binding on the disposition of monoclonal antibodies (mAbs) in murine models of colorectal cancer. A panel of anti-carcinoembryonic antigen (CEA) mAbs was developed via standard hybridoma technology and then evaluated for pH-dependent CEA binding. Binding was assessed via immunoassay and radioligand binding assays. 10H6, a murine IgG1 mAb with high affinity for CEA at pH = 7.4 (KD = 12.6 ± 1.7 nM) and reduced affinity at pH = 6.0 (KD = 144.6 ± 21.8 nM), and T84.66, which exhibits pH-independent CEA binding (KD = 1.1 ± 0.11 and 1.4 ± 0.16 nM at pH 7.4 and 6.0), were selected for pharmacokinetic investigations. We evaluated pharmacokinetics after intravenous administration to control mice and to mice bearing tumors with (MC38CEA+, LS174T) and without (MC38CEA-) CEA expression and with or without expression of murine FcRn, at doses of 0.1, 1, and 10 mg/kg. 10H6 displayed linear pharmacokinetics in mice bearing MC38CEA+ or MC38CEA- tumors. T84.66 displayed linear pharmacokinetics in mice with MC38CEA- tumors but dose-dependent nonlinear pharmacokinetics in mice bearing MC38CEA+ In addition to the improved plasma pharmacokinetic profile (i.e., linear pharmacokinetics, longer terminal half-life), 10H6 exhibited improved exposure in MC38CEA+ tumors relative to T84.66. In mice bearing tumors with CEA expression, but lacking expression of murine FcRn (LS174T), 10H6 demonstrated nonlinear pharmacokinetics, with rapid clearance at low dose. These data are consistent with the hypothesis that pH-dependent CEA binding allows mAb dissociation from target in acidified endosomes, enabling FcRn-mediated protection from target-mediated elimination in mice bearing MC38CEA+ tumors.
Collapse
Affiliation(s)
- Frank A Engler
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Joseph Ryan Polli
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Tommy Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Bo An
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Michael Otteneder
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo (F.A.E., J.R.P., T.L., B.A., J.Q., J.P.B.) and New York State Center of Excellence in Bioinformatics and Life Sciences (B.A., J.Q.), Buffalo, New York; and F. Hoffmann-La Roche Ltd., Roche Innovation Center, Basel, Switzerland (M.O.)
| |
Collapse
|
66
|
Sadeghipour N, Davis SC, Tichauer KM. Correcting for targeted and control agent signal differences in paired-agent molecular imaging of cancer cell-surface receptors. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-11. [PMID: 29931837 PMCID: PMC6013418 DOI: 10.1117/1.jbo.23.6.066004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/31/2018] [Indexed: 05/05/2023]
Abstract
Paired-agent kinetic modeling protocols provide one means of estimating cancer cell-surface receptors with in vivo molecular imaging. The protocols employ the coadministration of a control imaging agent with one or more targeted imaging agent to account for the nonspecific uptake and retention of the targeted agent. These methods require the targeted and control agent data be converted to equivalent units of concentration, typically requiring specialized equipment and calibration, and/or complex algorithms that raise the barrier to adoption. This work evaluates a kinetic model capable of correcting for targeted and control agent signal differences. This approach was compared with an existing simplified paired-agent model (SPAM), and modified SPAM that accounts for signal differences by early time point normalization of targeted and control signals (SPAMPN). The scaling factor model (SPAMSF) outperformed both SPAM and SPAMPN in terms of accuracy and precision when the scale differences between targeted and imaging agent signals (α) were not equal to 1, and it matched the performance of SPAM for α = 1. This model could have wide-reaching implications for quantitative cancer receptor imaging using any imaging modalities, or combinations of imaging modalities, capable of concurrent detection of at least two distinct imaging agents (e.g., SPECT, optical, and PET/MR).
Collapse
Affiliation(s)
- Negar Sadeghipour
- Illinois Institute of Technology, Biomedical Engineering, Chicago, Illinois, United States
| | - Scott C. Davis
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, United States
| | - Kenneth M. Tichauer
- Illinois Institute of Technology, Biomedical Engineering, Chicago, Illinois, United States
- Address all correspondence to: Kenneth M. Tichauer, E-mail:
| |
Collapse
|
67
|
Qiu Y, Ahn B, Sakurai Y, Hansen CE, Tran R, Mimche PN, Mannino RG, Ciciliano JC, Lamb TJ, Joiner CH, Ofori-Acquah SF, Lam WA. Microvasculature-on-a-chip for the long-term study of endothelial barrier dysfunction and microvascular obstruction in disease. Nat Biomed Eng 2018; 2:453-463. [PMID: 30533277 PMCID: PMC6286070 DOI: 10.1038/s41551-018-0224-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Alterations in the mechanical properties of erythrocytes occurring in inflammatory and hematologic disorders such as sickle cell disease (SCD) and malaria often lead to increased endothelial permeability, haemolysis, and microvascular obstruction. However, the associations among these pathological phenomena remain unknown. Here, we report a perfusable, endothelialized microvasculature-on-a-chip featuring an interpenetrating-polymer-network hydrogel that recapitulates the stiffness of blood-vessel intima, basement membrane self-deposition and self-healing endothelial barrier function for longer than 1 month. The microsystem enables the real-time visualization, with high spatiotemporal resolution, of microvascular obstruction and endothelial permeability under physiological flow conditions. We found how extracellular heme, a hemolytic byproduct, induces delayed but reversible endothelial permeability in a dose-dependent manner, and demonstrate that endothelial interactions with SCD or malaria-infected erythrocytes cause reversible microchannel occlusion and increased in situ endothelial permeability. The microvasculature-on-a-chip enables mechanistic insight into the endothelial barrier dysfunction associated with SCD, malaria and other inflammatory and haematological diseases.
Collapse
Affiliation(s)
- Yongzhi Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Byungwook Ahn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yumiko Sakurai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Caroline E Hansen
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Reginald Tran
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Patrice N Mimche
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Robert G Mannino
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jordan C Ciciliano
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Tracey J Lamb
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Clinton H Joiner
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Solomon F Ofori-Acquah
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Center for Translational and International Hematology, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.,School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Wilbur A Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA. .,Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
68
|
Zhang YR, Lin R, Li HJ, He WL, Du JZ, Wang J. Strategies to improve tumor penetration of nanomedicines through nanoparticle design. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2018; 11:e1519. [PMID: 29659166 DOI: 10.1002/wnan.1519] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/28/2018] [Accepted: 03/10/2018] [Indexed: 12/17/2022]
Abstract
Nanoparticles (NPs) have emerged as an effective means to deliver therapeutic drugs for cancer treatment, as they can preferentially accumulate at tumor site through the enhanced permeability and retention effect. Various forms of NPs including liposomes, polymeric micelles, and inorganic particles have been used for therapeutic applications. However, the therapeutic benefits of nanomedicines are suboptimal. Although many possible reasons may account for the compromised therapeutic efficacy, the inefficient tumor penetration can be a vital obstacle. Tumor develops characteristic pathological environment, such as abnormal vasculature, elevated interstitial fluid pressure, and dense extracellular matrix, which intrinsically hinder the transport of nanomedicines in the tumor parenchyma. The physicochemical properties of the NPs such as size, shape, and surface charge have profound effect on tumor penetration. In this review, we will highlight the factors that affect the transport of NPs in solid tumor, and then elaborate on designing strategies to improve NPs' penetration and uniform distribution inside the tumor interstitium. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Ya-Ru Zhang
- School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, China
| | - Run Lin
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hong-Jun Li
- School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, China
| | - Wei-Ling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jin-Zhi Du
- School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, China
| | - Jun Wang
- School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, Guangdong, China.,National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, Guangdong, China
| |
Collapse
|
69
|
Deonarain MP, Yahioglu G, Stamati I, Pomowski A, Clarke J, Edwards BM, Diez-Posada S, Stewart AC. Small-Format Drug Conjugates: A Viable Alternative to ADCs for Solid Tumours? Antibodies (Basel) 2018; 7:E16. [PMID: 31544868 PMCID: PMC6698822 DOI: 10.3390/antib7020016] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022] Open
Abstract
Antibody-Drug Conjugates (ADCs) have been through multiple cycles of technological innovation since the concept was first practically demonstrated ~40 years ago. Current technology is focusing on large, whole immunoglobulin formats (of which there are approaching 100 in clinical development), many with site-specifically conjugated payloads numbering 2 or 4. Despite the success of trastuzumab-emtansine in breast cancer, ADCs have generally failed to have an impact in solid tumours, leading many to explore alternative, smaller formats which have better penetrating properties as well as more rapid pharmacokinetics (PK). This review describes research and development progress over the last ~10 years obtained from the primary literature or conferences covering over a dozen different smaller format-drug conjugates from 80 kDa to around 1 kDa in total size. In general, these agents are potent in vitro, particularly more recent ones incorporating ultra-potent payloads such as auristatins or maytansinoids, but this potency profile changes when testing in vivo due to the more rapid clearance. Strategies to manipulate the PK properties, whilst retaining the more effective tumour penetrating properties could at last make small-format drug conjugates viable alternative therapeutics to the more established ADCs.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
- Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, UK.
| | - Gokhan Yahioglu
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
- Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, UK.
| | - Ioanna Stamati
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Anja Pomowski
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - James Clarke
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Bryan M Edwards
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Soraya Diez-Posada
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Ashleigh C Stewart
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| |
Collapse
|
70
|
Lu H, Stenzel MH. Multicellular Tumor Spheroids (MCTS) as a 3D In Vitro Evaluation Tool of Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1702858. [PMID: 29450963 DOI: 10.1002/smll.201702858] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/13/2017] [Indexed: 05/23/2023]
Abstract
Multicellular tumor spheroid models (MCTS) are often coined as 3D in vitro models that can mimic the microenvironment of tissues. MCTS have gained increasing interest in the nano-biotechnology field as they can provide easily accessible information on the performance of nanoparticles without using animal models. Considering that many countries have put restrictions on animals testing, which will only tighten in the future as seen by the recent developments in the Netherlands, 3D models will become an even more valuable tool. Here, an overview on MCTS is provided, focusing on their use in cancer research as most nanoparticles are tested in MCTS for treatment of primary tumors. Thereafter, various types of nanoparticles-from self-assembled block copolymers to inorganic nanoparticles, are discussed. A range of physicochemical parameters including the size, shape, surface chemistry, ligands attachment, stability, and stiffness are found to influence nanoparticles in MCTS. Some of these studies are complemented by animal studies confirming that lessons from MCTS can in part predict the behaviour in vivo. In summary, MCTS are suitable models to gain additional information on nanoparticles. While not being able to replace in vivo studies, they can bridge the gap between traditional 2D in vitro studies and in vivo models.
Collapse
Affiliation(s)
- Hongxu Lu
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, Sydney, New South Wales, 2052, Australia
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, Sydney, New South Wales, 2052, Australia
| |
Collapse
|
71
|
Ke W, Yin W, Zha Z, Mukerabigwi JF, Chen W, Wang Y, He C, Ge Z. A robust strategy for preparation of sequential stimuli-responsive block copolymer prodrugs via thiolactone chemistry to overcome multiple anticancer drug delivery barriers. Biomaterials 2018; 154:261-274. [DOI: 10.1016/j.biomaterials.2017.11.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/18/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
|
72
|
Leakage correction improves prognosis prediction of dynamic susceptibility contrast perfusion MRI in primary central nervous system lymphoma. Sci Rep 2018; 8:456. [PMID: 29323247 PMCID: PMC5765049 DOI: 10.1038/s41598-017-18901-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022] Open
Abstract
To evaluate whether the cerebral blood volume (CBV) measurement with leakage correction from dynamic susceptibility contrast perfusion weighted imaging can be useful in predicting prognosis for primary central nervous system lymphoma (PCNSL). 46 PCNSL patients were included and classified by radiation therapy (RT) stratification into RT (n = 30) and non-RT (n = 16) groups. The corresponding histogram parameters of normalized CBV (nCBV) maps with or without leakage correction were calculated on contrast-enhanced T1 weighted image (CE T1WI) or on fluid attenuated inversion recovery image. The 75th percentile nCBV with leakage correction based on CE T1WI (T1 nCBVL75%) had a significant difference between the short and long progression free survival (PFS) subgroups of the RT group and the non-RT group, respectively. Based on the survival analysis, patients in the RT group with high T1 nCBVL75% had earlier progression than the others with a low T1 nCBVL75%. However, patients in the non-RT group with a high T1 nCBVL75% had slower progression than the others with a low T1 nCBVL75%. Based on RT stratification, the CBV with leakage correction has potential as a noninvasive biomarker for the prognosis prediction of PCNSL to identify high risk patients and it has a different correlation with the PFS based on the presence of combined RT.
Collapse
|
73
|
Hajipour H, Hamishehkar H, Nazari Soltan Ahmad S, Barghi S, Maroufi NF, Taheri RA. Improved anticancer effects of epigallocatechin gallate using RGD-containing nanostructured lipid carriers. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:283-292. [PMID: 29310467 DOI: 10.1080/21691401.2017.1423493] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The global burden of cancer have encouraged oncologists to develop novel strategies for treatment. Present study was proposed to develop Arginyl-glycyl-aspartic acid (RGD)-containing nanostructured lipid carriers (NLC) as a delivery system for improving the anticancer capability of epigallocatechin gallate (EGCG) on breast cancer cell line by attaching to integrin superfamily on cancer cells. For this purpose, RGD-containing EGCG-loaded NLC were prepared by hot homogenization technique and characterized by different techniques. Then, cytotoxic and apoptotic effects of prepared nanoparticles and their uptake into cells was evaluated. As results, the nanoparticles with particle size of 85 nm, zeta potential of -21 mV, encapsulation of 83% were prepared. Cytotoxicity and apoptosis experiments demonstrated that EGCG-loaded NLC-RGD possessed greatest apoptotic activity. Furthermore, it has been shown that, EGCG-loaded NLC-RGD causes cell cycle arresting more effective than EGCG. Therefore, loading EGCG into NLC-RGD make it more effective in both targeting and accumulation into tumour cells, which results from specialized uptake mechanism by adhesion to αvβ3 integrin. The results strengthen our hope that loading EGCG into RGD-containing NLC could possibly overcome the therapeutic limitations of EGCG and make it more effective in cancer therapy.
Collapse
Affiliation(s)
- Hamed Hajipour
- a Student Research Committee and Department of Reproductive Biology, Faculty of Advanced Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran.,b Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Hamed Hamishehkar
- c Drug Applied Research Center , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Saeed Nazari Soltan Ahmad
- d Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Siamak Barghi
- e Stem Cell and Regenerative Medicine Institute , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Nazila Fathi Maroufi
- d Department of Biochemistry and Clinical Laboratories, Faculty of Medical Sciences , Tabriz University of Medical Sciences , Tabriz , Iran
| | - Ramezan Ali Taheri
- b Nanobiotechnology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
74
|
Ghosh D, Peng X, Leal J, Mohanty R. Peptides as drug delivery vehicles across biological barriers. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018; 48:89-111. [PMID: 29963321 PMCID: PMC6023411 DOI: 10.1007/s40005-017-0374-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/25/2017] [Indexed: 12/15/2022]
Abstract
Peptides are small biological molecules that are attractive in drug delivery and materials engineering for applications including therapeutics, molecular building blocks and cell-targeting ligands. Peptides are small but can possess complexity and functionality as larger proteins. Due to their intrinsic properties, peptides are able to overcome the physiological and transport barriers presented by diseases. In this review, we discuss the progress of identifying and using peptides to shuttle across biological barriers and facilitate transport of drugs and drug delivery systems for improved therapy. Here, the focus of this review is on rationally designed, phage display peptides, and even endogenous peptides as carriers to penetrate biological barriers, specifically the blood-brain barrier(BBB), the gastrointestinal tract (GI), and the solid tumor microenvironment (T). We will discuss recent advances of peptides as drug carriers in these biological environments. From these findings, challenges and potential opportunities to iterate and improve peptide-based approaches will be discussed to translate their promise towards the clinic to deliver drugs for therapeutic efficacy.
Collapse
Affiliation(s)
- Debadyuti Ghosh
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Xiujuan Peng
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Jasmim Leal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| | - Rashmi Mohanty
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, 2409 University Ave, Austin, TX 78712, USA
| |
Collapse
|
75
|
Specific Targeting of Melanotic Cells with Peptide Ligated Photosensitizers for Photodynamic Therapy. Sci Rep 2017; 7:15750. [PMID: 29146972 PMCID: PMC5691209 DOI: 10.1038/s41598-017-15142-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/18/2017] [Indexed: 02/05/2023] Open
Abstract
A strategy combining covalent conjugation of photosensitizers to a peptide ligand directed to the melanocortin 1 (MC1) receptor with the application of sequential LED light dosage at near-IR wavelengths was developed to achieve specific cytotoxicity to melanocytes and melanoma (MEL) with minimal collateral damage to surrounding cells such as keratinocytes (KER). The specific killing of melanotic cells by targeted photodynamic therapy (PDT) described in this study holds promise as a potentially effective adjuvant therapeutic method to control benign skin hyperpigmentation or superficial melanotic malignancy such as Lentigo Maligna Melanoma (LMM).
Collapse
|
76
|
Yazdani S, Bansal R, Prakash J. Drug targeting to myofibroblasts: Implications for fibrosis and cancer. Adv Drug Deliv Rev 2017; 121:101-116. [PMID: 28720422 DOI: 10.1016/j.addr.2017.07.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/20/2017] [Accepted: 07/12/2017] [Indexed: 12/21/2022]
Abstract
Myofibroblasts are the key players in extracellular matrix remodeling, a core phenomenon in numerous devastating fibrotic diseases. Not only in organ fibrosis, but also the pivotal role of myofibroblasts in tumor progression, invasion and metastasis has recently been highlighted. Myofibroblast targeting has gained tremendous attention in order to inhibit the progression of incurable fibrotic diseases, or to limit the myofibroblast-induced tumor progression and metastasis. In this review, we outline the origin of myofibroblasts, their general characteristics and functions during fibrosis progression in three major organs: liver, kidneys and lungs as well as in cancer. We will then discuss the state-of-the art drug targeting technologies to myofibroblasts in context of the above-mentioned organs and tumor microenvironment. The overall objective of this review is therefore to advance our understanding in drug targeting to myofibroblasts, and concurrently identify opportunities and challenges for designing new strategies to develop novel diagnostics and therapeutics against fibrosis and cancer.
Collapse
Affiliation(s)
- Saleh Yazdani
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Targeted Therapeutics Division, Department of Biomaterials, Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; ScarTec Therapeutics BV, Enschede, The Netherlands.
| |
Collapse
|
77
|
Lozada-Delgado EL, Grafals-Ruiz N, Vivas-Mejía PE. RNA interference for glioblastoma therapy: Innovation ladder from the bench to clinical trials. Life Sci 2017; 188:26-36. [PMID: 28864225 PMCID: PMC5617340 DOI: 10.1016/j.lfs.2017.08.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and deadliest type of primary brain tumor with a prognosis of 14months after diagnosis. Current treatment for GBM patients includes "total" tumor resection, temozolomide-based chemotherapy, radiotherapy or a combination of these options. Although, several targeted therapies, gene therapy, and immunotherapy are currently in the clinic and/or in clinical trials, the overall survival of GBM patients has hardly improved over the last two decades. Therefore, novel multitarget modalities are urgently needed. Recently, RNA interference (RNAi) has emerged as a novel strategy for the treatment of most cancers, including GBM. RNAi-based therapies consist of using small RNA oligonucleotides to regulate protein expression at the post-transcriptional level. Despite the therapeutic potential of RNAi molecules, systemic limitations including short circulatory stability and low release into the tumor tissue have halted their progress to the clinic. The effective delivery of RNAi molecules through the blood-brain barrier (BBB) represents an additional challenge. This review focuses on connecting the translational process of RNAi-based therapies from in vitro evidence to pre-clinical studies. We delineate the effect of RNAi in GBM cell lines, describe their effectiveness in glioma mouse models, and compare the proposed drug carriers for the effective transport of RNAi molecules through the BBB to reach the tumor in the brain. Furthermore, we summarize the most important obstacles to overcome before RNAi-based therapy becomes a reality for GBM treatment.
Collapse
Affiliation(s)
- Eunice L Lozada-Delgado
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR 00927, United States; Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States; Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States
| | - Nilmary Grafals-Ruiz
- Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States; Department of Physiology, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States
| | - Pablo E Vivas-Mejía
- Comprehensive Cancer Center, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States; Department of Biochemistry, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00935, United States.
| |
Collapse
|
78
|
de Souza ALR, Marra K, Gunn J, Samkoe KS, Hoopes PJ, Feldwisch J, Paulsen KD, Pogue BW. Fluorescent Affibody Molecule Administered In Vivo at a Microdose Level Labels EGFR Expressing Glioma Tumor Regions. Mol Imaging Biol 2017; 19:41-48. [PMID: 27379987 PMCID: PMC5209393 DOI: 10.1007/s11307-016-0980-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Purpose Fluorescence guidance in surgical oncology provides the potential to realize enhanced molecular tumor contrast with dedicated targeted tracers, potentially with a microdose injection level. For most glioma tumors, the blood brain barrier is compromised allowing some exogenous drug/molecule delivery and accumulation for imaging. The aberrant overexpression and/or activation of epidermal growth factor receptor (EGFR) is associated with many types of cancers, including glioblastoma, and so the use of a near-infrared (NIR) fluorescent molecule targeted to the EGFR receptor provides the potential for improving tumor contrast during surgery. Fluorescently labeled affibody molecule (ABY-029) has high EGFR affinity and high potential specificity with reasonably fast plasma clearance. In this study, ABY-29 was evaluated in glioma versus normal brain uptake from intravenous injection at a range of doses, down to a microdose injection level. Procedure Nude rats were inoculated with the U251 human glioma cell line in the brain. Tumors were allowed to grow for 3–4 weeks. ABY-029 fluorescence ex vivo imaging of brain slices was acquired at different time points (1–48 h) and varying injection doses from 25 to 122 μg/kg (from human protein microdose equivalent to five times microdose levels). Results The tumor was most clearly visualized at 1-h post-injection with 8- to 16-fold average contrast relative to normal brain. However, the tumor still could be identified after 48 h. In all cases, the ABY-029 fluorescence appeared to localize preferentially in EGFR-positive regions. Increasing the injected dose from a microdose level to five times, a microdose level increased the signal by 10-fold, and the contrast was from 8 to 16, showing that there was value in doses slightly higher than the microdose restriction. Normal tissue uptake was found to be affected by the tumor size, indicating that edema was a likely factor affecting the expected tumor to normal tissue contrast. Conclusion These results suggest that the NIR-labeled affibody molecules provide an excellent potential to increase surgical visualization of EGFR-positive tumor regions.
Collapse
Affiliation(s)
- Ana Luiza Ribeiro de Souza
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.,CAPES Foundation, Ministry of Education of Brazil, Brasilia, DF, 70040-020, Brazil
| | - Kayla Marra
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Jason Gunn
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.,Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH, 03756, USA
| | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.,Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH, 03756, USA
| | | | - Keith D Paulsen
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA.,Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH, 03756, USA
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, 14 Engineering Drive, Hanover, NH, 03755, USA. .,Department of Surgery, Geisel School of Medicine, Dartmouth College, Lebanon, NH, 03756, USA.
| |
Collapse
|
79
|
Jablonowski LJ, Teraphongphom NT, Wheatley MA. Drug Delivery from a Multi-faceted Ultrasound Contrast Agent: Influence of Shell Composition. Mol Pharm 2017; 14:3448-3456. [DOI: 10.1021/acs.molpharmaceut.7b00451] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lauren J. Jablonowski
- School
of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, United States
| | - Nutte T. Teraphongphom
- School
of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, United States
| | - Margaret A. Wheatley
- School
of Biomedical Engineering, Science, and Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
80
|
Bhandari A, Bansal A, Singh A, Sinha N. Transport of Liposome Encapsulated Drugs in Voxelized Computational Model of Human Brain Tumors. IEEE Trans Nanobioscience 2017; 16:634-644. [PMID: 28796620 DOI: 10.1109/tnb.2017.2737038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
There are many obstacles in the transport of chemotherapeutic drugs to tumor cells that lead to irregular and non-uniform uptake of drugs inside tumors. The study of these transport problems will help with accurate prediction of drug transport and optimizing treatment strategy. To this end, liposome mediated drug delivery has emerged as an excellent anticancer therapy due to its ability to deliver drugs at site of action and reducing the chances of side effects to the healthy tissues. In this paper, a computational fluid dynamics (CFD) model based on realistic vasculature of human brain tumor is presented. This model utilizes dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI) data to account for heterogeneity in tumor vasculature. Porosity of the interstitial space inside the tumor and normal tissue is determined voxel-wise by processing the DCE-MRI images by general tracer kinetic model (GTKM). The CFD model is applied to predict transport of two different types of liposomes (stealth and conventional) in tumors. The amount of accumulated liposomes is compared with accumulated free drug (doxorubicin) in the interstitial space. Simulation results indicate that stealth liposomes accumulate more and remain for longer periods of time in tumors as compared with conventional liposomes and free drug. The present model provides us a qualitative and quantitative examination on the transport and deposition of liposomes as well as free drugs in actual human brain tumors.
Collapse
|
81
|
Lei Q, Wang SB, Hu JJ, Lin YX, Zhu CH, Rong L, Zhang XZ. Stimuli-Responsive "Cluster Bomb" for Programmed Tumor Therapy. ACS NANO 2017; 11:7201-7214. [PMID: 28686414 DOI: 10.1021/acsnano.7b03088] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this paper, mesoporous silica nanoparticle (MSN) loaded with doxorubicin (DOX) and capped with tumor-homing/-penetrating peptide tLyP-1-modified tungsten disulfide quantum dots (WS2-HP) was designed and applied as a stimuli-responsive "Cluster Bomb" for high-performance tumor suppression. The peptide tLyP-1 on the surface can both facilitate the homing of DOX@MSN-WS2-HP to 4T1 tumor and greatly enhance the penetration of WS2-HP in tumor. The benzoic-imine bonds as the linkers between "bomblets" and "dispenser" are stable under normal physical conditions and quite labile at pH 6.8. After arriving at the mild acidic tumor microenvironment, the nanoplatform can rapidly break into two parts: (1) electropositive DOX@MSN-NH2 for efficient chemotherapy on surface tumor cells and (2) small-sized WS2-HP with improved tumor penetrating ability for near-infrared (NIR)-light-triggered photothermal therapy (PTT) among deep-seated tumor cells. Having killed the tumor cells in different depths, DOX@MSN-WS2-HP exhibited significant antitumor effect, which will find great potential in clinical trials.
Collapse
Affiliation(s)
- Qi Lei
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Shi-Bo Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Jing-Jing Hu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Yi-Xiong Lin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Cheng-Hui Zhu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Lei Rong
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry and ‡The Institute for Advanced Studies, Wuhan University , Wuhan 430072, China
| |
Collapse
|
82
|
Zhang M, Ma Y, Wang Z, Han Z, Gao W, Gu Y. Optimizing molecular weight of octyl chitosan as drug carrier for improving tumor therapeutic efficacy. Oncotarget 2017; 8:64237-64249. [PMID: 28969066 PMCID: PMC5609998 DOI: 10.18632/oncotarget.19452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/26/2017] [Indexed: 02/01/2023] Open
Abstract
Macromolecular drug carriers have attracted much attention taking advantage of passive tumor targeting property and excellent biocompatibility. For many biomedical applications, however, the effectiveness of the carriers is insufficient, which complicate further development into clinical use. Here, we systematically investigated the effects of molecular weight (from 1KDa to 300KDa) of macromolecular drug carrier, octyl chitosan, on tumor accumulation and penetration, as well as drug loading and releasing profiles. It was found that the molecular weight of chitosan influenced the cellular uptake and pharmacokinetic behavior of the nanocarriers, which ultimately determined their drug delivery efficiency. Interestingly, increased molecular weight of chitosan decreased its cellular uptake but increased its resident time in blood, which provided ample time for tumor accumulation. Moreover, the molecular weight altered the drug loading capability and release profile. Our results demonstrated that 10KDa octyl chitosan was an ideal candidate for anticancer drug delivery, which could deliver anticancer agent to tumor tissues and release drugs in tumor cells more effectively than those of other molecular weights, and finally result in better therapeutic effect.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaohui Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Weidong Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yueqing Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
83
|
Viger ML, Collet G, Lux J, Nguyen Huu VA, Guma M, Foucault-Collet A, Olejniczak J, Joshi-Barr S, Firestein GS, Almutairi A. Distinct ON/OFF fluorescence signals from dual-responsive activatable nanoprobes allows detection of inflammation with improved contrast. Biomaterials 2017; 133:119-131. [PMID: 28433935 PMCID: PMC5704950 DOI: 10.1016/j.biomaterials.2017.03.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/21/2017] [Accepted: 03/25/2017] [Indexed: 01/08/2023]
Abstract
Visualization of biochemical changes associated with disease is of great clinical significance, as it should allow earlier, more accurate diagnosis than structural imaging, facilitating timely clinical intervention. Herein, we report combining stimuli-responsive polymers and near-infrared fluorescent dyes (emission max: 790 nm) to create robust activatable fluorescent nanoprobes capable of simultaneously detecting acidosis and oxidative stress associated with inflammatory microenvironments. The spectrally-resolved mechanism of fluorescence activation allows removal of unwanted background signal (up to 20-fold reduction) and isolation of a pure activated signal, which enables sensitive and unambiguous localization of inflamed areas; target-to-background ratios reach 22 as early as 3 h post-injection. This new detection platform could have significant clinical impact in early detection of pathologies, individual tailoring of drug therapy, and image-guided tumor resection.
Collapse
Affiliation(s)
- Mathieu L Viger
- Skaggs School of Pharmacy and Pharmaceutical Sciences, KACST - UCSD Center for Excellence in Nanomedicine and Engineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0600, USA
| | - Guillaume Collet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, KACST - UCSD Center for Excellence in Nanomedicine and Engineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0600, USA
| | - Jacques Lux
- UT Southwestern Medical Center, Department of Radiology, 5323 Harry Hines Blvd., Dallas, TX 75390-8896, USA
| | - Viet Anh Nguyen Huu
- Department of Nanoengineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0448, USA
| | - Monica Guma
- Division of Rheumatology, Allergy and Immunology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0656, USA
| | - Alexandra Foucault-Collet
- Skaggs School of Pharmacy and Pharmaceutical Sciences, KACST - UCSD Center for Excellence in Nanomedicine and Engineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0600, USA
| | - Jason Olejniczak
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0332, USA
| | - Shivanjali Joshi-Barr
- Skaggs School of Pharmacy and Pharmaceutical Sciences, KACST - UCSD Center for Excellence in Nanomedicine and Engineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0600, USA
| | - Gary S Firestein
- Division of Rheumatology, Allergy and Immunology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0656, USA
| | - Adah Almutairi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, KACST - UCSD Center for Excellence in Nanomedicine and Engineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093-0600, USA.
| |
Collapse
|
84
|
Mohammed M, Abdel-Gawad E, Awwad S, Kandil E, El-Agamy B. Therapeutic role of a synthesized calcium phosphate nanocomposite material on hepatocarcinogenesis in rats. Biochem Cell Biol 2017; 94:279-88. [PMID: 27276232 DOI: 10.1139/bcb-2015-0135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nanotechnology research is booming worldwide, and the general belief is that medical and biological applications will form the greatest sector of expansion over the next decade. With this in mind, this study was designed to evaluate the therapeutic effects of a synthesized tricalcium phosphate nanocomposite material (nano-TCP) on hepatocarcinoma in a rat model, as initiated with diethylnitrosamine (DEN) and promoted with phenobarbital (PB). Hepatocarcinoma was induced with intraperitoneal injections of DEN (50 mg·(kg body mass)(-1)) 3 times a week for 2 weeks. Three weeks after the last dose of DEN, the rats received PB (0.05 %, w/v) in their drinking water for a further 6 weeks. Nano-TCP (100 mg·(kg body mass)(-1)) was administered intraperitoneally 3 times per week to rats with HCC. At the end of the experimental period, liver samples were collected from all animals for biochemical and histopathological analysis. The degree of DNA fragmentation was analyzed, in addition to immune status, by measuring the levels of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and interleukin-2 (IL-2). The activities of the most important free-radical scavengers of the antioxidant defense system as well as malondialdehyde (MDA) content and liver enzymes were measured. The levels of hepatic heat shock protein-70 (HSP-70), caspase-3, and metalloproteinase-9 were also measured as markers for inflammation and apoptosis. Histopathological examination of liver tissue was performed. The results revealed the potent efficacy of nano-TCP in repairing the fragmented DNA and ameliorating most of the investigated parameters by significant elevation in the levels of hepatic alanine aminotransferase (ALT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities. On the other hand, there was a significant decrease in hepatic gamma-glutamyl transpeptidase (γ-GT), MDA, IL-2, IFN-γ, TNF-α, matrix metalloproteinase-9 (MMP-9), HSP-70, and caspase-3 levels upon treatment. The findings form histopathological examination of the liver tissues agreed with the biochemical results and confirmed the difference between the control and treatment groups. In conclusion, nano-TCP succeeded in treating hepatocarcinoma efficiently, and presents a new hope for patients to get safe, fast, and effective treatment.
Collapse
Affiliation(s)
- Magdy Mohammed
- a Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Eman Abdel-Gawad
- b Radioisotopes Department, Atomic Energy Authority, Cairo, Egypt
| | | | - Eman Kandil
- a Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Basma El-Agamy
- a Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
85
|
Tan L, McGarry MDJ, Van Houten EEW, Ji M, Solamen L, Zeng W, Weaver JB, Paulsen KD. A numerical framework for interstitial fluid pressure imaging in poroelastic MRE. PLoS One 2017; 12:e0178521. [PMID: 28586393 PMCID: PMC5460821 DOI: 10.1371/journal.pone.0178521] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/15/2017] [Indexed: 11/18/2022] Open
Abstract
A numerical framework for interstitial fluid pressure imaging (IFPI) in biphasic materials is investigated based on three-dimensional nonlinear finite element poroelastic inversion. The objective is to reconstruct the time-harmonic pore-pressure field from tissue excitation in addition to the elastic parameters commonly associated with magnetic resonance elastography (MRE). The unknown pressure boundary conditions (PBCs) are estimated using the available full-volume displacement data from MRE. A subzone-based nonlinear inversion (NLI) technique is then used to update mechanical and hydrodynamical properties, given the appropriate subzone PBCs, by solving a pressure forward problem (PFP). The algorithm was evaluated on a single-inclusion phantom in which the elastic property and hydraulic conductivity images were recovered. Pressure field and material property estimates had spatial distributions reflecting their true counterparts in the phantom geometry with RMS errors around 20% for cases with 5% noise, but degraded significantly in both spatial distribution and property values for noise levels > 10%. When both shear moduli and hydraulic conductivity were estimated along with the pressure field, property value error rates were as high as 58%, 85% and 32% for the three quantities, respectively, and their spatial distributions were more distorted. Opportunities for improving the algorithm are discussed.
Collapse
Affiliation(s)
- Likun Tan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Matthew D. J. McGarry
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States of America
| | - Elijah E. W. Van Houten
- Department of Mechanical Engineering, University de Sherbrooke, Sherbrooke, Quebec J1K 2R1, Canada
| | - Ming Ji
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America
| | - Ligin Solamen
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Wei Zeng
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - John B. Weaver
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Department of Radiology, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 United States of America
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 United States of America
| |
Collapse
|
86
|
Bhandari A, Bansal A, Singh A, Sinha N. Perfusion kinetics in human brain tumor with DCE-MRI derived model and CFD analysis. J Biomech 2017. [PMID: 28623038 DOI: 10.1016/j.jbiomech.2017.05.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer is one of the leading causes of death all over the world. Among the strategies that are used for cancer treatment, the effectiveness of chemotherapy is often hindered by factors such as irregular and non-uniform uptake of drugs inside tumor. Thus, accurate prediction of drug transport and deposition inside tumor is crucial for increasing the effectiveness of chemotherapeutic treatment. In this study, a computational model of human brain tumor is developed that incorporates dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI) data into a voxelized porous media model. The model takes into account realistic transport and perfusion kinetics parameters together with realistic heterogeneous tumor vasculature and accurate arterial input function (AIF), which makes it patient specific. The computational results for interstitial fluid pressure (IFP), interstitial fluid velocity (IFV) and tracer concentration show good agreement with the experimental results. The computational model can be extended further for predicting the deposition of chemotherapeutic drugs in tumor environment as well as selection of the best chemotherapeutic drug for a specific patient.
Collapse
Affiliation(s)
- A Bhandari
- Department of Mechanical Engineering, Indian Institute of Technology, Kanpur 208016, India
| | - A Bansal
- Department of Mechanical and Industrial Engineering, Indian Institute of Technology, Roorkee 247677, India
| | - A Singh
- Centre for Biomedical Engineering, Indian Institute of Technology, Delhi 110016, India; Department of Biomedical Engineering, All India Institute of Medical Sciences, Delhi 110016, India
| | - N Sinha
- Department of Mechanical Engineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
87
|
Cao P, Mooney R, Tirughana R, Abidi W, Aramburo S, Flores L, Gilchrist M, Nwokafor U, Haber T, Tiet P, Annala AJ, Han E, Dellinger T, Aboody KS, Berlin JM. Intraperitoneal Administration of Neural Stem Cell-Nanoparticle Conjugates Targets Chemotherapy to Ovarian Tumors. Bioconjug Chem 2017; 28:1767-1776. [PMID: 28453256 DOI: 10.1021/acs.bioconjchem.7b00237] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ovarian cancer is particularly aggressive once it has metastasized to the abdominal cavity (stage III). Intraperitoneal (IP) as compared to intravenous (IV) administration of chemotherapy improves survival for stage III ovarian cancer, demonstrating that concentrating chemotherapy at tumor sites has therapeutic benefit; unfortunately, IP therapy also increases toxic side effects, thus preventing its completion in many patients. The ability to target chemotherapy selectively to ovarian tumors while sparing normal tissue would improve efficacy and decrease toxicities. We have previously shown that tumor-tropic neural stem cells (NSCs) dramatically improve the intratumoral distribution of nanoparticles (NPs) when given intracerebrally near an orthotopic brain tumor or into a flank xenograft tumor. Here, we show that NPs either conjugated to the surface of NSCs or loaded within the cells are selectively delivered to and distributed within ovarian tumors in the abdominal cavity following IP injection, with no evidence of localization to normal tissue. IP administration is significantly more effective than IV administration, and NPs carried by NSCs show substantially deeper penetration into tumors than free NPs. The NSCs and NPs target and localize to ovarian tumors within 1 h of administration. Pt-loaded silica NPs (SiNP[Pt]) were developed that can be transported in NSCs, and it was found that the NSC delivery of SiNP[Pt] (NSC-SiNP[Pt]) results in higher levels of Pt in tumors as compared to free drug or SiNP[Pt]. To the best of our knowledge, this work represents the first demonstration that cells given IP can target the delivery of drug-loaded NPs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Ernest Han
- Department of Surgery, City of Hope , 1500 East Duarte Road, Duarte, California 91010, United States
| | - Thanh Dellinger
- Department of Surgery, City of Hope , 1500 East Duarte Road, Duarte, California 91010, United States
| | | | | |
Collapse
|
88
|
Cao Z, Wang X, Cheng X, Wang J, Tang R. In vitro and in vivo antitumor study of folic acid-conjugated carboxymethyl chitosan and phenylboronic acid–based nanoparticles. INT J POLYM MATER PO 2017. [DOI: 10.1080/00914037.2016.1252346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Zhipeng Cao
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Xu Cheng
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Jun Wang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui University, Hefei, China
| |
Collapse
|
89
|
Sun Q, Zhou Z, Qiu N, Shen Y. Rational Design of Cancer Nanomedicine: Nanoproperty Integration and Synchronization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606628. [PMID: 28234430 DOI: 10.1002/adma.201606628] [Citation(s) in RCA: 733] [Impact Index Per Article: 91.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Indexed: 05/21/2023]
Abstract
Current cancer nanomedicines can only mitigate adverse effects but fail to enhance therapeutic efficacies of anticancer drugs. Rational design of next-generation cancer nanomedicines should aim to enhance their therapeutic efficacies. Taking this into account, this review first analyzes the typical cancer-drug-delivery process of an intravenously administered nanomedicine and concludes that the delivery involves a five-step CAPIR cascade and that high efficiency at every step is critical to guarantee high overall therapeutic efficiency. Further analysis shows that the nanoproperties needed in each step for a nanomedicine to maximize its efficiency are different and even opposing in different steps, particularly what the authors call the PEG, surface-charge, size and stability dilemmas. To resolve those dilemmas in order to integrate all needed nanoproperties into one nanomedicine, stability, surface and size nanoproperty transitions (3S transitions for short) are proposed and the reported strategies to realize these transitions are comprehensively summarized. Examples of nanomedicines capable of the 3S transitions are discussed, as are future research directions to design high-performance cancer nanomedicines and their clinical translations.
Collapse
Affiliation(s)
- Qihang Sun
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| |
Collapse
|
90
|
Abstract
Targeted cancer nanotherapeutics offers numerous opportunities for the selective uptake of toxic chemotherapies within tumors and cancer cells. The unique properties of nanoparticles, such as their small size, large surface-to-volume ratios, and the ability to achieve multivalency of targeting ligands on their surface, provide superior advantages for nanoparticle-based drug delivery to a variety of cancers. This review highlights various key concepts in the design of targeted nanotherapeutics for cancer therapy, and discusses physicochemical parameters affecting nanoparticle targeting, along with recent developments for cancer-targeted nanomedicines.
Collapse
Affiliation(s)
| | | | - Joseph Kaplinsky
- Department of Micro and Nanotechnology, DTU Nanotech, Technical University of Denmark, Produktionstorvet, 2800, Kongens Lyngby, Denmark
| | - Nazila Kamaly
- Department of Micro and Nanotechnology, DTU Nanotech, Technical University of Denmark, Produktionstorvet, 2800, Kongens Lyngby, Denmark.
| |
Collapse
|
91
|
Abramczyk H, Brozek-Pluska B, Surmacki J, Tondusson M, Freysz E. Photostability of biological systems—Femtosecond dynamics of zinc tetrasulfonated phthalocyanine at cancerous and noncancerous human Breast tissues. J Photochem Photobiol A Chem 2017. [DOI: 10.1016/j.jphotochem.2016.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
92
|
To exploit the tumor microenvironment: Since the EPR effect fails in the clinic, what is the future of nanomedicine? J Control Release 2016; 244:108-121. [DOI: 10.1016/j.jconrel.2016.11.015] [Citation(s) in RCA: 908] [Impact Index Per Article: 100.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/26/2016] [Accepted: 11/07/2016] [Indexed: 11/22/2022]
|
93
|
Alkholief M, Campbell RB. Investigating the role of mucin in the delivery of nanoparticles to cellular models of human cancer disease: an in vitro study. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1291-302. [DOI: 10.1016/j.nano.2016.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 10/22/2022]
|
94
|
Duvillard C, Romanet P, Cosmidis A, Beaudouin N, Chauffert B. Phase 2 Study of Intratumoral Cisplatin and Epinephrine Treatment for Locally Recurrent Head and Neck Tumors. Ann Otol Rhinol Laryngol 2016; 113:229-33. [PMID: 15053208 DOI: 10.1177/000348940411300312] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Local relapses of head and neck tumors are not often eligible for surgical and/or radiotherapy retreatment, and the efficacy of systemic chemotherapy is poor. A greater accumulation and efficacy of anticancer drugs with lower systemic toxicity is theoretically obtained with intratumoral chemotherapy. In experimental studies, epinephrine has been shown to increase the concentration and antitumor effect of intratumoral cisplatin. Fourteen patients with locally recurrent head and neck tumors (median age, 58.7 years) were included in this phase 2 trial. Recurrent tumors (squamous cell carcinomas) were located on the tongue, oral pharynx, or cervical nodes. Prior therapy was surgery and/or radiotherapy with or without intravenous chemotherapy. Inclusion criteria included an Eastern Cooperative Oncology Group/World Health Organization performance status of 0, 1, or 2, an anticipated survival of >3 months, adequate cardiac, kidney, liver, and bone marrow function, and no coagulopathy or carotid invasion. Fifty intratumoral injections of cisplatin-epinephrine (average, 3.6 injections per patient; range, 1 to 5 injections) were given to the 14 patients from November 1998 to July 2000. Patients were treated with cisplatin (1 mg/mL; maximum dose, 50 mg) at an injection volume corresponding to the tumor volume (1 mL/cm3 of tumor; maximum volume, 50 mL). Epinephrine was added at a concentration of 0.02 mg/mL. Intratumoral injections were repeated every 2 to 3 weeks at different locations in the tumors to obtain a homogeneous distribution. Tumor response was evaluated by clinical examination and computed tomography. Eight objective responses were registered among the 14 patients. Four were complete responses, and 4 were partial responses. The average time to disease progression was 11.5 ± 8.9 weeks. Local adverse effects were transient pain, swelling, and erythema at the site of the injection. No nephrotoxicity, neurotoxicity, or ototoxicity was observed. Intratumoral injection of cisplatin and epinephrine in an aqueous solution has a definite antitumor activity in recurrent head and neck cancer with acceptable local tolerance and no major systemic toxic effects except for transient tachycardia and high blood pressure at the time of injection.
Collapse
Affiliation(s)
- Christian Duvillard
- Department of Otolaryngology-Head and Neck Surgery, Hôpital Général, Dijon, France
| | | | | | | | | |
Collapse
|
95
|
Abstract
Chemotherapy has been the main modality of treatment for cancer patients; however, its success rate remains low, primarily due to limited accessibility of drugs to the tumor tissue, their intolerable toxicity, development of multi-drug resistance, and the dynamic heterogeneous biology of the growing tumors. Better understanding of tumor biology in recent years and new targeted drug delivery approaches that are being explored using different nanosystems and bioconjugates provide optimism in developing successful cancer therapy. This article reviews the possibilities and challenges for targeted drug delivery in cancer therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/pharmacology
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Drug Carriers
- Drug Delivery Systems
- Drug Resistance, Multiple
- Endothelium, Vascular/cytology
- Endothelium, Vascular/pathology
- Folate Receptors, GPI-Anchored
- Folic Acid/metabolism
- Gene Transfer Techniques
- Humans
- Immunotherapy
- Immunotoxins/chemistry
- Ligands
- Magnetics
- Mice
- Nanostructures/chemistry
- Neoplasms/genetics
- Neoplasms/therapy
- Neovascularization, Pathologic
- Oleic Acid/chemistry
- Receptors, Cell Surface/metabolism
- Receptors, LDL/chemistry
- Recombinant Fusion Proteins/chemistry
- Time Factors
- Trastuzumab
Collapse
Affiliation(s)
- Jaspreet K Vasir
- Department of Pharmaceutical Sciences, College of Pharmacy, 986025 Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | | |
Collapse
|
96
|
Tumor Heterogeneity, Single-Cell Sequencing, and Drug Resistance. Pharmaceuticals (Basel) 2016; 9:ph9020033. [PMID: 27322289 PMCID: PMC4932551 DOI: 10.3390/ph9020033] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 01/10/2023] Open
Abstract
Tumor heterogeneity has been compared with Darwinian evolution and survival of the fittest. The evolutionary ecosystem of tumors consisting of heterogeneous tumor cell populations represents a considerable challenge to tumor therapy, since all genetically and phenotypically different subpopulations have to be efficiently killed by therapy. Otherwise, even small surviving subpopulations may cause repopulation and refractory tumors. Single-cell sequencing allows for a better understanding of the genomic principles of tumor heterogeneity and represents the basis for more successful tumor treatments. The isolation and sequencing of single tumor cells still represents a considerable technical challenge and consists of three major steps: (1) single cell isolation (e.g., by laser-capture microdissection), fluorescence-activated cell sorting, micromanipulation, whole genome amplification (e.g., with the help of Phi29 DNA polymerase), and transcriptome-wide next generation sequencing technologies (e.g., 454 pyrosequencing, Illumina sequencing, and other systems). Data demonstrating the feasibility of single-cell sequencing for monitoring the emergence of drug-resistant cell clones in patient samples are discussed herein. It is envisioned that single-cell sequencing will be a valuable asset to assist the design of regimens for personalized tumor therapies based on tumor subpopulation-specific genetic alterations in individual patients.
Collapse
|
97
|
Abstract
Many patients with lung cancer, breast cancer, and melanoma develop brain metastases that are resistant to conventional therapy. The median survival for untreated patients is 1 to 2 months, which may be extended to 6 months with surgery, radiotherapy, and chemotherapy. The outcome of metastasis depends on multiple interactions of unique metastatic cells with host homeostatic mechanisms which the tumor cells exploit for their survival and proliferation. The blood-brain barrier is leaky in metastases that are larger than 0.5-mm diameter because of production of vascular endothelial growth factor by metastatic cells. Brain metastases are surrounded and infiltrated by microglia and activated astrocytes. The interaction with astrocytes leads to up-regulation of multiple genes in the metastatic cells, including several survival genes that are responsible for the increased resistance of tumor cells to cytotoxic drugs. These findings substantiate the importance of the "seed and soil" hypothesis and that successful treatment of brain metastases must include targeting of the organ microenvironment.
Collapse
|
98
|
Nanomicelles based on a boronate ester-linked diblock copolymer as the carrier of doxorubicin with enhanced cellular uptake. Colloids Surf B Biointerfaces 2016; 141:318-326. [DOI: 10.1016/j.colsurfb.2016.01.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
|
99
|
Kirui DK, Ferrari M. Intravital Microscopy Imaging Approaches for Image-Guided Drug Delivery Systems. Curr Drug Targets 2016; 16:528-41. [PMID: 25901526 DOI: 10.2174/1389450116666150330114030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/10/2014] [Accepted: 03/13/2015] [Indexed: 12/31/2022]
Abstract
Rapid technical advances in the field of non-linear microscopy have made intravital microscopy a vital pre-clinical tool for research and development of imaging-guided drug delivery systems. The ability to dynamically monitor the fate of macromolecules in live animals provides invaluable information regarding properties of drug carriers (size, charge, and surface coating), physiological, and pathological processes that exist between point-of-injection and the projected of site of delivery, all of which influence delivery and effectiveness of drug delivery systems. In this Review, we highlight how integrating intravital microscopy imaging with experimental designs (in vitro analyses and mathematical modeling) can provide unique information critical in the design of novel disease-relevant drug delivery platforms with improved diagnostic and therapeutic indexes. The Review will provide the reader an overview of the various applications for which intravital microscopy has been used to monitor the delivery of diagnostic and therapeutic agents and discuss some of their potential clinical applications.
Collapse
Affiliation(s)
| | - Mauro Ferrari
- Houston Methodist Research Institute, Department of NanoMedicine, 6670 Bertner Avenue, MS R8-460, Houston, TX 77030, USA.
| |
Collapse
|
100
|
Endostatin combined with platinum-based chemo-radiotherapy for advanced non-small cell lung cancer. Cell Biochem Biophys 2016; 71:571-7. [PMID: 25209742 DOI: 10.1007/s12013-014-0236-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This retrospective study was conducted to determine whether endostatin improves the efficacy and safety of platinum-based chemotherapy administered via both intravenous and arterial infusions for stage III/IV non-small-cell lung cancer (NSCLC). Seventy one patients with confirmed pathological or cytological diagnosis of NSCLC from January 2008 to March 2013 were enrolled for the study. Patients received three different therapeutic regimens until disease progression or an intolerable toxicity was evidenced. Group C received chemotherapy administered by intravenous injection and subsequent radiotherapy. Group IC received chemotherapy by intravenous injection and arterial infusion plus radiotherapy. Group IC + E patients were treated with chemotherapy plus Endostatin injected arterially and intravenously and subsequent radiotherapy. Group IC + E showed a progression-free survival (PFS) of 12 months as compared to 7 months of group C, a significant increase indeed (p = 0.037). Likewise, the overall survival time higher in Group IC + E and Group IC was compared to Group C (p = 0.001; p = 0.004, respectively). The adverse or toxic side effects exhibited by Group IC + E were not significantly different from either Group IC or Group C. Endostatin administered in combination with chemotherapeutic agents via both intravenous injection and arterial infusion enhanced the PFS and OS in advanced NSCLC without increasing the risk of toxicity.
Collapse
|