1
|
Haque M, Shakil MS, Mahmud KM. The Promise of Nanoparticles-Based Radiotherapy in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15061892. [PMID: 36980778 PMCID: PMC10047050 DOI: 10.3390/cancers15061892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Radiation has been utilized for a long time for the treatment of cancer patients. However, radiotherapy (RT) has many constraints, among which non-selectivity is the primary one. The implementation of nanoparticles (NPs) with RT not only localizes radiation in targeted tissue but also provides significant tumoricidal effect(s) compared to radiation alone. NPs can be functionalized with both biomolecules and therapeutic agents, and their combination significantly reduces the side effects of RT. NP-based RT destroys cancer cells through multiple mechanisms, including ROS generation, which in turn damages DNA and other cellular organelles, inhibiting of the DNA double-strand damage-repair system, obstructing of the cell cycle, regulating of the tumor microenvironment, and killing of cancer stem cells. Furthermore, such combined treatments overcome radioresistance and drug resistance to chemotherapy. Additionally, NP-based RT in combined treatments have shown synergistic therapeutic benefit(s) and enhanced the therapeutic window. Furthermore, a combination of phototherapy, i.e., photodynamic therapy and photothermal therapy with NP-based RT, not only reduces phototoxicity but also offers excellent therapeutic benefits. Moreover, using NPs with RT has shown promise in cancer treatment and shown excellent therapeutic outcomes in clinical trials. Therefore, extensive research in this field will pave the way toward improved RT in cancer treatment.
Collapse
Affiliation(s)
- Munima Haque
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Md Salman Shakil
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Kazi Mustafa Mahmud
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| |
Collapse
|
2
|
Zhang D, He J, Zhou M. Radiation-assisted strategies provide new perspectives to improve the nanoparticle delivery to tumor. Adv Drug Deliv Rev 2023; 193:114642. [PMID: 36529190 DOI: 10.1016/j.addr.2022.114642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/07/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022]
Abstract
Nanoparticles (NPs), with advantages in tumor targeting, have been extensively developed for anticancer treatment. However, the delivery efficacy of NPs tends to be heterogeneous in clinical research. Surprisingly, a traditional cancer treatment, radiotherapy (radiation), has been observed with the potential to improve the delivery of NPs by influencing the features of the tumor microenvironment, which provides new perspectives to overcome the barriers in the NPs delivery. Since the effect of radiation can also be enhanced by versatile NPs, these findings of radiation-assisted NPs delivery suggest innovative strategies combining radiotherapy with nanotherapeutics. This review summarizes the research on the delivery and therapeutic efficacy of NPs that are improved by radiation, focusing on relative mechanisms and existing challenges and opportunities.
Collapse
Affiliation(s)
- Dongxiao Zhang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Jian He
- The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
3
|
Boateng F, Ngwa W. Delivery of Nanoparticle-Based Radiosensitizers for Radiotherapy Applications. Int J Mol Sci 2019; 21:ijms21010273. [PMID: 31906108 PMCID: PMC6981554 DOI: 10.3390/ijms21010273] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/21/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Nanoparticle-based radiosensitization of cancerous cells is evolving as a favorable modality for enhancing radiotherapeutic ratio, and as an effective tool for increasing the outcome of concomitant chemoradiotherapy. Nevertheless, delivery of sufficient concentrations of nanoparticles (NPs) or nanoparticle-based radiosensitizers (NBRs) to the targeted tumor without or with limited systemic side effects on healthy tissues/organs remains a challenge that many investigators continue to explore. With current systemic intravenous delivery of a drug, even targeted nanoparticles with great prospect of reaching targeted distant tumor sites, only a portion of the administered NPs/drug dosage can reach the tumor, despite the enhanced permeability and retention (EPR) effect. The rest of the targeted NPs/drug remain in systemic circulation, resulting in systemic toxicity, which can decrease the general health of patients. However, the dose from ionizing radiation is generally delivered across normal tissues to the tumor cells (especially external beam radiotherapy), which limits dose escalation, making radiotherapy (RT) somewhat unsafe for some diseased sites despite the emerging development in RT equipment and technologies. Since radiation cannot discriminate healthy tissue from diseased tissue, the radiation doses delivered across healthy tissues (even with nanoparticles delivered via systemic administration) are likely to increase injury to normal tissues by accelerating DNA damage, thereby creating free radicals that can result in secondary tumors. As a result, other delivery routes, such as inhalation of nanoparticles (for lung cancers), localized delivery via intratumoral injection, and implants loaded with nanoparticles for local radiosensitization, have been studied. Herein, we review the current NP delivery techniques; precise systemic delivery (injection/infusion and inhalation), and localized delivery (intratumoral injection and local implants) of NBRs/NPs. The current challenges, opportunities, and future prospects for delivery of nanoparticle-based radiosensitizers are also discussed.
Collapse
Affiliation(s)
- Francis Boateng
- TIDTAC LLC, Orlando, FL 32828, USA
- Correspondence: ; Tel.: +1-7745264723
| | - Wilfred Ngwa
- TIDTAC LLC, Orlando, FL 32828, USA
- Department of Physics and Applied Physics, University of Massachusetts Lowell Lowell, MA 01854, USA
- Department of Radiation Oncology, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Radiation Oncology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Liu J, Zhang W, Kumar A, Rong X, Yang W, Chen H, Xie J, Wang Y. Acridine Orange Encapsulated Mesoporous Manganese Dioxide Nanoparticles to Enhance Radiotherapy. Bioconjug Chem 2019; 31:82-92. [PMID: 31809019 DOI: 10.1021/acs.bioconjchem.9b00751] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Manganese dioxide (MnO2) nanoparticles are a promising type of radiosensitizer for they can catalyze H2O2 decomposition to produce O2. Combining MnO2 nanoparticles with conventional, small molecule radiosensitizers would further enhance radiotherapy (RT) efficacy due to complementary mechanisms of action. However, solid MnO2 nanoparticles are suboptimal at drug loading, limiting the related progress. Herein we report a facile method to synthesize mesoporous MnO2 (mMnO2) nanoparticles, which can efficiently encapsulate small molecule therapeutics. In particular, we found that acridine orange (AO), a small molecule radiosensitizer, can be loaded onto mMnO2 nanoparticles at very high efficiency and released to the surroundings in a controlled fashion. We show that mMnO2 nanoparticles can efficiently produce O2 inside cells. This, together with AO-induced DNA damage, significantly enhances RT outcomes, which was validated both in vitro and in vivo. Meanwhile, mMnO2 nanoparticles slowly degrade in acidic environments to release Mn2+, providing a facile way to keep track of the nanoparticles through magnetic resonance imaging (MRI). Overall, our studies suggest mMnO2 as a promising nanoplatform that can be exploited to produce composite radiosensitizers for RT.
Collapse
Affiliation(s)
| | - Weizhong Zhang
- Department of Chemistry , University of Georgia , Athens , Georgia 30602 , United States
| | - Anil Kumar
- Department of Chemistry , University of Georgia , Athens , Georgia 30602 , United States
| | | | - Wei Yang
- Department of Chemistry , University of Georgia , Athens , Georgia 30602 , United States
| | - Hongmin Chen
- State Key Laboratory of Molecular Vaccinology and Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Fujian 361102 , China
| | - Jin Xie
- Department of Chemistry , University of Georgia , Athens , Georgia 30602 , United States
| | | |
Collapse
|
5
|
Ionizing radiation attracts tumor targeting and apoptosis by radiotropic lysyl oxidase traceable nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 24:102141. [PMID: 31830613 DOI: 10.1016/j.nano.2019.102141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 10/23/2019] [Accepted: 11/29/2019] [Indexed: 02/05/2023]
Abstract
Lysyl oxidase (LOX) is a cell-secreted amine oxidase that crosslinks collagen and elastin in extracellular microenvironment. LOX-traceable nanoparticles (LOXab-NPs) consisting of LOX antibodies (LOXab) and paclitaxel, can accumulate at high concentrations at radiation-treated target sites, as a tumor-targeting drug carrier for chemotherapy. Tumor-targeting and anticancer effects of PLGA based LOXab-NPs in vitro and in vivo were evaluated at radiation-targeted site. In the in vivo A549 lung carcinoma xenograft model, we showed highly specific tumor targeting (above 7.0 times higher) of LOXab-NPs on irradiated tumors. Notably, systemically administered NPs delayed tumor growth, reducing tumor volumes by more than 2 times compared with non-irradiated groups (222% vs. >500%) over 2 weeks. Radiotropic LOXab-NPs can serve as chemotherapeutic vehicles for combined targeted chemo-radiotherapy in clinical oncology.
Collapse
|
6
|
|
7
|
Sanoff HK, Moon DH, Moore DT, Boles J, Bui C, Blackstock W, O'Neil BH, Subramaniam S, McRee AJ, Carlson C, Lee MS, Tepper JE, Wang AZ. Phase I/II trial of nano-camptothecin CRLX101 with capecitabine and radiotherapy as neoadjuvant treatment for locally advanced rectal cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:189-195. [PMID: 30858085 DOI: 10.1016/j.nano.2019.02.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/13/2019] [Accepted: 02/24/2019] [Indexed: 12/19/2022]
Abstract
CRLX101 is a nanoparticle-drug conjugate with a camptothecin payload. We assessed the toxicity and pathologic complete response (pCR) rate of CRLX101 with standard neoadjuvant chemoradiotherapy (CRT) in locally advanced rectal cancer. A single-arm study was conducted with a 3 + 3 dose escalation phase Ib followed by phase II at the maximum tolerated dose (MTD). Thirty-two patients were enrolled with 29 (91%) patients having T3/4 and 26 (81%) N1/2 disease. In phase Ib, no patient experienced a dose limiting toxicity (DLT) with every other week dosing, while 1/9 patients experienced a DLT with weekly dosing. The weekly MTD was identified as 15 mg/m2. The most common grade 3-4 toxicity was lymphopenia, with only 1 grade 4 event. pCR was achieved in 6/32 (19%) patients overall and 2/6 (33%) patients at the weekly MTD. CRLX101 at 15 mg/m2 weekly with neoadjuvant CRT is a feasible combination strategy with an excellent toxicity profile. Clinicaltrials.gov registration NCT02010567.
Collapse
Affiliation(s)
- Hanna K Sanoff
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Dominic H Moon
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC
| | - Dominic T Moore
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | | | | | | | | | | | - Autumn J McRee
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Cheryl Carlson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Michael S Lee
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Joel E Tepper
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC
| | - Andrew Z Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC; Laboratory of Nano- and Translational Medicine, Carolina Center for Cancer Nanotechnology Excellent, Carolina Institute of Nanomedicine, University of North Carolina, Chapel Hill, NC.
| |
Collapse
|
8
|
Motea EA, Huang X, Singh N, Kilgore JA, Williams NS, Xie XJ, Gerber DE, Beg MS, Bey EA, Boothman DA. NQO1-dependent, Tumor-selective Radiosensitization of Non-small Cell Lung Cancers. Clin Cancer Res 2019; 25:2601-2609. [PMID: 30617135 DOI: 10.1158/1078-0432.ccr-18-2560] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Development of tumor-specific therapies for the treatment of recalcitrant non-small cell lung cancers (NSCLC) is urgently needed. Here, we investigated the ability of β-lapachone (β-lap, ARQ761 in clinical form) to selectively potentiate the effects of ionizing radiation (IR, 1-3 Gy) in NSCLCs that overexpress NAD(P)H:Quinone Oxidoreductase 1 (NQO1). EXPERIMENTAL DESIGN The mechanism of lethality of low-dose IR in combination with sublethal doses of β-lap was evaluated in NSCLC lines in vitro and validated in subcutaneous and orthotopic xenograft models in vivo. Pharmacokinetics and pharmacodynamics (PK/PD) studies comparing single versus cotreatments were performed to validate therapeutic efficacy and mechanism of action. RESULTS β-Lap administration after IR treatment hyperactivated PARP, greatly lowered NAD+/ATP levels, and increased double-strand break (DSB) lesions over time in vitro. Radiosensitization of orthotopic, as well as subcutaneous, NSCLCs occurred with high apparent cures (>70%), even though 1/8 β-lap doses reach subcutaneous versus orthotopic tumors. No methemoglobinemia or long-term toxicities were noted in any normal tissues, including mouse liver that expresses the highest level of NQO1 (∼12 units) of any normal tissue. PK/PD responses confirm that IR + β-lap treatments hyperactivate PARP activity, greatly lower NAD+/ATP levels, and dramatically inhibit DSB repair in exposed NQO1+ cancer tissue, whereas low NQO1 levels and high levels of catalase in associated normal tissue were protective. CONCLUSIONS Our data suggest that combination of sublethal doses of β-lap and IR is a viable approach to selectively treat NQO1-overexpressing NSCLC and warrant a clinical trial using low-dose IR + β-lap against patients with NQO1+ NSCLCs.
Collapse
Affiliation(s)
- Edward A Motea
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xiumei Huang
- Department of Radiation Oncology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Naveen Singh
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jessica A Kilgore
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Noelle S Williams
- Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xian-Jin Xie
- Department of Biostatistics, UT Southwestern Medical Center, Dallas, Texas
| | - David E Gerber
- Department of Internal Medicine, Division of Hematology-Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Muhammad S Beg
- Department of Internal Medicine, Division of Hematology-Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Erik A Bey
- Department of Pharmaceutical Sciences, West Virginia University Cancer Institute, Morgantown, West Virginia.
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
9
|
Chen X, Song J, Chen X, Yang H. X-ray-activated nanosystems for theranostic applications. Chem Soc Rev 2019; 48:3073-3101. [PMID: 31106315 DOI: 10.1039/c8cs00921j] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
X-rays are widely applied in clinical medical facilities for radiotherapy (RT) and biomedical imaging. However, the sole use of X-rays for cancer treatment leads to insufficient radiation energy deposition due to the low X-ray attenuation coefficients of living tissues and organs, producing unavoidable excessive radiation doses with serious side effects to healthy body parts. Over the past decade, developments in materials science and nanotechnology have led to rapid progress in the field of X-ray-activated tumor-targeting nanosystems, which are able to tackle even systemic tumors and relieve the burden of exposure to large radiation doses. Additionally, novel imaging contrast agents and techniques have also been developed. In comparison with conventional external light sources (e.g., near infrared), the X-ray technique is ideal for the activation of nanosystems for cancer treatment and biomedical imaging applications due to its nearly unlimited penetration depth in living tissues and organisms. In this review, we systematically describe the interaction mechanisms between X-rays and nanosystems, and provide an overview of X-ray-sensitive materials and the recent progress on X-ray-activated nanosystems for cancer-associated theranostic applications.
Collapse
Affiliation(s)
- Xiaofeng Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.
| | | | | | | |
Collapse
|
10
|
Remotely Triggered Nanotheranostics. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
11
|
Stapleton S, Dunne M, Milosevic M, Tran CW, Gold MJ, Vedadi A, Mckee TD, Ohashi PS, Allen C, Jaffray DA. Radiation and Heat Improve the Delivery and Efficacy of Nanotherapeutics by Modulating Intratumoral Fluid Dynamics. ACS NANO 2018; 12:7583-7600. [PMID: 30004666 DOI: 10.1021/acsnano.7b06301] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Nanomedicine drug delivery systems are capable of transporting significant payloads to solid tumors. However, only a modest increase in antitumor efficacy relative to the standard of care has been observed. In this study, we demonstrate that a single dose of radiation or mild hyperthermia can substantially improve tumor uptake and distribution of nanotherapeutics, resulting in improved treatment efficacy. The delivery of nanomedicine was driven by a reduction in interstitial fluid pressure (IFP) and small perturbation of steady-state fluid flow. The transient effects on fluid dynamics in tumors with high IFP was also shown to dominate over immune cell endocytic capacity, another mechanism suspected of improving drug delivery. Furthermore, we demonstrate the specificity of this mechanism by showing that delivery of nanotherapeutics to low IFP tumors with high leukocyte infiltration does not benefit from pretreatment with radiation or heat. These results demonstrate that focusing on small perturbations to steady-state fluid dynamics, rather than large sustained effects or uncertain immune cell recruitment strategies, can impart a vulnerability to tumors with high IFP and enhance nanotherapeutic drug delivery and treatment efficacy.
Collapse
Affiliation(s)
- Shawn Stapleton
- Department of Medical Biophysics , University of Toronto , Toronto , ON M5G 1L7 , Canada
| | - Michael Dunne
- Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , ON M5S 3M2 , Canada
| | - Michael Milosevic
- Department of Radiation Oncology , University of Toronto , Toronto , ON M5S 3E2 , Canada
| | - Charles W Tran
- Department of Immunology , University of Toronto , Toronto , ON M5S 1A1 , Canada
| | | | | | | | - Pamela S Ohashi
- Department of Immunology , University of Toronto , Toronto , ON M5S 1A1 , Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy , University of Toronto , Toronto , ON M5S 3M2 , Canada
| | - David A Jaffray
- Department of Medical Biophysics , University of Toronto , Toronto , ON M5G 1L7 , Canada
- Department of Radiation Oncology , University of Toronto , Toronto , ON M5S 3E2 , Canada
- Techna Institute , University Health Network , Toronto , ON M5G 1L5 , Canada
| |
Collapse
|
12
|
Hartshorn CM, Bradbury MS, Lanza GM, Nel AE, Rao J, Wang AZ, Wiesner UB, Yang L, Grodzinski P. Nanotechnology Strategies To Advance Outcomes in Clinical Cancer Care. ACS NANO 2018; 12:24-43. [PMID: 29257865 PMCID: PMC6589353 DOI: 10.1021/acsnano.7b05108] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Ongoing research into the application of nanotechnology for cancer treatment and diagnosis has demonstrated its advantages within contemporary oncology as well as its intrinsic limitations. The National Cancer Institute publishes the Cancer Nanotechnology Plan every 5 years since 2005. The most recent iteration helped codify the ongoing basic and translational efforts of the field and displayed its breadth with several evolving areas. From merely a technological perspective, this field has seen tremendous growth and success. However, an incomplete understanding of human cancer biology persists relative to the application of nanoscale materials within contemporary oncology. As such, this review presents several evolving areas in cancer nanotechnology in order to identify key clinical and biological challenges that need to be addressed to improve patient outcomes. From this clinical perspective, a sampling of the nano-enabled solutions attempting to overcome barriers faced by traditional therapeutics and diagnostics in the clinical setting are discussed. Finally, a strategic outlook of the future is discussed to highlight the need for next-generation cancer nanotechnology tools designed to address critical gaps in clinical cancer care.
Collapse
Affiliation(s)
- Christopher M Hartshorn
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| | - Michelle S Bradbury
- Department of Radiology and Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York, 10065, United States
| | - Gregory M Lanza
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63108, United States
| | - Andre E Nel
- Division of NanoMedicine, Department of Medicine, and California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jianghong Rao
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford School of Medicine, Stanford, California 94305, United States
| | - Andrew Z. Wang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ulrich B Wiesner
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14843, United States
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Piotr Grodzinski
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Corresponding Author,
| |
Collapse
|
13
|
Song G, Cheng L, Chao Y, Yang K, Liu Z. Emerging Nanotechnology and Advanced Materials for Cancer Radiation Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1700996. [PMID: 28643452 DOI: 10.1002/adma.201700996] [Citation(s) in RCA: 470] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 04/11/2017] [Indexed: 05/22/2023]
Abstract
Radiation therapy (RT) including external beam radiotherapy (EBRT) and internal radioisotope therapy (RIT) has been widely used for clinical cancer treatment. However, owing to the low radiation absorption of tumors, high doses of ionizing radiations are often needed during RT, leading to severe damages to normal tissues adjacent to tumors. Meanwhile, the RT efficacies are limited by different mechanisms, among which the tumor hypoxia-associated radiation resistance is a well-known one, as there exists hypoxia inside most solid tumors while oxygen is essential to enhance radiation-induced DNA damages. With the development in nanotechnology, there have been great interests in using nanomedicine strategies to enhance radiation responses of tumors. Nanomaterials containing high-Z elements to absorb radiation rays (e.g. X-ray) can act as radio-sensitizers to deposit radiation energy within tumors and promote treatment efficacy. Nanoscale carriers are able to deliver therapeutic radioisotopes into tumors for internal RIT, or chemotherapeutic drugs for synergistically combined chemo-radiotherapy. As uncovered in recent studies, the tumor microenvironment could be modulated by various nanomedicine approaches to overcome hypoxia-associated radiation resistance. Herein, the authors will summarize the applications of nanomedicine for RT cancer treatment, and pay particular attention to the latest development of 'advanced materials' for enhanced cancer RT.
Collapse
Affiliation(s)
- Guosheng Song
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, California, 94305-5484, USA
| | - Liang Cheng
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
14
|
Chen Y, Song G, Dong Z, Yi X, Chao Y, Liang C, Yang K, Cheng L, Liu Z. Drug-Loaded Mesoporous Tantalum Oxide Nanoparticles for Enhanced Synergetic Chemoradiotherapy with Reduced Systemic Toxicity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1602869. [PMID: 27957802 DOI: 10.1002/smll.201602869] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/29/2016] [Indexed: 06/06/2023]
Abstract
Combining chemotherapy and radiotherapy (chemoradiotherapy) has been widely applied in many clinical practices, showing promises in enhancing therapeutic outcomes. Nontoxic nanocarriers that not only are able to deliver chemotherapeutics into tumors, but could also act as radiosensitizers to enhance radiotherapy would thus be of great interest in the development of chemoradiotherapies. To achieve this aim, herein mesoporous tantalum oxide (mTa2 O5 ) nanoparticles with polyethylene glycol (PEG) modification are fabricated. Those mTa2 O5 -PEG nanoparticles could serve as a drug delivery vehicle to allow efficient loading of chemotherapeutics such as doxorubicin (DOX), whose release appears to be pH responsive. Meanwhile, owing to the interaction of Ta with X-ray, mTa2 O5 -PEG nanoparticles could offer an intrinsic radiosensitization effect to increase X-ray-induced DNA damages during radiotherapy. As a result, DOX-loaded mTa2 O5 -PEG (mTa2 O5 -PEG/DOX) nanoparticles can offer a strong synergistic therapeutic effect during the combined chemoradiotherapy. Furthermore, in chemoradiotherapy, such mTa2 O5 -PEG/DOX shows remarkably reduced side effects compared to free DOX, which at the same dose appears to be lethal to animals. This work thus presents a new type of mesoporous nanocarrier particularly useful for the delivery of safe and effective chemoradiotherapy.
Collapse
Affiliation(s)
- Yuyan Chen
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Guosheng Song
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ziliang Dong
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xuan Yi
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Chao Liang
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- School of Radiation Medicine and Protection and School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Liang Cheng
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, the Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, Jiangsu, 215123, China
| |
Collapse
|
15
|
Stapleton S, Jaffray D, Milosevic M. Radiation effects on the tumor microenvironment: Implications for nanomedicine delivery. Adv Drug Deliv Rev 2017; 109:119-130. [PMID: 27262923 DOI: 10.1016/j.addr.2016.05.021] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/22/2016] [Accepted: 05/24/2016] [Indexed: 01/24/2023]
Abstract
The tumor microenvironment has an important influence on cancer biological and clinical behavior and radiation treatment (RT) response. However, RT also influences the tumor microenvironment in a complex and dynamic manner that can either reinforce or inhibit this response and the likelihood of long-term disease control in patients. It is increasingly evident that the interplay between RT and the tumor microenvironment can be exploited to enhance the accumulation and intra-tumoral distribution of nanoparticles, mediated by changes to the vasculature and stroma with secondary effects on hypoxia, interstitial fluid pressure (IFP), solid tissue pressure (STP), and the recruitment and activation of bone marrow-derived myeloid cells (BMDCs). The use of RT to modulate nanoparticle drug delivery offers an exciting opportunity to improve antitumor efficacy. This review explores the interplay between RT and the tumor microenvironment, and the integrated effects on nanoparticle drug delivery and efficacy.
Collapse
Affiliation(s)
- Shawn Stapleton
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada
| | - David Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Michael Milosevic
- Radiation Medicine Program, Princess Margaret Cancer Centre and University Health Network, Toronto, ON, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
16
|
Neoantigen activation, protein translocation and targeted drug delivery in combination with radiotherapy. Ther Deliv 2016; 7:377-85. [PMID: 27250535 DOI: 10.4155/tde-2016-0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Concurrent chemo and radiation therapies are commonly used to treat locally advanced cancer. Despite improved efficacy, failure rates remain high due to healthy organ toxicity caused by chemo-radiotherapy. Recent technological advances such as nanoparticle encapsulation of anticancer agents, locally controlled irradiation and concurrent use of radio- and nano-medicines are providing innovative solutions for overcoming the limitations of systemic and local treatment toxicities. In this mini-review, we discuss the roles of radiotherapy in generating new therapeutic targets and altering the tumor microenvironment, and we propose their future applications in drug delivery in combination with radiotherapy.
Collapse
|
17
|
Buckel L, Savariar EN, Crisp JL, Jones KA, Hicks AM, Scanderbeg DJ, Nguyen QT, Sicklick JK, Lowy AM, Tsien RY, Advani SJ. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery. Cancer Res 2015; 75:1376-1387. [PMID: 25681274 DOI: 10.1158/0008-5472.can-14-1931] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022]
Abstract
Intrinsic tumor resistance to radiotherapy limits the efficacy of ionizing radiation (IR). Sensitizing cancer cells specifically to IR would improve tumor control and decrease normal tissue toxicity. The development of tumor-targeting technologies allows for developing potent radiosensitizing drugs. We hypothesized that the anti-tubulin agent monomethyl auristatin E (MMAE), a component of a clinically approved antibody-directed conjugate, could function as a potent radiosensitizer and be selectively delivered to tumors using an activatable cell-penetrating peptide targeting matrix metalloproteinases and RGD-binding integrins (ACPP-cRGD-MMAE). We evaluated the ability of MMAE to radiosensitize both established cancer cells and a low-passage cultured human pancreatic tumor cell line using clonogenic and DNA damage assays. MMAE sensitized colorectal and pancreatic cancer cells to IR in a schedule- and dose-dependent manner, correlating with mitotic arrest. Radiosensitization was evidenced by decreased clonogenic survival and increased DNA double-strand breaks in irradiated cells treated with MMAE. MMAE in combination with IR resulted in increased DNA damage signaling and activation of CHK1. To test a therapeutic strategy of MMAE and IR, PANC-1 or HCT-116 murine tumor xenografts were treated with nontargeted free MMAE or tumor-targeted MMAE (ACPP-cRGD-MMAE). While free MMAE in combination with IR resulted in tumor growth delay, tumor-targeted ACPP-cRGD-MMAE with IR produced a more robust and significantly prolonged tumor regression in xenograft models. Our studies identify MMAE as a potent radiosensitizer. Importantly, MMAE radiosensitization can be localized to tumors by targeted activatable cell-penetrating peptides.
Collapse
Affiliation(s)
- Lisa Buckel
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | - Angel M Hicks
- Department of Radiation Medicine and Applied Sciences
| | | | | | | | | | - Roger Y Tsien
- Department of Pharmacology.,Howard Hughes Medical Institute
| | - Sunil J Advani
- Department of Radiation Medicine and Applied Sciences.,Center for Advanced Radiotherapy Technologies University of California San Diego
| |
Collapse
|
18
|
Abstract
Nanotechnology, the manipulation of matter on atomic and molecular scales, is a relatively new branch of science. It has already made a significant impact on clinical medicine, especially in oncology. Nanomaterial has several characteristics that are ideal for oncology applications, including preferential accumulation in tumors, low distribution in normal tissues, biodistribution, pharmacokinetics, and clearance, that differ from those of small molecules. Because these properties are also well suited for applications in radiation oncology, nanomaterials have been used in many different areas of radiation oncology for imaging and treatment planning, as well as for radiosensitization to improve the therapeutic ratio. In this article, we review the unique properties of nanomaterials that are favorable for oncology applications and examine the various applications of nanotechnology in radiation oncology. We also discuss the future directions of nanotechnology within the context of radiation oncology.
Collapse
Affiliation(s)
- Andrew Z Wang
- All authors: Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Joel E Tepper
- All authors: Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
19
|
Shahin M, Safaei-Nikouei N, Lavasanifar A. Polymeric micelles for pH-responsive delivery of cisplatin. J Drug Target 2014; 22:629-37. [PMID: 24878378 DOI: 10.3109/1061186x.2014.921925] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Methoxy poly(ethylene oxide)-block-poly-(α-carboxylate-ε-caprolactone) (PEO-b-PCCL) was used to develop pH-responsive polymeric micelles for the delivery of cisplatin (CDDP). Micelles were prepared through complexation of CDDP with the pendant carboxyl groups on the poly(ε-caprolactone) core, perhaps through coordinate bonding. The obtained micelles were characterized using dynamic light scattering (DLS) measurement for size and stability. The in vitro release of CDDP at different pHs (7.4, 6.0 and 5.0) was evaluated. The in vitro cell uptake as well as cytotoxicity of developed micelles against two breast cancer cell lines, i.e. MDA-MB-435 and MDA-MB-231, were also assessed and compared to free CDDP as control. DLS results showed PEO-b-PCCL to form stable micelles with an average diameter of <50 nm upon complexation with CDDP. Developed polymeric micelles were capable of slowly releasing CDDP in physiological pH. However, CDDP release from polymeric micelles was triggered upon exposure to electrolytes and/or acidic pHs mimicking that of extracellular tumor microenvironment or intracellular organelles. Consistent with the slow release of CDDP from its polymeric micellar formulation, polymeric micellar CDDP exhibited lower cytotoxicity and CDDP intracellular uptake compared to free drug. The results indicate a great potential for the developed formulation in platinum therapy of breast cancer.
Collapse
|
20
|
Nanoparticles containing insoluble drug for cancer therapy. Biotechnol Adv 2013; 32:778-88. [PMID: 24113214 DOI: 10.1016/j.biotechadv.2013.10.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/26/2013] [Accepted: 10/01/2013] [Indexed: 01/04/2023]
Abstract
Nanoparticle drug formulations have been extensively researched and developed in the field of drug delivery as a means to efficiently deliver insoluble drugs to tumor cells. By mechanisms of the enhanced permeability and retention effect, nanoparticle drug formulations are capable of greatly enhancing the safety, pharmacokinetic profiles and bioavailability of the administered treatment. Here, the progress of various nanoparticle formulations in both research and clinical applications is detailed with a focus on the development of drug/gene delivery systems. Specifically, the unique advantages and disadvantages of polymeric nanoparticles, liposomes, solid lipid nanoparticles, nanocrystals and lipid-coated nanoparticles for targeted drug delivery will be investigated in detail.
Collapse
|