1
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
2
|
Neves CL, Barbosa CMV, Ranéia-Silva PA, Faquim-Mauro EL, Sampaio SC. Crotoxin Modulates Macrophage Phenotypic Reprogramming. Toxins (Basel) 2023; 15:616. [PMID: 37888647 PMCID: PMC10611389 DOI: 10.3390/toxins15100616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Macrophage plasticity is a fundamental feature of the immune response since it favors the rapid and adequate change of the functional phenotype in response to the pathogen or the microenvironment. Several studies have shown that Crotoxin (CTX), the major toxin of the Crotalus durissus terrificus snake venom, has a long-lasting antitumor effect both in experimental models and in clinical trials. In this study, we show the CTX effect on the phenotypic reprogramming of macrophages in the mesenchymal tumor microenvironment or those obtained from the peritoneal cavity of healthy animals. CTX (0.9 or 5 μg/animal subcutaneously) administered concomitantly with intraperitoneal inoculation of tumor cells (1 × 107/0.5 mL, injected intraperitoneally) of Ehrlich Ascitic Tumor (EAT) modulated the macrophages phenotype (M1), accompanied by increased NO• production by cells from ascites, and was evaluated after 13 days. On the other hand, in healthy animals, the phenotypic profile of macrophages was modulated in a dose-dependent way at 0.9 μg/animal: M1 and at 5.0 μg/animal: M2; this was accompanied by increased NO• production by peritoneal macrophages only for the dose of 0.9 μg/animal of CTX. This study shows that a single administration of CTX interferes with the phenotypic reprogramming of macrophages, as well as with the secretory state of cells from ascites, influencing events involved with mesenchymal tumor progression. These findings may favor the selection of new therapeutic targets to correct compromised immunity in different systems.
Collapse
Affiliation(s)
- Camila Lima Neves
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, Brazil;
| | | | | | - Eliana L. Faquim-Mauro
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05503-900, Brazil;
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-220, Brazil
| | - Sandra Coccuzzo Sampaio
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, Brazil;
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-220, Brazil
| |
Collapse
|
3
|
Fumoto S, Yamamoto T, Okami K, Maemura Y, Terada C, Yamayoshi A, Nishida K. Understanding In Vivo Fate of Nucleic Acid and Gene Medicines for the Rational Design of Drugs. Pharmaceutics 2021; 13:159. [PMID: 33530309 PMCID: PMC7911509 DOI: 10.3390/pharmaceutics13020159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid and genetic medicines are increasingly being developed, owing to their potential to treat a variety of intractable diseases. A comprehensive understanding of the in vivo fate of these agents is vital for the rational design, discovery, and fast and straightforward development of the drugs. In case of intravascular administration of nucleic acids and genetic medicines, interaction with blood components, especially plasma proteins, is unavoidable. However, on the flip side, such interaction can be utilized wisely to manipulate the pharmacokinetics of the agents. In other words, plasma protein binding can help in suppressing the elimination of nucleic acids from the blood stream and deliver naked oligonucleotides and gene carriers into target cells. To control the distribution of these agents in the body, the ligand conjugation method is widely applied. It is also important to understand intracellular localization. In this context, endocytosis pathway, endosomal escape, and nuclear transport should be considered and discussed. Encapsulated nucleic acids and genes must be dissociated from the carriers to exert their activity. In this review, we summarize the in vivo fate of nucleic acid and gene medicines and provide guidelines for the rational design of drugs.
Collapse
Affiliation(s)
- Shintaro Fumoto
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8501, Japan; (T.Y.); (K.O.); (Y.M.); (C.T.); (A.Y.); (K.N.)
| | | | | | | | | | | | | |
Collapse
|
4
|
Alavi S, Haeri A, Mahlooji I, Dadashzadeh S. Tuning the Physicochemical Characteristics of Particle-Based Carriers for Intraperitoneal Local Chemotherapy. Pharm Res 2020; 37:119. [DOI: 10.1007/s11095-020-02818-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
|
5
|
Pro-inflammatory macrophage polarization enhances the anti-cancer efficacy of self-assembled galactomannan nanoparticles entrapped with hydrazinocurcumin. Drug Deliv Transl Res 2019; 9:1159-1188. [DOI: 10.1007/s13346-019-00661-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
6
|
Wang SW, Bai YF, Weng YY, Fan XY, Huang H, Zheng F, Xu Y, Zhang F. Cinobufacini Ameliorates Dextran Sulfate Sodium-Induced Colitis in Mice through Inhibiting M1 Macrophage Polarization. J Pharmacol Exp Ther 2019; 368:391-400. [PMID: 30606760 DOI: 10.1124/jpet.118.254516] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/31/2018] [Indexed: 03/08/2025] Open
Abstract
Cinobufacini is a traditional Chinese medicine used clinically that has antitumor and anti-inflammatory effects. It improves colitis outcomes in the clinical setting, but the mechanism underlying its function yet to be uncovered. We investigated the protective effects and mechanisms of cinobufacini on colitis using a dextran sulfate sodium (DSS)-induced colitis mouse model, mainly focusing on the impact of macrophage polarization. Our results showed that cinobufacini dramatically ameliorated DSS-induced colitis in mice. Cinobufacini treatment reduced the infiltration of activated F4/80+ and/or CD68+ macrophages into the colon in DSS-induced colitis mice. More importantly, cinobufacini significantly decreased the quantity of M1 macrophages and the expression of proinflammatory cytokines such as interleukin-6, tumor necrosis factor α, and inducible nitric oxide synthase. Cinobufacini also increased the population of M2 macrophages and the expression of anti-inflammatory factors such as interleukin-10 and arginase-1 in DSS-induced colitis mice. Furthermore, our study demonstrated that cinobufacini inhibited M1 macrophage polarization in lipopolysaccharide-induced RAW 264.7 cells. Mechanistically, our in vivo and in vitro results showed that cinobufacini inhibition of M1 macrophage polarization may be associated with the suppression of nuclear factor κB activation. Our study suggests that cinobufacini could ameliorate DSS-induced colitis in mice by inhibiting M1 macrophage polarization.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Yong-Feng Bai
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Yuan-Yuan Weng
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Xue-Yu Fan
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Hui Huang
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Fang Zheng
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Yi Xu
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| | - Feng Zhang
- Department of Core Facility (S.W., Fa.Z., Fe.Z.), Clinical Laboratory (Y.-B., Y.-W., X.-F., H.H., Fe. Z.), and Department of Urology (Y.X.), People's Hospital of Quzhou, Quzhou, People's Republic of China
| |
Collapse
|
7
|
Dosekova E, Filip J, Bertok T, Both P, Kasak P, Tkac J. Nanotechnology in Glycomics: Applications in Diagnostics, Therapy, Imaging, and Separation Processes. Med Res Rev 2017; 37:514-626. [PMID: 27859448 PMCID: PMC5659385 DOI: 10.1002/med.21420] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
This review comprehensively covers the most recent achievements (from 2013) in the successful integration of nanomaterials in the field of glycomics. The first part of the paper addresses the beneficial properties of nanomaterials for the construction of biosensors, bioanalytical devices, and protocols for the detection of various analytes, including viruses and whole cells, together with their key characteristics. The second part of the review focuses on the application of nanomaterials integrated with glycans for various biomedical applications, that is, vaccines against viral and bacterial infections and cancer cells, as therapeutic agents, for in vivo imaging and nuclear magnetic resonance imaging, and for selective drug delivery. The final part of the review describes various ways in which glycan enrichment can be effectively done using nanomaterials, molecularly imprinted polymers with polymer thickness controlled at the nanoscale, with a subsequent analysis of glycans by mass spectrometry. A short section describing an active glycoprofiling by microengines (microrockets) is covered as well.
Collapse
Affiliation(s)
- Erika Dosekova
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Jaroslav Filip
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Tomas Bertok
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Peter Both
- School of Chemistry, Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DNUK
| | - Peter Kasak
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Jan Tkac
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| |
Collapse
|
8
|
Kalish S, Lyamina S, Manukhina E, Malyshev Y, Raetskaya A, Malyshev I. M3 Macrophages Stop Division of Tumor Cells In Vitro and Extend Survival of Mice with Ehrlich Ascites Carcinoma. Med Sci Monit Basic Res 2017; 23:8-19. [PMID: 28123171 PMCID: PMC5291087 DOI: 10.12659/msmbr.902285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background M1 macrophages target tumor cells. However, many tumors produce anti-inflammatory cytokines, which reprogram the anti-tumor M1 macrophages into the pro-tumor M2 macrophages. We have hypothesized that the problem of pro-tumor macrophage reprogramming could be solved by using a special M3 switch phenotype. The M3 macrophages, in contrast to the M1 macrophages, should respond to anti-inflammatory cytokines by increasing production of pro-inflammatory cytokines to retain its anti-tumor properties. Objectives of the study were to form an M3 switch phenotype in vitro and to evaluate the effect of M3 macrophages on growth of Ehrlich ascites carcinoma (EAC) in vitro and in vivo. Material/Methods Tumor growth was initiated by an intraperitoneal injection of EAC cells into C57BL/6J mice. Results 1) The M3 switch phenotype can be programed by activation of M1-reprogramming pathways with simultaneous inhibition of the M2 phenotype transcription factors, STAT3, STAT6, and/or SMAD3. 2) M3 macrophages exerted an anti-tumor effect both in vitro and in vivo, which was superior to anti-tumor effects of cisplatin or M1 macrophages. 3) The anti-tumor effect of M3 macrophages was due to their anti-proliferative effect. Conclusions Development of new biotechnologies for restriction of tumor growth using in vitro reprogrammed M3 macrophages is very promising.
Collapse
Affiliation(s)
- Sergey Kalish
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Svetlana Lyamina
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Eugenia Manukhina
- Department of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,University of North Texas Health Science Center, Fort Worth, TX, USA.,South Ural State University Biomedical School, Chelyabinsk, Russian Federation
| | | | - Anastasiya Raetskaya
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation
| | - Igor Malyshev
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry n.a. A.I. Evdokimov, Moscow, Russian Federation.,Department of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation.,University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
9
|
Kalish SV, Lyamina SV, Usanova EA, Manukhina EB, Larionov NP, Malyshev IY. Macrophages Reprogrammed In Vitro Towards the M1 Phenotype and Activated with LPS Extend Lifespan of Mice with Ehrlich Ascites Carcinoma. Med Sci Monit Basic Res 2015; 21:226-34. [PMID: 26471744 PMCID: PMC4612464 DOI: 10.12659/msmbr.895563] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background The majority of tumors trigger macrophage reprogramming from an anti-tumor M1 phenotype towards a pro-tumor M2 phenotype. The M2 phenotype promotes tumor growth. We hypothesized that increasing the number of M1 macrophages in a tumor would limit carcinogenesis and extend the lifespan of the tumor host. The aim of this study was to verify this hypothesis in Ehrlich ascites carcinoma (EAC). The objectives were to evaluate effects of 1) EAC on a macrophage phenotype and NO-producing macrophage activity in vivo; 2) ascitic fluid from mice with EAC on a macrophage phenotype and NO-producing macrophage activity in vitro; and 3) in vitro reprogrammed M1 macrophages on lifespan of mice with EAC. Material/Methods The study was conducted using C57BL/6J mice. Results Concentration of nitrite, a stable NO metabolite and an index of NO production, was measured spectrophotometrically. Shifts of macrophage phenotype were assessed by changes in NO production as well as by amounts of CD80, a marker of M1 phenotype, and CD206, a marker of M2 phenotype. The CD markers were measured by flow cytometry. Macrophages were reprogrammed towards the M1 phenotype using two reprogramming factors: 0% FBS and 20 ng/ml IFN-γ. The study results showed that 1) EAC inhibited the macrophage NO production in vivo and reprogrammed macrophages towards the M2 phenotype; 2) ascitic fluid of mice with EAC inhibited the macrophage NO production in vitro and reprogrammed macrophages towards the M2 phenotype; and 3) injection of in vitro reprogrammed M1 macrophages into mice with EAC significantly increased the lifespan of mice. Conclusions These findings suggest that promising biotechnologies for restriction of tumor growth could be developed based on the in vitro macrophage reprogramming.
Collapse
Affiliation(s)
- Sergey V Kalish
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry, Moscow, Russian Federation
| | - Svetlana V Lyamina
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry, Moscow, Russian Federation
| | - Elena A Usanova
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry, Moscow, Russian Federation
| | - Eugenia B Manukhina
- Department of Stress and Adaptation, Institute of General Pathology and Pathophysiology, Moscow, Russian Federation
| | - Nikolai P Larionov
- Department of Biology, Vladimir State University, Vladimir, Russian Federation
| | - Igor Y Malyshev
- Department of Pathophysiology, Moscow State University of Medicine and Dentistry, Moscow, Russian Federation
| |
Collapse
|
10
|
Xiao X, Gang Y, Wang H, Wang J, Zhao L, Xu L, Liu Z. Double-stranded RNA transcribed from vector-based oligodeoxynucleotide acts as transcription factor decoy. Biochem Biophys Res Commun 2014; 457:221-6. [PMID: 25550185 DOI: 10.1016/j.bbrc.2014.12.091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/20/2014] [Indexed: 12/24/2022]
Abstract
In this study, we designed a short hairpin RNA vector-based oligodeoxynucleotide (VB-ODN) carrying transcription factor (TF) consensus sequence which could function as a decoy to block TF activity. Specifically, VB-ODN for Nuclear factor-κB (NF-κB) could inhibit cell viability and decrease downstream gene expression in HEK293 cells without affecting expression of NF-κB itself. The specific binding between VB-ODN produced double-stranded RNA and NF-κB was evidenced by electrophoretic mobility shift assay. Moreover, similar VB-ODNs designed for three other TFs also inhibit their downstream gene expression but not that of themselves. Our study provides a new design of decoy for blocking TF activity.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China
| | - Yi Gang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China; Department of Infectious Diseases, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China
| | - Honghong Wang
- No. 518 Hospital of Chinese People's Liberation Army, Xi'an 710043, Shaanxi Province, PR China
| | - Jiayin Wang
- The Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China
| | - Li Xu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China.
| | - Zhiguo Liu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, PR China.
| |
Collapse
|