Recent Progress in Materials  (ISSN 2689-5846) is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc. This periodical is devoted to publishing high-quality papers that describe the most significant and cutting-edge research in all areas of Materials. Its aim is to provide timely, authoritative introductions to current thinking, developments and research in carefully selected topics. Also, it aims to enhance the international exchange of scientific activities in materials science and technology.
Recent Progress in Materials publishes original high quality experimental and theoretical papers and reviews on basic and applied research in the field of materials science and engineering, with focus on synthesis, processing, constitution, and properties of all classes of materials. Particular emphasis is placed on microstructural design, phase relations, computational thermodynamics, and kinetics at the nano to macro scale. Contributions may also focus on progress in advanced characterization techniques.          

Main research areas include (but are not limited to):
Characterization & evaluation of materials
Metallic materials 
Inorganic nonmetallic materials 
Composite materials
Polymer materials
Biomaterials
Sustainable materials and technologies
Special types of materials
Macro-, micro- and nano structure of materials
Environmental interactions, process modeling
Novel applications of materials

Publication Speed (median values for papers published in 2023): Submission to First Decision: 5.3 weeks; Submission to Acceptance: 12.6 weeks; Acceptance to Publication: 7.5 days (1-2 days of FREE language polishing included)

Current Issue: 2024  Archive: 2023 2022 2021 2020 2019
Open Access Review

Viral/Non-Viral Vectors in DNA/RNA Delivery Technology

Hossein Hosseinkhani *

  1. Innovation Center for Advanced Technology, Matrix HT, Inc., New York, NY 10019, USA

Correspondence: Hossein Hosseinkhani

Academic Editor: Pedro Fonte

Received: August 26, 2024 | Accepted: December 10, 2024 | Published: December 12, 2024

Recent Progress in Materials 2024, Volume 6, Issue 4, doi:10.21926/rpm.2404027

Recommended citation: Hosseinkhani H. Viral/Non-Viral Vectors in DNA/RNA Delivery Technology. Recent Progress in Materials 2024; 6(4): 027; doi:10.21926/rpm.2404027.

© 2024 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Viral and non-viral vectors for nucleic acids (DNA/RNA) therapeutics are important in development of pharmaceutical industries. Although viral vectors have shown significant impact in transfection of DNA/RNA, but numerus studies have shown that non-viral vectors are also effective for the safe and targeted delivery of DNA/RNA to exact location in the body with high percentage of efficacy, safety, and low-immunogenicity. It is also important to protect the nucleic acids from degradation and large-scale production capability at low cost. This review article discusses the potential applications of viral vectors in DNA/RNA delivery technology, while some of the applications of non-viral vectors are compared with viral vectors.

Keywords

Viral; non-viral; DNA/RNA therapeutics; nanoparticles; biodegradable polymers

1. Introduction

DNA and RNA-based therapeutics is an emerging technology for the delivery of genetic materials to the cells for stable transfection. There are mainly two approaches for the delivery of DNA/RNA into the cells and are divided into viral and non-viral vectors. Although among them, viral vectors are very powerful and common for the delivery of genetic materials such as DAN and RNA enable individualized approaches to the treatment of diseases including cancer, diabetes, AIDS, and tuberculosis. Development of viral vectors in delivery of DNA/RNA is due to their high performance in transfecting many in vitro cell lines as well as in vivo applications. Therefore, there are many attempts in using different viral vectors as carrier of genetic materials. It is also well known that different viruses in nature transfect their host body in a similar way with high level of efficacy. This makes viral vectors as the potential available candidates for DNA and RNA delivery.

This review article focuses and emphasizes on the potential applications of viral vectors in DNA/RNA delivery systems in comparison with non-viral vectors.

2. Viral Vectors Technology

2.1 Vaccinia Virus

Vaccinia virus is a member of the Poxviridae family that contains a linear double-stranded DNA genome of about 190 kbp in length with a high number of genes (~250) [1]. It has been vastly used as a smallpox vaccine and an oncolytic agent with comparable safety [2,3,4]. In addition, poxvirus vectors have also been employed in the fabrication of vaccination, cancer immunotherapy, and oncolytic cancer therapy due to the intensive expression, but short-lasting of the transgene and cytolytic properties [5,6]. The complex life cycle of vaccinia involves two mechanisms of infection via separate forms of infectious particles, namely intracellular mature virions and extracellular enveloped virions. The former are the main product of viral lysis, while the latter are actively shed by infected cells [7]. In comparison to other agents, vaccinia allows high levels of viral gene expression, the capacity to spread cell-to-cell, rapid replication, and lysis of infected cells. Its activity is not prevented by therapeutic irradiation [8] and hypoxia [9]. Various strains of vaccinia virus are currently being investigated in preclinical and clinical trials, including Lister, Western Reserve, the Wyeth, and Copenhagen strains [10,11,12,13]. Pexa-Vec (pexastimogene devacirepvec; JX-594) is derived from a Wyeth strain and is regarded as the most advanced vaccinia virus oncolytic product. It is an armed and targeted oncolytic and immunotherapeutic vaccinia virus, causing the disruption of the viral thymidine kinase gene and expression of the hGM-CSF and β-galactosidase transgenes via adjusting the synthetic early-late and p7.5 promoters, respectively [14,15]. Vaccinia virus has now entered a randomized controlled Phase III to express a granulocyte/macrophage-colony-stimulating factor (GM-CSF) [16]. GL-ONC1 is another class of oncolytic and immunotherapeutic vaccinia virus that has been developed for preclinical and clinical studies [17,18]. Recently, GL-ONC1, which is administrated intravenously with chemo radiotherapy, met efficacy requirements in a Phase I clinical trial for treatment of advanced non-metastatic head and neck cancer [13]. Although oncolytic viruses are a promising gene therapy strategy for cancer therapy, several limitations are associated with their applications. The ability to infect disseminated cancer cells over distance, as well as the limited efficiency of delivering and propagating oncolytic viruses throughout the entire tumor, are challenging. This drawback might be overcome by the extracellular enveloped virus through escaping clearance and allowing infection of distant tumor sites [19]. The need to increase systemic antitumor immunity to affect disseminated disease that generally evolves over time is another issue [20]. The potential effect of microbiota in host responsiveness to oncolytic viruses or other vaccinia viruses remains unknown. Microbiota is typically assumed to be significant in the initiation, progression, and dissemination of several cancers, as well as in patient responsiveness to interventional immunotherapies, such as immune checkpoint blockade and immunogenic tumor cell death, inducing chemotherapies [19]. Also, the functions of a large number of genes encoded by the viral genome of vaccinia virus are unclear.

2.2 Herpes Simplex Virus

Herpes simplex virus (HSV) is a double-stranded DNA genome capable of delivering up to 50 kbp of transgenic DNA when used as a vector with more than 150 kb in length. HSV is classified into short and long unique segments and flanked by inverted repeated sequences [21]. Although HSV vectors are mainly able to evade inactivation through host immune response, pre-existing immunity to HSV infection is prevalent within the common population in the same way as the adenovirus [22,23]. Similar to the adenovirus, HSV vector genomes are episomal, thus they suffer from the same challenges of transient expression faced by adenovirus [24,25]. Due to their excellent properties, HSVs are desirable vectors for gene therapy particularly for the treatment of nervous system disorders [21,25]. Certain studies reveal the successful use of HSV vectors for the treatment of cancerous cells and tumors of neuronal origin [26,27,28]. Almost 90 genes are encoded by the HSV genome, many of which are non-essential for its replication cycle, enabling the delivery of very large transgenic DNA. HSV demonstrates diverse cell types with high infectivity and an ability to persist and transduce in both dividing and non-dividing cells. The hidden HSV genome remains episomal as a closed circular molecule, while it does not integrate into cellular DNA [29,30,31]. This unique ability, therefore, prevents isks of insertional mutagenesis. Generally, HSV is divided into three main categories for gene therapy applications, namely defective helper-dependent vectors (known as amplicons), replication-competent attenuated vectors, and replication-incompetent recombinant vectors. The amplicon possesses low immunogenicity and cell toxicity, since it carries minimal viral sequences. It is generated by means of a plasmid vector comprising the HSV origin of DNA replication, transgene(s) of interest, and HSV cleavage-packaging sequences. The replication-competent HSV vectors are attenuated versions of HSV that have the genes required for pathogenicity but are unessential for replication [24]. The replication-incompetent vectors can be produced by deleting genes essential for the lytic cycle of HSV that evoke a shorter immune response than wild-type HSV vectors and are less toxic [25]. Various forms of replication-defective HSV vectors have been formed via deleting several combinations of the immediate-early genes. The immediate-early genes encode infected cell protein (ICP)0, ICP4, ICP22, ICP27 and ICP47. These genes (except ICP47) control the expression of late and early genes that encode the essential functions for the virion structural proteins and viral genome replication, respectively. The biggest barrier to the therapeutic use of HSV is pre-existing immunity that effectively inactivates vector particles and limits transduced cells. The presence of latently infected cells is another safety challenge that may be transduced and present a good environment for the HSV vector to recombine with the wild-type genome [32,33]. Most of these limitations can be efficiently addressed by removal of immediate-early genes from the HSV vector that lead to lower cytotoxic activity and host immune response to viral gene products [34]. Such features enable HSV vector genome to expresses the transgene(s) for almost 30 days in vivo and 21 days in vitro [25]. These barriers are considered a benefit for cancer therapy due to their inherent ability to target the central nervous system. Thus, a wide range of studies demonstrates the use of HSV in combination with oncolytic vectors for the treatment of melanoma, gliomas, glioblastomas, ovarian, and other solid tumors [25,35,36,37].

2.3 Lentiviral Vectors

Lentivirus is a single-stranded RNA that packages two copies of positive-strand RNA comprising three genes: env, gag, and pol. These genes have their own functions: env encodes the envelope proteins that coat the virus, gag encodes structural proteins, and pol encodes the reverse transcriptase, integrase, and protease enzymes that are packaged with the RNA strands [38]. The human immunodeficiency virus (HIV) was first engineered into an integrating lentiviral vector with effective delivery to both non-dividing and mitotic cells [39]. Other lentiviruses and HIV transfer six gene encoding accessory proteins, termed nef, rev, tat, vif, vpr, and vpu [38]. The lentiviral regulatory genes (tat and rev) and accessory genes (vif, vpr, vpu, and nef), are involved in first-generation lentiviral vectors. Specifically, tat and rev are essential for viral replication; vif, vpr, vpu, and nef enable survival benefits for lentiviral replication in vivo, while they are not required for the in vitro growth of the virus [40]. The accessory genes were removed in the second-generation [24], which does not prevent the delivery of genetic component to the host cell. Further attempts caused the development of third-generation lentiviral vectors that divided many cis DNA elements and accessory viral proteins, resulting in the development of self-inactivating (SIN) with enhanced safety [41]. Lentiviral vectors can carry almost 9 Kbp heterologous DNA organized in one or more genes. The availability of non-human lentiviruses, and the absence of pre-existing immunity in the human population capable of transducing human cells upon suitable modification, are advantages of lentiviruses [24]. They are also able to slowly proliferate or effectively transduce non-proliferating cells. Therefore, because of these unique properties, lentiviral vector-based gene delivery has been employed in diverse clinical trials such as X-linked adrenoleukodystrophy [42], Wiskott - Aldrich syndrome [43], and metachromatic leukodystrophy [44,45]. The risk of activating oncogenes, inactivating tumor suppressor genes, or changing many other fundamental regulatory genes (a phenomenon known as “insertional mutagenesis”) is highly dependent on the site of integration and may potentially cause cancer [46,47,48]. The risk of mutagenesis might be decreased by using lentiviral vectors, if it is not eliminated [49]. Thus, safety regarding the development of cancer by insertional mutagenesis remains for lentiviral vectors in gene therapy. Large-scale fabrication of lentiviral vector, particularly for clinical applications, is another hurdle. The lack of a packaging cell line (defined as fabrication of lentiviral vectors centers around the use of a cell line) with stable transfection of the core packaging plasmids increases costs of lentiviral vectors and affects the production process consistency [40]. A recent approach successfully solves this challenge by applying Cre recombinase-mediated insertion of a codon modified HIV-1 gag-pol create into a constitutively expressed locus in 293FT cells [50]. In vivo gene therapy-based lentiviral vectors to the eye raises several difficulties including efficiency, immunogenicity, and the need for tissue-restricted promoters. Thus, the need for in-depth studies considering several methods for modifying lentiviral vectors with improved safety will likely be vital for in vivo applications.

2.4 Other Viral Vectors

In addition to the above-mentioned viral vectors-based gene therapy, there is now a wealth of pre-clinical and clinical studies with various vectors containing polio, measles, and vesicular stomatitis virus (VSV).

2.4.1 Poliovirus

Poliovirus is a member of the Picornaviridae family and is regarded as the causative factor in paralytic poliomyelitis through selective destruction of motor neurons [51,52]. The productive poliovirus replication exists in two areas in humans, including the spinal cord motor neurons and the gastrointestinal tract (GIT), as well as its associated lymphatic structures [53,54]. These sites for poliovirus propagation occur simultaneously with CD155 (also known as poliovirus receptor) expression [55]. Poliovirus propagation in the GIT generates unclear enteric pathology and is typically silent, leading to severe safety issues. Consequently, the potential neuropathogenicity of the poliovirus should be completely removed before clinical studies. The favourable polioviruses should be: a) non-pathogenic, b) genetically stable on intratumoral replication, c) target and affect heterogeneous tumors, d) cause effective destruction of cancer cells, e) function in natural antiviral immune activation and neutralizing antibodies, f) reverse the immune suppressive microenvironment, and g) be able to employ immune effectors in tumor [55]. Currently, poliovirus is used in Phase I clinical trials for treatment of glioblastoma as the most aggressive cancer [56].

2.4.2 Measles Virus

Measles is an enveloped negative single-strand RNA virus belonging to the Paramyxoviridae family. A measles virus genome contains six genes that encode eight proteins: fusion (F), hemagglutinin (H), large (L) protein, nucleocapsid (N), phospho- (P), matrix (M), as well as the accessory proteins V and C that are encoded by the P-cistron [57]. The F and H proteins cause viral attachment and entry into host cells. There are three cellular receptors known for the bmeasles virus: the signal lymphocyte-activation molecule (SLAM; also known as CD150), membrane cofactor protein (CD46), and Nectin4 (also known as Polio virus receptor-related 4 (PVRL4) [58]. SLAM is a transmembrane glycoprotein fundamentally expressed on the surface of activated B- and T- lymphocytes, immature thymocytes, memory lymphocytes, and dendritic cells [59]. Most of the wild-type measles virus strains enter cells through SLAM. The CD46 is ubiquitously present on most normal human cells and is only used via laboratory adapted/vaccine strains MV. However, Nectin4 is used by both the laboratory/vaccine and wild-type strain measles virus. It is generally overexpressed on the surface of adenocarcinoma cells [58]. Clinically, measles virus has been developed in Phase I/II trials against malignant mesothelioma (NCT01503177), metastatic head and neck cancer, breast cancer (NCT01846091), fallopian, ovarian, or peritoneal cancer (NCT02068794, NCT02364713, NCT00408590), and multiple myeloma (NCT00450814, NCT02192775) [60].

2.4.3 Vesicular Stomatitis Virus

Vesicular stomatitis virus (VSV) belongs to the Rhabdoviridae family, genus Vesiculovirus, and is a nonsegmented, negative-sense, single-stranded RNA virus. Upon entering the cell the viral genomic RNA is released into the cytoplasm, where the positive-sense RNA (viral mRNA) is formed through the viral RNA-dependent RNA polymerase protein (RdRp) in the viral particle. The viral RNA polymerase produces antigenomic RNA in the absence of poly A tail and cap structure to replicate its viral genome in the cytoplasm of the host cells [61]. VSV does not go through a DNA intermediate during replication, similarly to other cytoplasmic RNA viruses. The viral RNAs also do not enter the nucleus of the host cell during the viral life cycle, and the viral RNAs are produced in the cytoplasm. VSV has been suggested as a desirable candidate for antitumor therapy, since it is a safe and powerful inducer of apoptosis with diverse cancer cells susceptible to killing by VSV [62,63,64]. Most cancers are irresponsive to the role of interferon (IFN), while in the presence of IFN normal healthy tissues are not affected by the virus [65]. A main challenge regarding VSV-based tumor therapy is whether the virus can be tolerated by cancer patients who are immunologically compromised. Various strategies have been introduced to solve this problem, including administering IFN systemically during VSV infection or development of VSV M-protein mutant [65], fabricating a recombinant VSV expressing IFNβ [66], and creating a recombinant replicating VSV with a modified surface glycoprotein [67].

3. Non-Viral Vectors Technology

Engineering viral-like vectors for RNA therapeutics development in order to improve their delivery methods and stability in vivo is one the challenge in pharmaceutical industries [68,69,70,71,72,73,74,75,76,77]. Numerus studies have shown that polymeric nanoparticulate systems are an efficient method for the safe and targeted delivery of DNA/RNA. Of particular note, it has found that artificial virus vehicles present several advantages including safety, low-immunogenicity, a capacity to deliver large RNA, and large-scale production capability at low cost [78,79,80,81,82,83,84,85,86,87,88,89,90,91]. These advantages make novel technology a boon to RNA therapeutics development and a strong tool for his field in creating delivery vehicles for this new class of therapeutic intervention intended for the treatment of viral and bacterial infections. Quantum dots are one of the newest materials that currently in use for delivery of genetic materials [92,93,94]. There are also concern of the use of biodegradable materials for successful delivery of DNA both in vitro and in vivo [95,96,97,98,99,100]. In this regards, polymeric materials have shown great interest in the development of no-viral vectors [100,101,102,103,104,105]. Among many polymeric materials, biodegradable types are more applicable for the delivery of genetic materials, because they are timely degradable and will be adsorbed to the host body after controlled release of the DNA molecules into the cells [106,107,108,109,110]. Although, there are more successful progress in the use of biodegradable polymers, but more researches are still needed to overcome many of their limitations [111,112,113,114,115,116,117].

4. Materials Technology

Materials design and combinational technology of biomaterials development have been significantly impacted on the new area of biodegradable polymeric materials [118,119,120,121]. Biomaterials are in the core of materials development for biomedical applications from the view point of early stage detection of various diseases to treatment of end stage-therapeutic applications [122,123,124,125,126,127,128,129,130]. Materials for medical applications are divided into non-degradable and bio-degradable and each of them has advantages and disadvantages. From the view point of stability, non- non-degradable materials are more stable to use for some of medical implants [130,131,132,133,134,135], while they may cause some issue concerning to toxicity. In this regards, bio-degradable materials are the best choice for in vivo and in vitro application [136,137,138,139,140,141,142]. Delivery of genetic materials to the cytoplasm of cells is one the biggest challenge in the treatment of various e disorder diseases [143,144,145,146,147,148,149,150]. There are many steps in gene therapy technology in order to be successful both in vitro and in vivo [150,151,152,153,154,155]. The first step is to incorporate the negatively charged DNA or RNA into the gene carriers. Therefore, the stability of the formation between the DNA/RNA and the carrier is one of the most important factors in order to protect the genetic materials during the applications [156,157,158,159,160]. One the easiest technology is to use positively charged biomaterials to electrostatically interact with negatively charged DNA/RNA due the presence of the phosphate groups in their chemical structure backbone and to form stable micro- or nanoparticles [161,162,163,164,165,166,167,168,169]. The stability of the formation of micro- or nanoparticles is important to protect the degradation of genetic materials before transfection and after transfection of the host cells [170,171,172,173,174,175]. Combinational technology of three dimensional (3D) biomaterials with gene carriers can also make stable delivery of genetic materials to the cells [176,177,178,179,180,181,182,183]. In this regard, biomaterials are the best choice to form a platform for reverse transfection where cells first are seeded on the biomaterials and next the gene carrier are added on the top of 3D biomaterials and slowly diffuses into the 3D scaffolds and transfect the seeded cells [184,185,186,187,188,189,190,191,192]. 3D scaffolding biomaterials that are mainly used for tissue engineering applications are considered for stable gene transfection in vitro and apply to implant the transfected cells into the defect area of the body such as bone for tissue regeneration. Also, these 3D scaffolding biomaterials have been widely applied in regenerative medicine technology [193,194,195,196,197,198,199,200]. One of the advantages of biomaterials is combinational technology of tissue engineering and gene therapy where the genetic materials are incorporate into the biodegradable biomaterials and the host cells are seeded on the biomaterials [201,202,203,204,205,206,207,208,209]. The delivery mechanisms of DNA/RNA from biodegradable biomaterials are based on combination of the degradation of the carriers and diffusion of the genetic materials into the host cells. Therefore, it is important that the degradation of the biomaterials should later after the controlled release of the DNA/RNA from the carrier [210,211,212]. Also, biocompatibility of the materials is important factors for various cells and tissue both in vitro and in vivo to overcome toxicity of implanted of the materials [213,214].

Arginine based polymers as gene carriers have been widely designed as non-viral vectors gene carrier with high level of transfection efficacy [215,216,217,218]. These biodegradable and biocompatible polymers of arginine-based poly (ester amide) were synthesized with a tunable structure for such a comprehensive biopolymer and have shown great impact in DNA delivery technology.

5. RNA Technology

RNA technology is a prospective new form of medicine in which in one area of RNA-based medicine, mRNAs have been created to serve as efficient vaccines to protect from the SARS-CoV-2 virus. This marks the new generation of nucleotide-based biological drugs and vaccines. Cancer in general, as well as viral infections such as COVID-19, are serious threats to the public health. RNA technology is a major achievement in pharmaceutical development, and various RNA drugs have already been approved by the United States Food and Drug Administration. As such, it is also a boon to the national pharmaceutical industry, which is a significant contributor to the economy. The pharmaceutical industry had revenue of about $425 billion in 2020 alone, and it accounts for almost 50 percent of the pharmaceutical industry worldwide.

The development of viral-like vectors for RNA therapeutic treatments is of great economic value.

Among RNA molecules, siRNA have been widely developed for therapeutic applications [219,220,221]. siRNA molecules are powerful tools in developing products for pharmaceutical, biotechnology, and academic research. Several studies have shown that biodegradable nanoparticles are capable to carry siRNA molecules with high percentage of transfection and can protect the molecules from in vitro or in vivo degradation by surrounding enzymes [219]. Biodegradable natural polysaccharides have been widely used for delivery of RNA/DNA molecules and showed high level of transfection efficacy [86,87,88,89,90,91].

6. Future Prospects

The delivery technologies of genetic materials have become very effective in treating various diseases. Viruses such as retroviruses, adenoviruses, adeno-associated virus and herpes virus used for gene therapy to date are very powerful tools for therapeutics applications. However, they have several concerns for their future applications such as: risks of virals infection, immunogenicity, toxicity, limited size of gene, one copy, expensive, long procedure, stability and regulatory issues, and limited to certain tissues and cells. In this regard, no-viral vectors have become great interests to overcome the above problems of viral vectors as they shows: no infection, low toxicity, no limited on the size of gene, cheap, easy to prepare, stable, and applicable to all tissues and cells. Their main problem is very low transfection efficiency. The future prospects of using non-viral vectors in pharmaceutical industries must focus on designing nanoparticles that acts like a virus and it would be called artificial virus having similar high transfection efficiency.

Author Contributions

The author did all the research work of this study.

Competing Interests

The author has declared that no competing interests exist.

References

  1. Vannucci L, Lai M, Chiuppesi F, Ceccherini-Nelli L, Pistello M. Viral vectors: A look back and ahead on gene transfer technology. New Microbiol. 2013; 36: 1-22.
  2. Poland GA, Grabenstein JD, Neff JM. The US smallpox vaccination program: A review of a large modern era smallpox vaccination implementation program. Vaccine. 2005; 23: 2078-2081. [CrossRef]
  3. Chan WM, McFadden G. Oncolytic poxviruses. Annu Rev Virol. 2014; 1: 119-141. [CrossRef]
  4. Mansfield DC, Kyula JN, Rosenfelder N, Chao-Chu J, Kramer-Marek G, Khan AA, et al. Oncolytic vaccinia virus as a vector for therapeutic sodium iodide symporter gene therapy in prostate cancer. Gene Ther. 2016; 23: 357-368. [CrossRef]
  5. E Gomez C, L Najera J, Krupa M, Perdiguero B, Esteban M. MVA and NYVAC as vaccines against emergent infectious diseases and cancer. Curr Gene Ther. 2011; 11: 189-217. [CrossRef]
  6. Kim JW, Gulley JL. Poxviral vectors for cancer immunotherapy. Expert Opin Biol Ther. 2012; 12: 463-478. [CrossRef]
  7. Smith GL, Vanderplasschen A, Law M. The formation and function of extracellular enveloped vaccinia virus. J Gen Virol. 2002; 83: 2915-2931. [CrossRef]
  8. Mansfield D, Pencavel T, Kyula JN, Zaidi S, Roulstone V, Thway K, et al. Oncolytic vaccinia virus and radiotherapy in head and neck cancer. Oral Oncol. 2013; 49: 108-118. [CrossRef]
  9. Hiley CT, Yuan M, Lemoine NR, Wang Y. Lister strain vaccinia virus, a potential therapeutic vector targeting hypoxic tumours. Gene Ther. 2010; 17: 281-287. [CrossRef]
  10. Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, et al. Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer. Nat Med. 2013; 19: 329-336. [CrossRef]
  11. Zeh HJ, Downs-Canner S, McCart JA, Guo ZS, Rao UN, Ramalingam L, et al. First-in-man study of western reserve strain oncolytic vaccinia virus: Safety, systemic spread, and antitumor activity. Mol Ther. 2015; 23: 202-214. [CrossRef]
  12. Foloppe J, Kintz J, Futin N, Findeli A, Cordier P, Schlesinger Y, et al. Targeted delivery of a suicide gene to human colorectal tumors by a conditionally replicating vaccinia virus. Gene Ther. 2008; 15: 1361-1671. [CrossRef]
  13. Mell LK, Brumund KT, Daniels GA, Advani SJ, Zakeri K, Wright ME, et al. Phase I trial of intravenous oncolytic vaccinia virus (GL-ONC1) with cisplatin and radiotherapy in patients with locoregionally advanced head and neck carcinoma. Clin Cancer Res. 2017; 23: 5696-5702. [CrossRef]
  14. Kim JH, Oh JY, Park BH, Lee DE, Kim JS, Park HE, et al. Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. Mol Ther. 2006; 14: 361-370. [CrossRef]
  15. Kirn DH, Thorne SH. Targeted and armed oncolytic poxviruses: A novel multi-mechanistic therapeutic class for cancer. Nat Rev Cancer. 2009; 9: 64-71. [CrossRef]
  16. Breitbach CJ, Burke J, Jonker D, Stephenson J, Haas AR, Chow LQ, et al. Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humans. Nature. 2011; 477: 99-102. [CrossRef]
  17. Advani SJ, Buckel L, Chen NG, Scanderbeg DJ, Geissinger U, Zhang Q, et al. Preferential replication of systemically delivered oncolytic vaccinia virus in focally irradiated glioma xenografts. Clin Cancer Res. 2012; 18: 2579-2590. [CrossRef]
  18. Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, et al. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: A phase I trial. Lancet Oncol. 2008; 9: 533-542. [CrossRef]
  19. Guo ZS, Lu B, Guo Z, Giehl E, Feist M, Dai E, et al. Vaccinia virus-mediated cancer immunotherapy: Cancer vaccines and oncolytics. J Immunother Cancer. 2019; 7: 6. [CrossRef]
  20. Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, Martins MM, et al. Systemic immunity is required for effective cancer immunotherapy. Cell. 2017; 168: 487-502.e15. [CrossRef]
  21. Manservigi R, Argnani R, Marconi P. HSV recombinant vectors for gene therapy. Open Virol J. 2010; 4: 123-156. [CrossRef]
  22. Waehler R, Russell SJ, Curiel DT. Engineering targeted viral vectors for gene therapy. Nat Rev Genet. 2007; 8: 573-587. [CrossRef]
  23. Lachmann R. Herpes simplex virus‐based vectors. Int J Exp Pathol. 2004; 85: 177-190. [CrossRef]
  24. Robbins PD, Ghivizzani SG. Viral vectors for gene therapy. Pharmacol Ther. 1998; 80: 35-47. [CrossRef]
  25. Lentz TB, Gray SJ, Samulski RJ. Viral vectors for gene delivery to the central nervous system. Neurobiol Dis. 2012; 48: 179-188. [CrossRef]
  26. Goss JR, Harley CF, Mata M, O'Malley ME, Goins WF, Hu X, et al. Herpes vector-mediated expression of proenkephalin reduces bone cancer pain. Ann Neurol. 2002; 52: 662-665. [CrossRef]
  27. Goins WF, Goss JR, Chancellor MB, De Groat WC, Glorioso JC, Yoshimura N. Herpes simplex virus vector-mediated gene delivery for the treatment of lower urinary tract pain. Gene Ther. 2009; 16: 558-569. [CrossRef]
  28. Wolfe D, Mata M, Fink DJ. A human trial of HSV-mediated gene transfer for the treatment of chronic pain. Gene Ther. 2009; 16: 455-460. [CrossRef]
  29. Garber DA, Beverley SM, Coen DM. Demonstration of circularization of herpes simplex virus DNA following infection using pulsed field gel electrophoresis. Virology. 1993; 197: 459-462. [CrossRef]
  30. Efstathiou S, Minson AC, Field HJ, Anderson JR, Wildy P. Detection of herpes simplex virus-specific DNA sequences in latently infected mice and in humans. J Virol. 1986; 57: 446-455. [CrossRef]
  31. Jackson SA, DeLuca NA. Relationship of herpes simplex virus genome configuration to productive and persistent infections. Proc Natl Acad Sci. 2003; 100: 7871-7876. [CrossRef]
  32. De Silva S, Bowers WJ. Herpes virus amplicon vectors. Viruses. 2009; 1: 594-629. [CrossRef]
  33. Marconi P, Argnani R, Epstein AL, Manservigi R. HSV as a vector in vaccine development and gene therapy. In: Pharmaceutical biotechnology. New York, NY: Springer; 2009. pp. 118-144. [CrossRef]
  34. Grant KG, Krisky DM, Ataai MM, Glorioso III JC. Engineering cell lines for production of replication defective HSV‐1 gene therapy vectors. Biotechnol Bioeng. 2009; 102: 1087-1097. [CrossRef]
  35. Pulkkanen KJ, Yla-Herttuala S. Gene therapy for malignant glioma: Current clinical status. Mol Ther. 2005; 12: 585-598. [CrossRef]
  36. Marconi P, Manservigi R, Epstein AL. HSV-1-derived helper-independent defective vectors, replicating vectors and amplicon vectors, for the treatment of brain diseases. Curr Opin Drug Discov Devel. 2010; 13: 169-183.
  37. Wong J, Lee C, Zhang K, S Rennie P, Jia W. Targeted oncolytic herpes simplex viruses for aggressive cancers. Curr Pharm Biotechnol. 2012; 13: 1786-1794. [CrossRef]
  38. Escors D, Breckpot K. Lentiviral vectors in gene therapy: Their current status and future potential. Arch Immunol Ther Exp. 2010; 58: 107-119. [CrossRef]
  39. Naldini L, Blömer U, Gallay P, Ory D, Mulligan R, Gage FH, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science. 1996; 272: 263-267. [CrossRef]
  40. Milone MC, O’Doherty U. Clinical use of lentiviral vectors. Leukemia. 2018; 32: 1529-1541. [CrossRef]
  41. Dull T, Zufferey R, Kelly M, Mandel RJ, Nguyen M, Trono D, et al. A third-generation lentivirus vector with a conditional packaging system. J Virol. 1998; 72: 8463-8471. [CrossRef]
  42. Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, Kutschera I, et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science. 2009; 326: 818-823. [CrossRef]
  43. Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 2013; 341: 1233151. [CrossRef]
  44. Biffi A, Montini E, Lorioli L, Cesani M, Fumagalli F, Plati T, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science. 2013; 341: 1233158. [CrossRef]
  45. Sessa M, Lorioli L, Fumagalli F, Acquati S, Redaelli D, Baldoli C, et al. Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: An ad-hoc analysis of a non-randomized, open-label, phase 1/2 trial. Lancet. 2016; 388: 476-487. [CrossRef]
  46. Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry CC, et al. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N Engl J Med. 2010; 363: 355-364. [CrossRef]
  47. Fox JL. US authorities uphold suspension of SCID gene therapy. Nat Biotechnol. 2003; 21: 217. [CrossRef]
  48. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch PA, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003; 302: 415-419. [CrossRef]
  49. Maldarelli F, Wu X, Su L, Simonetti FR, Shao W, Hill S, et al. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science. 2014; 345: 179-183. [CrossRef]
  50. Sanber KS, Knight SB, Stephen SL, Bailey R, Escors D, Minshull J, et al. Construction of stable packaging cell lines for clinical lentiviral vector production. Sci Rep. 2015; 5: 9021. [CrossRef]
  51. Gromeier M, Lachmann S, Rosenfeld MR, Gutin PH, Wimmer E. Intergeneric poliovirus recombinants for the treatment of malignant glioma. Proc Natl Acad Sci. 2000; 97: 6803-6808. [CrossRef]
  52. Cello J, Toyoda H, DeJesus N, Dobrikova EY, Gromeier M, Wimmer E. Growth phenotypes and biosafety profiles in poliovirus‐receptor transgenic mice of recombinant oncolytic polio/human rhinoviruses. J Med Virol. 2008; 80: 352-359. [CrossRef]
  53. Bodian D. Emerging concept of poliomyelitis infection. Science. 1955; 122: 105-108. [CrossRef]
  54. Sabin AB. Pathogenesis of poliomyelitis reappraisal in the light of new data. Science. 1956; 123: 1151-1157. [CrossRef]
  55. Iwasaki A, Welker R, Mueller S, Linehan M, Nomoto A, Wimmer E. Immunofluorescence analysis of poliovirus receptor expression in Peyer’s patches of humans, primates, and CD155 transgenic mice: Implications for poliovirus infection. J Infect Dis. 2002; 186: 585-592. [CrossRef]
  56. Gromeier M, Nair SK. Recombinant poliovirus for cancer immunotherapy. Annu Rev Med. 2018; 69: 289-299. [CrossRef]
  57. Yanagi Y, Takeda M, Ohno S. Measles virus: Cellular receptors, tropism and pathogenesis. J Gen Virol. 2006; 87: 2767-2779. [CrossRef]
  58. Lin LT, Richardson CD. The host cell receptors for measles virus and their interaction with the viral hemagglutinin (H) protein. Viruses. 2016; 8: 250. [CrossRef]
  59. Navaratnarajah CK, Leonard VH, Cattaneo R. Measles virus glycoprotein complex assembly, receptor attachment, and cell entry. In: Measles: History and Basic Biology. Berlin, Heidelberg: Springer; 2009. pp. 59-76. [CrossRef]
  60. Tai CJ, Liu CH, Pan YC, Wong SH, Tai CJ, Richardson CD, et al. Chemovirotherapeutic treatment using camptothecin enhances oncolytic measles virus-mediated killing of breast cancer cells. Sci Rep. 2019; 9: 6767. [CrossRef]
  61. Dietzgen RG, Kondo H, Goodin MM, Kurath G, Vasilakis N. The family Rhabdoviridae: Mono-and bipartite negative-sense RNA viruses with diverse genome organization and common evolutionary origins. Virus Res. 2017; 227: 158-170. [CrossRef]
  62. Galivo F, Diaz RM, Thanarajasingam U, Jevremovic D, Wongthida P, Thompson J, et al. Interference of CD40L-mediated tumor immunotherapy by oncolytic vesicular stomatitis virus. Hum Gene Ther. 2010; 21: 439-450. [CrossRef]
  63. Moussavi M, Fazli L, Tearle H, Guo Y, Cox M, Bell J, et al. Oncolysis of prostate cancers induced by vesicular stomatitis virus in PTEN knockout mice. Cancer Res. 2010; 70: 1367-1376. [CrossRef]
  64. Wollmann G, Rogulin V, Simon I, Rose JK, van den Pol AN. Some attenuated variants of vesicular stomatitis virus show enhanced oncolytic activity against human glioblastoma cells relative to normal brain cells. J Virol. 2010; 84: 1563-1573. [CrossRef]
  65. Stojdl DF, Lichty B, Knowles S, Marius R, Atkins H, Sonenberg N, et al. Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus. Nat Med. 2000; 6: 821-825. [CrossRef]
  66. Obuchi M, Fernandez M, Barber GN. Development of recombinant vesicular stomatitis viruses that exploit defects in host defense to augment specific oncolytic activity. J Virol. 2003; 77: 8843-8856. [CrossRef]
  67. Gao Y, Whitaker-Dowling P, Griffin JA, Bergman I. Treatment with targeted vesicular stomatitis virus generates therapeutic multifunctional anti-tumor memory CD4 T cells. Cancer Gene Ther. 2012; 19: 282-291. [CrossRef]
  68. Hosseinkhani H, Azzam T, Tabata Y, Domb AJ. Dextran-spermine polycation: An efficient nonviral vector for in vitro and in vivo gene transfection. Gene Ther. 2004; 11: 194-203. [CrossRef]
  69. Abedini F, Hosseinkhani H, Ismail M, Chen YR, Omar A, Chong P, et al. In vitro intracellular trafficking of biodegradable nanoparticles dextran-spermine in cancer cell lines. Int J Nanotechnol. 2011; 8: 712-723. [CrossRef]
  70. Hosseinkhani H, Abedini F, Ou KL, Domb AJ. Polymers in gene therapy technology. Polym Adv Technol. 2015; 26: 198-211. [CrossRef]
  71. Abdullah S, Wendy-Yeo WY, Hosseinkhani H, Hosseinkhani M, Masrawa E, Ramasamy R, et al. Gene transfer into the lung by nanoparticle dextran‐spermine/plasmid DNA complexes. Biomed Res Int. 2010; 2010: 284840. [CrossRef]
  72. Abedini F, Ismail M, Hosseinkhani H, Azmi T, Omar AR, Chong PP, et al. Toxicity evaluation of dextran-spermine polycation as a tool for gene therapy in vitro. J Cell Anim Biol. 2010; 4: 170-176.
  73. Abedini F, Ismail M, Hosseinkhani H, Ibrahim TA, Omar AR, Chong PP, et al. Effects of CXCR4 siRNA/dextran-spermine nanoparticles on CXCR4 expression and serum LDH levels in a mouse model of colorectal cancer metastasis to the liver. Cancer Manage Res. 2011; 3: 301-309. [CrossRef]
  74. Yeo WW, Hosseinkhani H, Rahman SA, Rosli R, Domb AJ, Abdullah S. Safety profile of dextran-spermine gene delivery vector in mouse lungs. J Nanosci Nanotechnol. 2014; 14: 3328-3336. [CrossRef]
  75. Ghadiri M, Vasheghani-Farahani E, Atyabi F, Kobarfard F, Hosseinkhani H. In-vitro assessment of magnetic dextran-spermine nanoparticles for capecitabine delivery to cancerous cells. Iran J Pharm Res. 2017; 16: 1320-1334.
  76. Jain A, Hosseinkhani H, Domb AJ, Khan W. Cationic polymers for the delivery of therapeutic nucleotides. In: Polysaccharides. Cham: Springer; 2015. pp. 1969-1990. [CrossRef]
  77. He W, Hosseinkhani H, Mohammadinejad R, Roveimiab Z, Hueng DY, Ou KL, et al. Polymeric nanoparticles for therapy and imaging. Polym Adv Technol. 2014; 25: 1216-1225. [CrossRef]
  78. Hosseinkhani H, Chen YR, He W, Hong PD, Yu DS, Domb AJ. Engineering of magnetic DNA nanoparticles for tumor-targeted therapy. J Nanopart Res. 2013; 15: 1345. [CrossRef]
  79. Hosseinkhani H, He WJ, Chiang CH, Hong PD, Yu DS, Domb AJ, et al. Biodegradable nanoparticles for gene therapy technology. J Nanopart Res. 2013; 15: 1794. [CrossRef]
  80. Amini R, Jalilian FA, Abdullah S, Veerakumarasivam A, Hosseinkhani H, Abdulamir AS, et al. Dynamics of PEGylated-dextran-spermine nanoparticles for gene delivery to leukemic cells. Appl Biochem Biotechnol. 2013; 170: 841-853. [CrossRef]
  81. He WJ, Hosseinkhani H, Hong PD, Chiang CH, Yu DS. Magnetic nanoparticles for imaging technology. Int J Nanotechnol. 2013; 10: 930-944. [CrossRef]
  82. Khan W, Hosseinkhani H, Ickowicz D, Hong PD, Yu DS, Domb AJ. Polysaccharide gene transfection agents. Acta Biomater. 2012; 8: 4224-4232. [CrossRef]
  83. Abedini F, Hosseinkhani H, Ismail M, Domb AJ, Omar AR, Chong PP, et al. Cationized dextran nanoparticle-encapsulated CXCR4-siRNA enhanced correlation between CXCR4 expression and serum alkaline phosphatase in a mouse model of colorectal cancer. Int J Nanomed. 2012; 7: 4159-4168. [CrossRef]
  84. Mohammad-Taheri M, Vasheghani-Farahani E, Hosseinkhani H, Shojaosadati SA, Soleimani M. Fabrication and characterization of a new MRI contrast agent based on a magnetic dextran-spermine nanoparticle system. Iran Polym J. 2012; 21: 239-251. [CrossRef]
  85. Amini R, Hosseinkhani H, Abdulamir AS, Rosli R, Jalilian FA. Engineered smart biomaterials for gene delivery. Gene Ther Mol Biol. 2012; 14: 72-86.
  86. Hosseinkhani H, Hosseinkhani M, Chen YR, Subramani K, Domb AJ. Innovative technology of engineering magnetic DNA nanoparticles for gene therapy. Int J Nanotechnol. 2011; 8: 724-735. [CrossRef]
  87. Hosseinkhani M, Hosseinkhani H, Chen YR, Subramani K. In vitro physicochemical evaluation of DNA nanoparticles. Int J Nanotechnol. 2011; 8: 736-748. [CrossRef]
  88. Hosseinkhani H. DNA nanoparticles for gene delivery to cells and tissue. Int J Nanotechnol. 2006; 3: 416-461. [CrossRef]
  89. Abedini F, Ebrahimi M, Hosseinkhani H. Technology of RNA interference in advanced medicine. MicroRNA. 2018; 7: 74-84. [CrossRef]
  90. Abedini F, Ebrahimi M, Roozbehani AH, Domb AJ, Hosseinkhani H. Overview on natural hydrophilic polysaccharide polymers in drug delivery. Polym Adv Technol. 2018; 29: 2564-2573. [CrossRef]
  91. Ghadiri M, Vasheghani‐Farahani E, Atyabi F, Kobarfard F, Mohamadyar‐Toupkanlou F, Hosseinkhani H. Transferrin‐conjugated magnetic dextran‐spermine nanoparticles for targeted drug transport across blood‐brain barrier. J Biomed Mater Res A. 2017; 105: 2851-2864. [CrossRef]
  92. Alibolandi M, Abnous K, Sadeghi F, Hosseinkhani H, Ramezani M, Hadizadeh F. Folate receptor-targeted multimodal polymersomes for delivery of quantum dots and doxorubicin to breast adenocarcinoma: In vitro and in vivo evaluation. Int J Pharm. 2016; 500: 162-178. [CrossRef]
  93. Alibolandi M, Abnous K, Ramezani M, Hosseinkhani H, Hadizadeh F. Synthesis of AS1411-aptamer-conjugated CdTe quantum dots with high fluorescence strength for probe labeling tumor cells. J Fluoresc. 2014; 24: 1519-1529. [CrossRef]
  94. Sarabi RS, Sadeghi EL, Hosseinkhani HO, Mahmoudi MO, Kalantari M, Adeli MO. Polyrotaxane capped quantum dots as new candidates for cancer diagnosis and therapy. J Nanostruct Polym Nanocompos. 2011; 7: 18-31.
  95. Khan M. Polymers as Efficient Non-Viral Gene Delivery Vectors: The Role of the Chemical and Physical Architecture of Macromolecules. Polym. 2024; 16: 2629-2671. [CrossRef]
  96. Mottaghitalab F, Farokhi M, Shokrgozar MA, Atyabi F, Hosseinkhani H. Silk fibroin nanoparticle as a novel drug delivery system. J Control Release. 2015; 206: 161-176. [CrossRef]
  97. Chiang CH, Hosseinkhani H, Cheng WS, Chen CW, Wang CH, Lo YL. Improving drug loading efficiency and delivery performance of micro-and nanoparticle preparations through optimising formulation variables. Int J Nanotechnol. 2013; 10: 996-1006. [CrossRef]
  98. Steinman NY, Campos LM, Feng Y, Domb AJ, Hosseinkhani H. Cyclopropenium nanoparticles and gene transfection in cells. Pharmaceutics. 2020; 12: 768. [CrossRef]
  99. Hu CS, Tang SL, Chiang CH, Hosseinkhani H, Hong PD, Yeh MK. Characterization and anti-tumor effects of chondroitin sulfate-chitosan nanoparticles delivery system. J Nanopart Res. 2014; 16: 2672. [CrossRef]
  100. Hosseinkhani H, Hosseinkhani M. Biodegradable polymer-metal complexes for gene and drug delivery. Curr Drug Saf. 2009; 4: 79-83. [CrossRef]
  101. Abdallah B, Hassan A, Benoist C, Goula D, Behr JP, Demeneix BA. A powerful nonviral vector for in vivo gene transfer into the adult mammalian brain: Polyethylenimine. Hum Gene Ther. 1996; 7: 1947-1954. [CrossRef]
  102. Hosseinkhani H, Azzam T, Kobayashi H, Hiraoka Y, Shimokawa H, Domb AJ, et al. Combination of 3D tissue engineered scaffold and non-viral gene carrier enhance in vitro DNA expression of mesenchymal stem cells. Biomaterials. 2006; 27: 4269-4278. [CrossRef]
  103. Domb AJ, Sharifzadeh G, Nahum V, Hosseinkhani H. Safety evaluation of nanotechnology products. Pharmaceutics. 2021; 13: 1615. [CrossRef]
  104. Hosseinkhani H, Domb AJ. Biodegradable polymers in gene‐silencing technology. Polym Adv Technol. 2019; 30: 2647-2655. [CrossRef]
  105. Hosseinkhani H. Innovation technology to engineer 3D living organs as intelligent diagnostic tools. In: Characterization and development of biosystems and biomaterials. Berlin, Heidelberg: Springer; 2013. pp. 183-192. [CrossRef]
  106. Mottaghitalab F, Rastegari A, Farokhi M, Dinarvand R, Hosseinkhani H, Ou KL, et al. Prospects of siRNA applications in regenerative medicine. Int J Pharm. 2017; 524: 312-329. [CrossRef]
  107. Hosseinkhani H, Domb AJ, Sharifzadeh G, Nahum V. Gene therapy for regenerative medicine. Pharmaceutics. 2023; 15: 856. [CrossRef]
  108. Hosseinkhani H, Aoyama T, Ogawa O, Tabata Y. Ultrasound enhances the transfection of plasmid DNA by non-viral vectors. Curr Pharm Biotechnol. 2003; 4: 109-122. [CrossRef]
  109. Hosseinkhani H, Aoyama T, Ogawa O, Tabata Y. Liver targeting of plasmid DNA by pullulan conjugation based on metal coordination. J Control Release. 2002; 83: 287-302. [CrossRef]
  110. Hosseinkhani H. siRNA delivery systems in cancer therapy. Org Med Chem Int J. 2020; 10: 1-3. [CrossRef]
  111. Dermani FK, Jalilian FA, Hossienkhani H, Ezati R, Amini R. SiRNA delivery technology for cancer therapy: Promise and challenges. Acta Med Iran. 2019; 57: 83-93. [CrossRef]
  112. Hosseinkhani H. Nanomaterials in advanced medicine. Hoboken, NJ: John Wiley & Sons; 2019. [CrossRef]
  113. Hosseinkhani H. Biomedical engineering: Materials, technology, and applications. Hoboken, NJ: John Wiley & Sons; 2022. [CrossRef]
  114. Hosseinkhani H. Developing 3D technology for drug discovery. Curr Drug Deliv. 2022; 19: 813-814. [CrossRef]
  115. Hosseinkhani H, Inatsugu Y, Hiraoka Y, Inoue S, Shimokawa H, Tabata Y. Impregnation of plasmid DNA into three-dimensional scaffolds and medium perfusion enhance in vitro DNA expression of mesenchymal stem cells. Tissue Eng. 2005; 11: 1459-1475. [CrossRef]
  116. Medina-Kauwe L, Xie J, Hamm-Alvarez S. Intracellular trafficking of nonviral vectors. Gene Ther. 2005; 12: 1734-1751. [CrossRef]
  117. Farokhi M, Mottaghitalab F, Shokrgozar MA, Ou KL, Mao C, Hosseinkhani H. Importance of dual delivery systems for bone tissue engineering. J Control Release. 2016; 225: 152-169. [CrossRef]
  118. Mottaghitalab F, Hosseinkhani H, Shokrgozar MA, Mao C, Yang M, Farokhi M. Silk as a potential candidate for bone tissue engineering. J Control Release. 2015; 215: 112-128. [CrossRef]
  119. Hosseinkhani H, Hosseinkhani M, Gabrielson NP, Pack DW, Khademhosseini A, Kobayashi H. DNA nanoparticles encapsulated in 3D tissue‐engineered scaffolds enhance osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A. 2008; 85: 47-60. [CrossRef]
  120. Shahrezaei M, Habibzadeh S, Babaluo AA, Hosseinkhani H, Haghighi M, Hasanzadeh A, et al. Study of synthesis parameters and photocatalytic activity of TiO2 nanostructures. J Exp Nanosci. 2017; 12: 45-61. [CrossRef]
  121. Hosseinkhani H, Chaswal V, Chang J, Shi D. The world scientific encyclopedia of nanomedicine and bioengineering I. 1st ed. Singapore: World Scientific Publishing Company; 2016. pp. 150-220.
  122. Han HC, Lo HC, Wu CY, Chen KH, Chen LC, Ou KL, et al. Nano‐textured fluidic biochip as biological filter for selective survival of neuronal cells. J Biomed Mater Res A. 2015; 103: 2015-2023. [CrossRef]
  123. Konishi M, Tabata Y, Kariya M, Hosseinkhani H, Suzuki A, Fukuhara K, et al. In vivo anti-tumor effect of dual release of cisplatin and Adriamycin from biodegradable gelatin hydrogel. J Control Release. 2005; 103: 7-19. [CrossRef]
  124. Hosseinkhani H. Controlled release system for bone regeneration. In: Polymeric biomaterials. 3rd ed. Boca Raton, Florida: CRC Press; 2020. pp. 643-654.
  125. Baheiraei N, Azami M, Hosseinkhani H. Investigation of magnesium incorporation within gelatin/calcium phosphate nanocomposite scaffold for bone tissue engineering. Int J Appl Ceram Technol. 2015; 12: 245-253. [CrossRef]
  126. Ou SF, Chen CS, Hosseinkhani H, Yu CH, Shen YD, Ou KL. Surface properties of nano-structural silicon-doped carbon films for biomedical applications. Int J Nanotechnol. 2013; 10: 945-958. [CrossRef]
  127. Hosseinkhani H. Special issue on nanomedicine. Int J Nanotechnol. 2011; 8: 615-617.
  128. Hosseinkhani H, Chen KH. Special issue on nanotechnology research in Taiwan. Int J Nanotechnol. 2013; 10: 837-839.
  129. Hosseinkhani H, Tabata Y. Self assembly of DNA nanoparticles with polycations for the delivery of genetic materials into cells. J Nanosci Nanotechnol. 2006; 6: 2320-2328. [CrossRef]
  130. Mahmoudi M, Hosseinkhani H, Hosseinkhani M, Boutry S, Simchi A, Journeay WS, et al. Magnetic resonance imaging tracking of stem cells in vivo using iron oxide nanoparticles as a tool for the advancement of clinical regenerative medicine. Chem Rev. 2011; 111: 253-280. [CrossRef]
  131. Akinc A, Zumbuehl A; Goldberg M, Leshchiner ES, Busini V, Hossain N, et al. A Combinatorial Library of Lipid-Like Materials for Delivery of RNAi Therapeutics. Nat. Biotechnol. 2008; 26: 561-569. [CrossRef]
  132. Hosseinkhani H, Ou KL. The nanoscience in translational medicine. Taiwan, China: Wu-Nan Book Inc.; 2018.
  133. Sharifzadeh G, Hosseinkhani H. Biomolecule‐responsive hydrogels in medicine. Adv Healthc Mater. 2017; 6: 1700801. [CrossRef]
  134. Hosseinkhani H. Potential applications of nanomedicine. Recent Prog Mater. 2023; 5: 036. [CrossRef]
  135. Hosseinkhani H, Ou KL. Advanced biomaterials for biomedical engineering. Taiwan, China: Wu-Nan Book Inc.; 2018.
  136. Hosseinkhani H, Hosseinkhani M, Subramani K. Bone regeneration using self-assembled nanoparticle-based scaffolds. In: Emerging nanotechnologies in dentistry. New York, NY: William Andrew Publishing; 2012. pp. 225-237. [CrossRef]
  137. Hosseinkhani H, Hosseinkhani M. Tissue engineered scaffolds for stem cells and regenerative medicine. Trend in stem cells biology and technology. Totowa, NJ: Humana Press Inc.; 2009. [CrossRef]
  138. Hosseinkhani H, Hosseinkhani M, Zhang S, Subramani K. Self-assembly of nanomaterials for engineering cell microenvironment. Micro and nanoengineering of the cell microenvironment: Applications and technologies. Norwood, MA: Artech House; 2008.
  139. Hosseinkhani H, Hosseinkhani M, Khademhosseini A. Emerging technology of hydrogels in drug discovery. Topics in multifunctional biomaterials and devices. Oulu, Finland: University of Oulu; 2007.
  140. Hosseinkhani H, Aoyama T, Ogawa O, Tabata Y. Tumor targeting of plasmid DNA by dextran conjugation based on metal coordination. Key Eng Mater. 2005; 288: 109-112. [CrossRef]
  141. Yamamoto M, Hosseinkhani H. Liver targeting of plasmid DNA by polymer-metal conjugation. Japan: Yodosha Tech Publications, Japanese version, Med Tech Publications; 2004. pp. 119-132.
  142. Hosseinkhani H, Hosseinkhani M. Design of 3D culture systems to enhance in vitro gene expression of mesenchymal stem cells. Mol Ther. 2009; 17: S270 [CrossRef]
  143. Hosseinkhani H, Hosseinkhani M, Khademhosseini A. A new injectable tissue engineered scaffold induces angiogenesis. Proceedings of the AIChE Annual Meeting; 2007 November 4-9; Salt Palace Convention Center, Salt Lake City, UT. New York, NY: American Institute of Chemical Engineers.
  144. Hosseinkhani M, Hosseinkhani H, Khademhosseini A. Post-translational modification of GATA-4 involved in the differentiation of monkey ES cell into cardiac myocytes. Circulation. 2007; 116: 202-203. [CrossRef]
  145. Hosseinkhani M, Hosseinkhani H. Bone morphogenetic protein-4 enhances cardiomyocyte differentiation of cynomolgus monkey ES cells in knockout serum replacement medium. Eur Heart J. 2007; 28: 230-231. [CrossRef]
  146. Hosseinkhani H, Kobayashi H, Tabata Y. Design of a nano-vessel-like network for controlled proliferation and differentiation of mesenchymal stem cells for regenerative medicine. Tissue Eng. 2006; 12: 993-994.
  147. Hosseinkhani H, Hosseinkhani M, Kobayashi H. A new injectable tissue engineered scaffold for regenerative medicine. Proceedings of the 2006 International Conference on Microtechnologies in Medicine and Biology; 2006 May 9-12; Okinawa, Japan. Piscataway, NJ: IEEE. [CrossRef]
  148. Hosseinkhani H. Selective differentiation of cardiomyocyte cells by using peptide amphiphile nanofibers. Proceedings of the 42nd Japanese Peptide Symposium; 2005 October 27-29; Osaka, Japan. Minoh, Japan: The Japanese Peptide Society. [CrossRef]
  149. Hosseinkhani H. Design of tissue-engineered nano-scaffold using peptide-amphiphile for regenerative medicine. Proceedings of the 42nd Japanese Peptide Symposium; 2005 October 27-29; Osaka, Japan. Minoh, Japan: The Japanese Peptide Society.
  150. Hosseinkhani H. 3D In Vitro Living Systems for Biological Application. ALTEX Proceedings of Animal Alternatives in Teaching, Toxicity Testing and Medicine; 2012 January 3-7; Tiruchirappalli, India.
  151. Hosseinkhani H, Tabata Y. Tumor targeting of plasmid DNA by spermine derivative of dextran combined with ultrasound. Polym Prepr. 2004; 53: 2PE179.
  152. Hosseinkhani H, Tabata Y. Ultrasound enhances expression level of plasmid DNA by PEGylation of cationized dextran in tumor. J Code X0225A. 2004; 19: 299.
  153. Lynn DM, Langer R. Degradable Poly (Beta-Amino Esters): Synthesis, Characterization, and Self-Assembly with Plasmid DNA. J Am Chem Soc. 2000; 122: 10761-10768. [CrossRef]
  154. Vasheghani-Farahani E, Hosseinkhani H, Nekomanesh M. Effect of preparation conditions on theophylline release from biodegradable poly (DL-lactic acid) microspheres. J Control Release. 2001; 72: 287-291.
  155. Tzeng SY, Yang PH, Grayson WL, Green JJ. Synthetic Poly(ester amine) and Poly(amido amine) Nanoparticles for Efficient DNA and siRNA Delivery to Human Endothelial Cells. Int J Nanomed. 2011; 6: 3309-3322. [CrossRef]
  156. Bhise NS, Gray RS, Sunshine JC, Htet S, Ewald AJ, Green JJ. The Relationship between Terminal Functionalization and Molecular Weight of a Gene Delivery Polymer and Transfection Efficacy in Mammary Epithelial 2-D Cultures and 3-D Organotypic Cultures. Biomater. 2010; 31: 8088-8096. [CrossRef]
  157. Hosseinkhani H, Hosseinkhani M. Suppression effect of basic fibroblast growth factor on mesenchymal stem cell proliferation activity-Part I: Release characteristics. Chem Today. 2008; 26: 30-32.
  158. Hosseinkhani H, Hosseinkhani M. Suppression effect of basic fibroblast growth factor on mesenchymal stem cell proliferation activity-Part II: Biological characteristics. Chem Today. 2008; 26: 35-37.
  159. Khalaji S, Golshan Ebrahimi N, Hosseinkhani H. Enhancement of biocompatibility of PVA/HTCC blend polymer with collagen for skin care application. Int J Polym Mater Polym Biomater. 2021; 70: 459-468. [CrossRef]
  160. Hosseini V, Maroufi NF, Saghati S, Asadi N, Darabi M, Ahmad SN, et al. Current progress in hepatic tissue regeneration by tissue engineering. J Transl Med. 2019; 17: 383. [CrossRef]
  161. Saberianpour S, Heidarzadeh M, Geranmayeh MH, Hosseinkhani H, Rahbarghazi R, Nouri M. Tissue engineering strategies for the induction of angiogenesis using biomaterials. J Biol Eng. 2018; 12: 36. [CrossRef]
  162. Toosi S, Naderi-Meshkin H, Kalalinia F, Pievandi MT, Hosseinkhani H, Bahrami AR, et al. Long bone mesenchymal stem cells (Lb-MSCs): Clinically reliable cells for osteo-diseases. Cell Tissue Bank. 2017; 18: 489-500. [CrossRef]
  163. Abbasi N, Hashemi SM, Salehi M, Jahani H, Mowla SJ, Soleimani M, et al. Influence of oriented nanofibrous PCL scaffolds on quantitative gene expression during neural differentiation of mouse embryonic stem cells. J Biomed Mater Res A. 2016; 104: 155-164. [CrossRef]
  164. Toosi S, Naderi‐Meshkin H, Kalalinia F, Peivandi MT, HosseinKhani H, Bahrami AR, et al. PGA‐incorporated collagen: Toward a biodegradable composite scaffold for bone‐tissue engineering. J Biomed Mater Res A. 2016; 104: 2020-2028. [CrossRef]
  165. Toosi S, Naderi-Meshkin H, Kalalinia F, Peivandi MT, Khani HH, Bahrami AR, et al. Comparative characteristics of mesenchymal stem cells derived from reamer-irrigator-aspirator, iliac crest bone marrow, and adipose tissue. Cell Mol Biol. 2016; 62: 68-74.
  166. Jahani H, Jalilian FA, Wu CY, Kaviani S, Soleimani M, Abbasi N, et al. Controlled surface morphology and hydrophilicity of polycaprolactone toward selective differentiation of mesenchymal stem cells to neural like cells. J Biomed Mater Res A. 2015; 103: 1875-1881. [CrossRef]
  167. Rahbarghazi R, Nassiri SM, Ahmadi SH, Mohammadi E, Rabbani S, Araghi A, et al. Dynamic induction of pro-angiogenic milieu after transplantation of marrow-derived mesenchymal stem cells in experimental myocardial infarction. Int J Cardiol. 2014; 173: 453-466. [CrossRef]
  168. Ghodsizadeh A, Hosseinkhani H, Piryaei A, Pournasr B, Najarasl M, Hiraoka Y, et al. Galactosylated collagen matrix enhanced in vitro maturation of human embryonic stem cell-derived hepatocyte-like cells. Biotechnol Lett. 2014; 36: 1095-1106. [CrossRef]
  169. Shi D, Tatu R, Liu Q, Hosseinkhani H. Stem cell-based tissue engineering for regenerative medicine. Nano Life. 2014; 4: 1430001. [CrossRef]
  170. Ou KL, Hosseinkhani H. Development of 3D in vitro technology for medical applications. Int J Mol Sci. 2014; 15: 17938-17962. [CrossRef]
  171. Hosseinkhani H, Hong PD, Yu DS. Self-assembled proteins and peptides for regenerative medicine. Chem Rev. 2013; 113: 4837-4861. [CrossRef]
  172. Hosseinkhani H, Hiraoka Y, Li CH, Chen YR, Yu DS, Hong PD, et al. Engineering three-dimensional collagen-IKVAV matrix to mimic neural microenvironment. ACS Chem Neurosci. 2013; 4: 1229-1235. [CrossRef]
  173. Hosseinkhani H. 3D in vitro technology for drug discovery. Curr Drug Saf. 2012; 7: 37-43. [CrossRef]
  174. Subramani K, Mathew R, Hosseinkhani H, Hosseinkhani M. Bone regeneration around dental implants as a treatment for peri-implantitis: A review of the literature. J Biomim Biomater Tissue Eng. 2011; 11: 21-33. [CrossRef]
  175. Hosseinkhani H, Hosseinkhani M, Hattori S, Matsuoka R, Kawaguchi N. Micro and nano‐scale in vitro 3D culture system for cardiac stem cells. J Biomed Mater Res A. 2010; 94: 1-8. [CrossRef]
  176. Mohajeri S, Hosseinkhani H, Golshan Ebrahimi N, Nikfarjam L, Soleimani M, Kajbafzadeh AM. Proliferation and differentiation of mesenchymal stem cell on collagen sponge reinforced with polypropylene/polyethylene terephthalate blend fibers. Tissue Eng Part A. 2010; 16: 3821-3830. [CrossRef]
  177. Tian F, Hosseinkhani H, Hosseinkhani M, Khademhosseini A, Yokoyama Y, Estrada GG, et al. Quantitative analysis of cell adhesion on aligned micro‐and nanofibers. J Biomed Mater Res A. 2008; 84: 291-299. [CrossRef]
  178. Hosseinkhani H, Hosseinkhani M, Tian F, Kobayashi H, Tabata Y. Osteogenic differentiation of mesenchymal stem cells in self-assembled peptide-amphiphile nanofibers. Biomaterials. 2006; 27: 4079-4086. [CrossRef]
  179. Bielinska AU, Kukowska-Latallo JF, Baker Jr JR. The interaction of plasmid DNA with polyamidoamine dendrimers: mechanism of complex formation and analysis of alterations induced in nuclease sensitivity and transcriptional activity of the complexed DNA. Biochim Biophys Acta Gene Struct Expression. 1997; 1353: 180-190. [CrossRef]
  180. Hosseinkhani H, Hosseinkhani M, Tian F, Kobayashi H, Tabata Y. Ectopic bone formation in collagen sponge self-assembled peptide–amphiphile nanofibers hybrid scaffold in a perfusion culture bioreactor. Biomaterials. 2006; 27: 5089-5098. [CrossRef]
  181. Hosseinkhani H, Hosseinkhani M, Khademhosseini A, Kobayashi H, Tabata Y. Enhanced angiogenesis through controlled release of basic fibroblast growth factor from peptide amphiphile for tissue regeneration. Biomaterials. 2006; 27: 5836-5844. [CrossRef]
  182. Hosseinkhani H, Hosseinkhani M, Kobayashi H. Proliferation and differentiation of mesenchymal stem cells using self-assembled peptide amphiphile nanofibers. Biomed Mater. 2006; 1: 8. [CrossRef]
  183. Hosseinkhani H, Hosseinkhani M, Kobayashi H. Design of tissue-engineered nanoscaffold through self-assembly of peptide amphiphile. J Bioact Compat Polym. 2006; 21: 277-296. [CrossRef]
  184. Hosseinkhani H, Hosseinkhani M, Khademhosseini A. Emerging applications of hydrogels and microscale technologies in drug discovery. Drug Discov. 2006; 1: 32-34.
  185. Hosseinkhani H, Inatsugu Y, Hiraoka Y, Inoue S, Tabata Y. Perfusion culture enhances osteogenic differentiation of rat mesenchymal stem cells in collagen sponge reinforced with poly (glycolic acid) fiber. Tissue Eng. 2005; 11: 1476-1488. [CrossRef]
  186. Hosseinkhani H, Hong PD, Yu DS, Chen YR, Ickowicz D, Farber IY, et al. Development of 3D in vitro platform technology to engineer mesenchymal stem cells. Int J Nanomed. 2012; 7: 3035-3043. [CrossRef]
  187. Liu YM, Reineke TM. Hydroxyl stereochemistry and amine number within poly(glycoamidoamine)s affect intracellular DNA delivery. J Am Chem Soc. 2005; 127: 3004-3015. [CrossRef]
  188. Kloeckner J, Wagner E, Ogris M. Degradable gene carriers based on oligomerized polyamines. Eur J Pharm Sci. 2006; 29: 414-425. [CrossRef]
  189. Hosseinkhani H. 3‐D culture systems for cell culture technology. J Cell Res. 2018; 1: 1-4.
  190. Gholivand MB, Mohammadi-Behzad L, Hosseinkhani H. Application of a Cu-chitosan/multiwalled carbon nanotube film-modified electrode for the sensitive determination of rutin. Anal Biochem. 2016; 493: 35-43. [CrossRef]
  191. Huang CF, Chiang HJ, Lin HJ, Hosseinkhani H, Ou KL, Peng PW. Comparison of cell response and surface characteristics on titanium implant with SLA and SLAffinity functionalization. J Electrochem Soc. 2014; 161: G15. [CrossRef]
  192. Hosseinkhani H, Chen KH. On nanotechnology research in Taiwan. Int J Nanotechnol. 2013; 10: 191.
  193. Wong SY, Sood N, Putnam D. Combinatorial Evaluation of Cations, pH-sensitive and Hydrophobic Moieties for Polymeric Vector Design. Mol Ther. 2009; 17: 480-490. [CrossRef]
  194. Gonzalez H, Hwang SJ, Davis ME. New class of polymers for the delivery of macromolecular therapeutics. Bioconjug Chem. 1999; 10: 1068-1074. [CrossRef]
  195. Subramani K, Pathak S, Hosseinkhani H. Recent trends in diabetes treatment using nanotechnology. Dig J Nanomater Biostruct. 2012; 7: 85-95.
  196. Kalhor HR, Shahin VF, Fouani MH, Hosseinkhani H. Self-assembly of tissue transglutaminase into amyloid-like fibrils using physiological concentration of Ca2+. Langmuir. 2011; 27: 10776-10784. [CrossRef]
  197. Amini R, Rosli R, Abdullah S, Hosseinkhani H, Veerakumarasivam A. Delivery of plasmid expressing green fluorescent protein by PEGylated dextran‐spermine to acute myeloid leukemic cells. Med J Malaysia. 2010; 65: 118-120.
  198. Balbarini A, Magera A, Barsotti MC, Hosseinkhani H, Chiellini F, Solaro R, et al. Self assembling peptide amphiphile nanofibers as a scaffold for endothelial progenitor cells. J Am Coll Cardiol. 2010; 55: A176.E1652. [CrossRef]
  199. Lindstrom S, Iles A, Persson J, Hosseinkhani H, Hosseinkhani M, Khademhosseini A, et al. Nanoporous titania coating of microwell chips for stem cell culture and analysis. J Biomech Sci Eng. 2010; 5: 272-279. [CrossRef]
  200. Ebrahimi N, Soleimani AA, Rashidiani J, Malekafzali B, Abedini F, Hosseinkhani H. Chitosan/Fluoride Nanoparticles for Preventing Dental Caries. Curr Dent. 2019; 1: 61-67. [CrossRef]
  201. Das J, Han JW, Choi YJ, Song H, Cho SG, Park C, et al. Cationic lipid-nanoceria hybrids, a novel nonviral vector-mediated gene delivery into mammalian cells: Investigation of the cellular uptake mechanism. Sci Rep. 2016; 6: 29197. [CrossRef]
  202. Subramani K, Hosseinkhani H, Khraisat A, Hosseinkhani M, Pathak Y. Targeting nanoparticles as drug delivery systems for cancer treatment. Curr Nanosci. 2009; 5: 135-140. [CrossRef]
  203. Kreofsky NW, Roy P, Reineke TM. pH-Responsive Micelles Containing Quinine Functionalities Enhance Intracellular Gene Delivery and Expression. Bioconjug Chem. 2024; 35: 1762-1778. [CrossRef]
  204. Hosseinkhani H, Hosseinkhani M, Farahani EV, Haghighi MN. In vitro sustained release and degradation study of biodegradable poly (D, L-lactic acid) microspheres loading theophylline. Adv Sci Lett. 2009; 2: 70-77. [CrossRef]
  205. Di Stefano R, Barsotti MC, Magera A, Hosseinkhani H, Chiellini F, Dinucci D, et al. A biological self assembling amphiphilic peptide enhances endothelial progenitor cells growth and paracrine releaser. Eur Heart J. 2009; 29. Available from: https://hdl.handle.net/11568/134562.
  206. Barsotti MC, Di Stefano R, Magera A, Alderighi M, Chiellini F, Dinucci D, et al. A biological self assembling amphiphilic peptide with pro-angiogenic function. Proceedings of the II Convegno Nazionale INSTM sulla Scienza e Tecnologia dei Materiali; 2009 June 9-12; Tirrenia, Italy. Sassari, Italy: IRIS.
  207. Simonetti E, Barsotti MC, Magera A, Hosseinkhani H, Sorbo S, Dinucci D, et al. A biological self-assembling peptide suitable for cell delivery in ischemic tissues. Proceedings of the XV Congresso Nazionale della Società Italiana di Ricerche Cardiologiche; 2008 October 9-11; Imola, Italy. Sassari, Italy: IRIS.
  208. Di Stefano R, Barsotti MC, Magera A, Hosseinkhani H, Lemmi M, Arici R, et al. Biological self-assembling peptide amphiphile nanofiber scaffold enhances endothelial progenitor cell growth. G Ital Cardiol. 2008; 12: 56s.
  209. Hosseinkhani H. Towards the development of advanced medicine by use of nanotechnology. Cell J. 2008; 10: 34-35.
  210. Tian F, Hosseinkhani H, Estrada GG, Kobayashi H. Quantitative method for the analysis of cell attachment using aligned scaffold structures. J Phys Conf Ser. 2007; 61: 587. [CrossRef]
  211. Hosseinkhani M, Hosseinkhani H, Khademhosseini A, Bolland F, Kobayashi H, Gonzalez SP. Bone morphogenetic protein-4 enhances cardiomyocyte differentiation of cynomolgus monkey ESCs in knockout serum replacement medium. Stem Cells. 2007; 25: 571-580. [CrossRef]
  212. Hosseinkhani H, Hosseinkhani M, Khademhosseini A. Tissue regeneration through self-assembled peptide amphiphile nanofibers. Cell J. 2006; 8: 204-209. [CrossRef]
  213. Hosseinkhani H. Selective differentiation of cardiomyocytes by using self-assembly of peptide amphiphile nanofiber nanoscaffolds. Cell J. 2005; 7: 38-39.
  214. Di Stefano R, Magera A, Barsotti MC, Felice F, Hosseinkhani H, Chiellini F, et al. Endothelial progenitor cell differentiation and paracrine activity on self-assembling peptide amphiphile nanofibers. Tissue engineering and regenerative medicine international society - EU meeting. Galway, Ireland: University of Galway; 2010.
  215. Yamanouchi D, Wu J, Lazar AN, Kent KC, Chu CC, Liu B. Biodegradable arginine-based poly (ester-amide) s as non-viral gene delivery reagents. Biomaterials. 2008; 29: 3269-3277. [CrossRef]
  216. Wu J, Yamanouchi D, Liu B, Chu CC. Biodegradable arginine-based poly (ether ester amide) s as a non-viral DNA delivery vector and their structure-function study. J Mater Chem. 2012; 22: 18983-18991. [CrossRef]
  217. Zhou Y, Han S, Liang Z, Zhao M, Liu G, Wu J. Progress in arginine-based gene delivery systems. J Mater Chem B. 2020; 8: 5564-5577. [CrossRef]
  218. You X, Gu Z, Huang J, Kang Y, Chu CC, Wu J. Arginine-based poly (ester amide) nanoparticle platform: From structure-property relationship to nucleic acid delivery. Acta Biomater. 2018; 74: 180-191. [CrossRef]
  219. Liu Y, Gunda V, Zhu X, Xu X, Wu J, Askhatova D, et al. Theranostic near-infrared fluorescent nanoplatform for imaging and systemic siRNA delivery to metastatic anaplastic thyroid cancer. Proc Natl Acad Sci. 2016; 113: 7750-7755. [CrossRef]
  220. Xu X, Wu J, Liu Y, Saw PE, Tao W, Yu M, et al. Multifunctional envelope-type siRNA delivery nanoparticle platform for prostate cancer therapy. ACS Nano. 2017; 11: 2618-2627. [CrossRef]
  221. Zhang L, Dong Z, Yu S, Li G, Kong W, Liu W, et al. Ionic liquid-based in situ dynamically self-assembled cationic lipid nanocomplexes (CLNs) for enhanced intranasal siRNA delivery. Chin Chem Lett. 2024; 35: 109101. [CrossRef]
Newsletter
Download PDF Download Citation
0 0

TOP