Skip to main content
Iranian Journal of Basic Medical Sciences logoLink to Iranian Journal of Basic Medical Sciences
. 2022 Feb;25(2):134–147. doi: 10.22038/IJBMS.2022.59718.13245

Recent advances in development of nano-carriers for immunogene therapy in various complex disorders

Sanaz Shahgordi 1,2, Fatemeh Oroojalian 3, Ezzat Hashemi 4, Maryam Hashemi 2,5,*
PMCID: PMC9124536  PMID: 35655600

Abstract

Immunotherapy is a novel preference for the treatment of various complex diseases. Considering the application of varying agents for suppression or activation of the immune system, immunogene therapy was confirmed to stand as a proper alternative for other immunotherapeutic strategies due to its capability in targeting cells with more specificity that leads to controlling the expression of therapeutic genes. This method facilitates the local and single-dose application of most gene therapies that result in the usage of high therapeutic doses with a low risk of systemic side effects while being cost-efficient in long-term administrations. However, the existing barriers between the administration site and cell nucleus limited the clinical uses of genetic materials. These challenges can be overcome through the promising method of exerting non-carriers with high stability, low toxicity/immunogenicity, and simple modifications. In this study, we attempted to review the potential of nanoparticle application throughout the immunogene therapy of different diseases including cancer, microbial diseases, allergies, inflammatory bowel disease, rheumatoid arthritis, and respiratory infections. We included the outline of some challenges and opportunities in regards to the delivery of genetic materials that are based on nano-systems through immunotherapy of these disorders. Next to the promising future of these vectors, more detailed analyses are required to overcome the current limitations in clinical approaches.

Key Words: Gene delivery, Immunogene therapy, Immunotherapy, Nano-carriers

Introduction

Immunotherapy is an effective strategy for preventing and treating various disorders including viral infections, allergies, cancer, and autoimmune diseases ( 1 ). The application of numerous agents was trialed for suppression or activation of the immune system such as modified dendritic cells, monoclonal antibodies, stem cells, peptides, nucleic acids (NAs), and nanoparticles (NPs) ( 2 ).

The procedure of gene therapy attempted to introduce exogenous NAs into specific cells for the modulation of gene expression, which could also counteract or replace a malfunctioning gene ( 3 ). Immunogene therapy is a novel preference for the treatment of various complex disorders and a proper alternative for other immunotherapeutic strategies. This method is capable of targeting cells with more specificity, which leads to controlling the expression of therapeutic genes. Consequently, this route facilitates the local and single-dose application of most gene therapies that result in the usage of high therapeutic doses with a low risk of systemic side effects, while being cost-efficient in long-term administration.

Therapeutic genetic materials are categorized into DNA (Deoxyribonucleic acid) and RNA (Ribonucleic acid) types with limited clinical uses due to the existing barriers between the administration site and cell nucleus. The delivery efficiency of genetic materials was attempted to be improved through the development of various gene carriers, which are required to have the potential of overcoming certain obstacles such as containing the ability to circulat e in the bloodstream, remain undetected by the immune system, survive in the extracellular environment, successfully cross the cell membrane, protect the NAs from nuclease degradation, and release the functional form of NAs in the nucleus ( 4 , 5 ).

The application of viral vectors with high transfection efficiency is extensively considered for gene delivery purposes. However, there are disadvantages to this approach such as irregular cytotoxicity and immune system stimulation, limited targeting of specific cell types, low DNA carrying capacity, inability to infect non-dividing cells ( 6 ), mutagenesis ( 7 , 8 ), high risk of wild-type virus reversion (9, 10), and inflammation ( 11 , 12 ). Therefore, nano-carriers ( 13 ) attracted the focus of many owing to their high stability, low toxicity, decreased immune responses , flexib le designs, and simple modifications ( 14 - 16 ). On the other hand, the main challenge of nano-delivery systems is to develop a delivery vehicle with high cellular uptake efficiency and the ability to promote endosomal escape, inhibit its transference to lysosomes, and ensure its localization in appropriate cellular compartments ( 17 ).

NPs proved to stand as a promising method for overcoming the mentioned challenges (NPs) due to offering notable advantages such as the ability to prevent degradation, deliver high concentrations of the payload for targeting the desired site, and function as a controlled release system ( 18 - 20 ). All of the nano-carriers that were exerted in immunogene therapy, including polymeric NPs, lipid-based NPs, liposomes, carbon nanotubes, and gold NPs ( 3 , 21 , 22 ), could be designed to produce efficient long-term immune responses ( 23 ).

The objective of this study was to review the applications of nano-carriers, which are based on immunogene therapy, for the treatment of various diseases.

Genetic materials used in immunotherapy

Various genetic materials were applied in immunogene therapy including synthetic oligonucleotide (ODN), plasmid DNA, and RNA interference (RNAi).

Oligonucleotides

ODNs are known as short, single-stranded synthetic DNA molecules and according to several studies, they contain unmethylated CpG (Cytosine-Phosphate-Guanine) motifs that are capable of arranging potent immune systems, which consequently improve the obtained responses to vaccines, reduce allergic responses, and provide protective effects in immunotherapy and cancer therapy (24). Figure 1 depicts the action mechanisms of ODNs throughout the immune system.

Figure 1.

Mechanism of effects of ODNs on immune system

Figure 1

As CpG ODN oligonucleotides are internalized in APCs, activated APCs release various cytokines and chemokines, causing effector immune cell differentiation and activation; TLR: toll-like receptor, IRF: interferon regulatory transcription factor, NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells, Mon: monocytes, MQ: macrophage

Recognized by toll-like receptor 9 (TLR9) ( 25 ) in antigen-presenting cells (APCs), the activation of CpG ODN oligonucleotides requires the Myeloid differentiation primary response 88(MYD88)-IRF7 (interferon regulatory transcription factor) signaling, which leads to up-regulation of IRF and nuclear factor- κB (NF- κB ). The activated APCs are able to stimulate the secretion of immunomodulatory cytokines, including interleukin-6 (IL-6), IL-1, IL-2, IL-10, IL-12, IL-18, Tumor necrosis factor-α (TNFα), and Interferon-α/β (IFN α / β ) ( 26 ), that consequently activate the innate immune effector cells (e.g., natural killer cells [NK]). These cytokines provide the production of other immune-stimulatory cytokines and chemokines, as well as improve the adoptive immunity through the induction of Ag-specific T cells ( 27 - 29 ) and increase neutrophil migration, monocyte, and macrophage activation, and acute-phase protein production ( 30 , 31 ). Another capability of CpG ODNs is to initiate the activation of B cells as an APC in order to produce IgM and cytokines such as IL-6 and IL-10. Additionally, IL-6 can facilitate the activation and differentiation of long-lived plasma cells for secret ion of other kinds of antibodies such as IgG ( 32 ).

According to recent studies, CpG formulations could inhibit immune responses through enzyme up-regulation of indoleamine-pyrrole 2, 3-dioxygenase (IDO), and T regulatory cell (Treg)-mediated suppression ( 33 ). The induction of general immune responses stands as the main difficulty of naked CpGs, which is caused by the release of inflammatory cytokines and systemic activation of lymphocytes ( 33 ). However, this issue can be surpassed by exertion of specific vehicles designed for carry ing CpGs.

Plasmid DNA

Another candidate for immunogene therapy is plasmid DNA (pDNA) due to its ability in expressing particular antigens (34). The produced antigens can increase the antigen expression of traveling APCs towards draining lymph nodes that present the antigenic peptide-MHC complexes for T cell stimulation, which results in the generation of long-term cellular immunity. Furthermore, humoral immunity can be generated through this process since it initiates the antibody production cascade (35-38). The sequences of CpG throughout bacterial plasmid were exerted as potent adjuvants in pDNA-mediated vaccination (25, 39, 40). In addition to adaptive immunity, the unmethylated CpG oligonucleotide sequences can turn the functionality of pDNA into a potent innate immunity inducer. Figure 2 displays the immune cell modulation that involves the internalization of NPs, which carry the plasmids throughout the suppression of allergic reactions.

Figure 2.

Figure 2

Immune cell modulation through internalization of NP carrying plasmids in suppression of allergic reactions

The internalized NPs, which carry plasmid DNA into the targeted APCs end up engulfed by endosomes and the plasmid DNA is translated to the intended protein to result in the activation of APCs. The activated APCs induce T helper cell (Th1) differentiation to suppress the Th2 cells through IFN-γ secretion and decrease the allergic cytokine secretions (IL-4 and IL-5). Moreover, these APCs can create Treg cells with the ability to suppress Th2 cells, as well as arrange the secretion of IgG2a and IgA from B cells through inhibitory cytokines, suppress mucus production, and inhibit other allergic immune cells such as basophils, mast cells, and eosinophils ( 41 , 42 ).

RNA interference (RNAi)

RNA interference (RNAi) mediated by short interfering RNA (siRNA), short hairpin RNA (shRNA), and microRNA (miRNA) ( 43 ) is a natural cellular process that acts as a gene silencer throughout the eukaryotic system at transcriptional, posttranscriptional, or translational levels ( 44 - 46 ). In this process, siRNA molecules function as the key mediators for inhibiting target gene expression s. Owing to their high efficiency, these molecules are considered promising gene silencers and biotherapeutic agents for a wide array of diseases. In addition, siRNA can serve as the guide sequence for creating the cleavage of target-specific Messenger RNA (mRNA) through activation of highly conserved regulatory mechanisms ( 47 ). It has been strongly proven that siRNA can be recognized by endosomal (e.g., TLR 3, 7, 8) and cytoplasmic pathways such as retinoic acid-inducible gene I, melanoma differentiation-associated antigen 5, and RNA-activated protein kinase. This process leads to activation of various transcription factors, including NF- κB , interferon regulatory factor 3 (IRF-3), and IRF-7, and consequently result in IFN secretion. Furthermore, T cells, NK cells, and DCs can be also activated through this process ( 48 ).

Figure 3 demonstrates the silencing mechanism of programmed death-ligand 1 (PDL1) by PDL-1 siRNA which is a model for reduction of tumor cell survival. Accordingly, long double-stranded RNA (dsRNA) was cleaved through the dicer enzyme (endoribonuclease) next to the production of siRNA , which enabled the cells to assemble the RNA-induced silencing complex (RISC). siRNA becomes unwound upon incorporation of siRNA into RISC and forms a single-strand siRNA that binds to its PDL1 complementary mRNA and creates mRNA cleavage; considerably, this type of mRNA cannot be translated to PDL1 protein ( 49 ). Due to lack of PDL1 expression on tumor cell surfaces, immune cells with PD1 on their surface cause a reduction in tumor cell survival, which include cytotoxic T cells (CTL), macrophages (MQ), NK cells, and invariant natural killer T cells (iNKT) ( 50 , 51 ).

Figure 3.

Figure 3

Silencing of programmed death-ligand 1 (PDL1) to decrease tumor cell survival

Nano-carriers in immunogene therapy

Nowadays, various biodegradable and biocompatible NPs are exerted in the form of gene delivery vehicles. In addition to their applications as protective genetic materials against enzymatic degradation, nano-carriers can efficiently deliver therapeutic agents to the targeted cells. In the following section, we discussed the potential of nano-carriers in immunogene therapy of various diseases including cancer, microbial diseases, allergies, inflammatory bowel disease, rheumatoid arthritis, and respiratory infections.

Cancer

According to recent findings, the applied nano-carriers for gene delivery are capable of modulating immune responses and affecting the tumor microenvironment (TME). In this regard, s everal studies attempted to focus on this method for treatment of colon and ovarian cancers and melanoma. Figure 4 exhibits the immunomodulatory effects of NPs on cancer. It can be observed that the entrance of NPs containing various immune modulatory genes into the tumor environment leads to the occurrence of changes including the minimized infiltration of tumor-associated microphages (TAM), modulation of cellular immunity cells (e.g., CTL, Treg, and B lymphocytes), innate immune cell modulation (e.g., NK cells), and modulated secretion of soluble mediators from immune cells (e.g., cytokines and chemokines). All of these alterations may result in to tumor cell necrosis or apoptosis.

Figure 4.

Figure 4

Immune modulatory effects of NPs in the tumor microenvironment

Table 1 displays some of the exerted nano-carriers for forming a complex with various genetic materials for cancer immunotherapy.

Table 1.

Nanoparticles (NPs)-based immunogene delivery used in cancer treatment

Cancer type Gene therapeutic agents NPs Functional mechanisms Ref.
Melanoma Tlr9 agonist
Trp2 peptide
Liposome High-activity CTL generation
DC Activation
(52)
Melanoma Tlr9 agonist
Trp2 peptide
Lipid-calcium-phosphate (LCP) Modest CTL response
Moderate DC activation
(53)
Melanoma Tlr9 agonist Gelatinase NP T Cell activation
Immune response amplification
(33)
Melanoma Hsp-70 gene Magnetic cationic liposomes (MCL) CTL activation
Elicit Pro-inflammatory cytokines
MHC-I Up-regulation
(54)
Melanoma C-myc Antisense Liposome Immune Cell activation
Increased CD69,86 expression
Increased CD8⁺ T Cells
Increased IFN-γ, IgG1, and IgG2a secretion
Induction of humoral immune responses
Induction of balance between Th1 and Th2
Breaking self-antigen tolerance
(26)
Melanoma Ovalbumin Lipid NP Increased CD8⁺ T Cell levels
Increased innate immune system pattern Recognition receptors
Type I IFN up-regulation
(55)
Melanoma Ovalbumin-TLR9 ligand MgAl layered double hydroxide
(MgAL-LDH)
Increased IgG2a/IgG1 ratio
Induced Th1/Th2 balance
Increased CD8⁺ T Cell recruitment
(56)
Leukemia PDL1-siRNA Lipid NP Increased CD8⁺ T Cell proliferation
CTL degranulation upon Ag re-stimulation
(57)
Leukemia β-galactosidase Chitosan Increased MMP-9 levels
No Pro-inflammatory and IL-10 changes
APC activation
(58)
Leukemia IL-12 Chitosan Increased IL-12 and IFN-γ levels
Modulated immune system
(59)
Leukemia IL-12 LNP (lipid nanoparticles) Increased IL-12 and IFN-γ levels
Activated innate immunity
Recruited monocytes, NK cells, and T cells
Activated MYD88 and STING signaling pathways
(60)
Ovarian cancer PDL1-siRNA Polyethylenimine 
(PEI)
TLR 3, 5, 7 and MYD88 stimulation
IFN-γ secretion
Innate immune system activation
Tumor-associated DC activation
Silenced PDL-1
(50)
Ovarian cancer PDL1-siRNA PEI PDL-1 knockdown
Increased CAR-T Cell function
(51)
Ovarian cancer miR-155 ds-RNA duplexes PEI DC activation
Increased Ag presentation
Produced Th1 cytokines
Tumor-reactive T cell expansion
(61)
Ovarian cancer IL-12 Liposome Prime antitumor immunity
Increased CD8⁺ T Cell levels
Increased perforin-positive Cell Infiltration to TME
(62)
Lung cancer Anti-VEGF siRNA Gold NP Decreased MQs in lung tissues
No changes in cytokine levels or TLR activation
Modulate TAM recruitment
(63)
Lung cancer IL-12 PEI-DNA NP Increased IL-12 and IFN-γ levels
Activate antitumor immune responses
(64)
Lung cancer IL-12 tumor-targeted micelleplexes Increased IL-12, IFNγ and TNFα levels
T4+, T+, and NK cell activation
Switching M2 MQs into M1 MQs
(65)
Hepatocellular carcinoma IL-12 PLGA DOTAP Increased IL-12 and IFN-γ levels
Induced immune responses
NK cell activation
(66)
Hepatocellular carcinoma PDL-1
IL-2
Tumor targeted lipid-dendrimer-calcium-phosphate
(TT-LDCP NP)
Increased IL-2 level
Decreased PD-L1 level
Increased T CD8⁺ activation and infiltration to TME
(67)
Hepatocellular carcinoma IL-12 PAMAM Increased IL-12 level (68)
Pancreatic cancer HIFα siRNA Lipid polymer hybrid NP Decreased IL-6 and IFN-γ Levels (69)
Colon cancer IL-12 DMP micelle T cell infiltration to the tumor microenvironment
Increased TNF-α levels
(70)
Colon cancer IL-12 Cholesterol-conjugated PEGylated PAMAM Stimulated TH1 differentiation (71)
Colorectal cancer CCL19 Folate-modified nanoparticle Increased TNF-α and IFN-γ secretion
Activated T cells
Decreased T regulatory cells
Decreased M2 macrophages
Decreased MDSCs
(72)
Colon cancer IL-15 Protamine-liposome system (CLPP) Increased NK cells
Activated TCD8⁺ cells
Increased TNF-α secretion
(73)
Brain cancer TGFβ Polybutyl cyanoacrylate nanoparticles Increased T CD25+ cells
Decreased TGFβ2 levels
(74)

Melanoma

Melanoma has yielded numerous immunogenic epitopes such as tyrosinase-related protein (TRP-2) in the form of a tumor-associated antigens ( 52 ) and low levels of MHC-I ( 75 ). Application of MHC-I-dependent CTLs proved to be essential for suppression of tumor growth and inhibition of tumor recurrence. Down-regulation of MHC-I is a fundamental mechanism for the escape of tumor cells from T-cell-mediated anti-tumor immunity ( 76 , 77 ).

Xu et al . reported the usage of TRP-2-loaded lipid-calcium-phosphate (LCP) NPs in conjugation with CpG ODN (TLR-9 agonist). Accordingly, the surface of NPs was modified with mannose due to expression of its receptor on DCs. The prepared formulation consisted of 50 µg of TRP-2 antigen, and 20 µg of CpG was administered to melanoma-bearing female C57BL/6 mice. In comparison with the other groups, the obtained results were indicative of modest CTL response, mild DC activation signals, and modest antitumor activity (53).

In another study, the melanoma-bearing C57BL/6 mice were pretreated with Heat shock proteins (HSP-70) gene (total amount: 20 µg/mouse), which was combined with induced hyperthermia by magnetic cationic liposomes (MCLs) , and succeeded in strongly arresting tumor growth; presumably, activation of CTL through MHC-I up-regulation could be the reason behind this achievement ( 54 ). HSP-70 is also capable of acting as an extracellular protein with regulatory effects on monocytes eliciting proinflammatory cytokines for providing innate immunity ( 78 ).

According to the findings of Oberli et al . the administration of a lipid NP complex with unmodified mRNA-encoding ovalbumin (10 µg of mRNA/mouse) to melanoma-bearing C57BL/6 mice was resulted in blocking tumor growth through the induction of IFN-I up-regulation comparing with control group. IFN-I is an essential factor for CD8⁺ T cell protective immunity. This data could be transfected to various immune cells such as DCs, MQs, Neutrophil (NEU), and B cells due to resulting in strong activation of CD 8⁺ T cells. In this regard, the gathered evidence confirmed the effectiveness of this formulation as an immune system activator and modulator ( 55 ).

Another related research was performed and in which the exertion of MgAl-layered double hydroxide (LDH) with a similar chemical composition to Alum was applied in conjugation with TLR-9 ligand CpG and ovalbumin (OVA) for the treatment of melanoma in female C57BL/6 mice. The findings indicated that this nanoformulation induced an approximately 6-fold higher IgG2a/IgG1 ratio than Alum-CpG-OVA ( P <0.01), which can strongly direct the immune responses towards Th1 ( 56 ) . LDH is capable of effectively causing immune responses through several mechanisms including the efficient loading and protection of OVA and CpG, as well as increased uptake of these cargos by APCs and endosomal processing, in which the released CpG interacts with TLR-9 receptor on the surface of endosomes ( 79 ). The proposed formulation could also increase the recruitment of CD8⁺ T-cell to TME and consequently block tumor growth. In the mentioned study, the application of MgAl-LDH with immune modulation can efficiently deliver Ag and CpG to APCs in order to provoke the immune system ( 56 ).

Li et al . reported that the administration of self-replicated IL-12 RNA along with lipid nanoparticles (LNP) to C57BL6/J and BALB/C mice (10 µg/mice) was lead to a higher IL-12 and IFNγ expression and recruited monocytes, as well as observed the presence of NK cells and T cells throughout the tumor microenvironment and reported the inducement of STING and MYD88 signaling pathway. Furthermore, the outcomes of in vitro assays on B16F10 tumor cells indicated the occurrence of innate immunity activation caused by the upregulation of TLR3, MDA5 and IFNI along with an increase in IL-12 gene expression. In total, this therapy could modulate tumor microenvironment and increase the rate of immunogenic cell death ( 60 ).

Leukemia

Leukemia is a malignant disease that involves the release of an increased number of immature or abnormal leukocytes into the bloodstream or bone marrow. DC vaccines function as T-cell immune response enhancers and stand among the optimal treatment options for leukemia.

A group of researchers conducted an study on human DC vaccine with an improved immunogenic potential by forcing siRNAs to target the incorporated PD-L genes into the lipid NPs (LNPs) in conjugation with Minor histocompatibility antigen (MiHA, a key Ag in the antitumor responses expressed by malignant cells) and mRNA electroporation. Moreover, Immature DCs (iDCs) were generated by culturing the isolated monocytes from peripheral blood mononuclear cells. Their findings pointed out the enhanced proliferation of Ag-specific CD8⁺ T cells in comparison with the control group. However, these specialized DCs were not capable of expressing PDL-1, while the activated T cells remained in highly activated states. In addition, the expanded MiHA-specific CD8⁺ T cells could be efficiently de-granulated upon Ag re-stimulation. In the mentioned study, LNPs were exerted due to their ability to efficiently encapsulate and deliver siRNA through the cell membrane ( 57 ) . Moreover, considering the common usage of monocytes for the generation of DCs ( 79 ), NPs were covered by Polyethylene glycol (PEG) in order to take advantage of its transfection efficiency for human monocyte-derived DCs ( 57 ).

CAR-T (chimeric antigen receptor) cells are T lymphocytes engineered in laborator ies to express the artificial receptors that would be targeted towards specific antigens in tumor cells.

According to another findings, the delivery of PDL-1-siRNA by the combination of PEI NPs with anti-PDL-1 antibody was results in knockdown of PDL-1 on epithelial ovarian cancer (EOC) cells, which consequently increases the functionality of chimeric CAR-T through higher IFN-γ secretion ( P <0.01) and CD107a expression on the surface of CAR-T cells ( P <0.05). CD107a is a lysosomal marker that involves the degranulation activity of CAR-T cells ( 51 ). However, CAR-T cell therapy has been undermined by various immunosuppressive mechanisms that are based on EOC cells, as well as the interaction between PD-1 and its ligand that leads to the hyporesponsiveness of T-cells ( 80 - 83 ). Therefore, this method can modulate the immune system through the blockage of PDL-1 on ovarian cancer cells.

Previous findings confirmed the essentiality of miRNAs for regulation of every immune cell type ( 84 , 85 ). MiR-155 is basally expressed at low levels in B cells ( 85 , 86 ), T cells ( 87 ), MQs ( 88 ), and DCs ( 85 ) and stands as a perfect example of miRNA functionality throughout the immune system ( 61 ). On the other hand, MiR-155 is expressed at high levels in human cancer cells ( 61 ). S ignals activation (e.g., Ags) can rapidly increase the expression of miR-155 in leukocyte subsets that contain bone marrow DCs and MQs ( 85 - 87 ).

In a study in 2012, the encapsulated miR-155 ds-RNA duplexes (50 µg/mice) by PEI-based NPs were transmitted to C57BL/6 mice that suffered from ovarian cancer. Active miRNA can be bound to the miRNA recognition element on multiple mRNAs, resulting in simultaneously silencing multiple target genes. Therefore, it is indicated that the treatment can reverse the tolerance activity of ovarian cancer-associated DCs and promote their potential to enhance antitumor immunity through enhancement of robust Ag presentation, production of Th1 cytokines ( P <0.01), and expansion of tumor-reactive T cells. According to the mentioned study, the duplexes (double-stranded molecules of nucleic acid) were processed by tumor-associated DCs in order to generate mature miR-155 ( 61 ).

Lung cancer

Bronchogenic carcinoma is currently a worldwide highly prevalent malignant disease ( 64 ). The infiltrating leukocytes of malignant solid tumors are abundantly filled with MQs ( 63 ). The inflammatory and regulatory phenotypes of these tumor-associated MQs (TAM) are exhibited through their M2 markers ( 89 ). In fact, TAMs have been recently identified as the key mediators in cancer progression, metastasis, and resistance to therapy ( 90 , 91 ).

M2 peptide was coupled with gold NPs (AuNPs) and used to deliver anti-VEGF siRNA (0.05 mg/kg) in the sample of BALB/c mice with lung cancer. According to the findings, this hybrid approach could reduce the number of existing MQs in lung tissues when compared with the results of the treated group with solitary AuNPs and siRNA. However, there were no changes observed in the cytokines at the mRNA level along with the lack of any TLR activation. It is notable that the mentioned study succeeded in targeting an angiogenic factor (VEGF) that was produced by inflammatory MQs and lung cancer cells and modulated the recruitment of TAMs ( 63 ). Therefore, these observations proved the capability of this therapy in modulating the immune cell infiltration into inflammatory sites.

It was shown that the combination therapy of cisplatin (CDDP) and pIL-12 (plasmid encoding IL-12) lead to achieving a higher rate of antitumor activity and cancer cells apoptosis. Besides, the delivery of this formulation to C57 mice bearing lung cancer cells ((50 µg/mice) resulted in a higher IL-12 expression, increased immune effector cells (T4+, T8+, and NK cells), conversion of M2 MQs into M1 MQs, and higher levels of IFNγ and TNFα. Therefore, this formulation proved to be capable of enhancing the immune responses in the tumor microenvironment ( 65 ).

Colon cancer

Men et al . introduced the application of 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP)- modified mPEG–PCL micelles (DMP) for the delivery of plasmid encoding IL-12 gene to colon carcinoma-bearing female BALB/c mice, which increased the level of IL-12 in treated mice in comparison with the control group ( P <0.001) ( 70 ). IL-12 is an immunomodulatory cytokine with the ability to regulate the immune system through induction of immune cell proliferation ( 92 ) . According to the obtained results, the proposed method could provide a strong T cell infiltration into the tumor microenvironment and increase the rate of TNF-α secretion from immune cells such as macrophages and neutrophils ( 70 ) . Thus, it is indicated that successful expression of IL-12 in the tumor microenvironment leads to activation of immune cells and causes inducement of strong antitumor immune responses.

In conformity to another study, the delivery of cholesterol-conjugated PEGylated Polyamidoamine (PAMAM) complexed with plasmid encoding IL-12 into HT29 cells was resulted in increasing the production of IL-12 when compared with the control group ( 71 ). Doubtlessly , IL-12 initiated an immunomodulatory effect in this method through stimulation of TH1 differentiation, which augmented the antitumor activity of the immune system ( 93 ).

Considering the findings of Liu et al . the administration of folate-modified NPs containing Chemokine (C-C motif ligand) (CCL19) gene to female BALB/c mice with colorectal cancer caused a higher expression of CCL19 when compared with the control group ( P <0.05). A higher level of CCL19 can intensify the expressions of IFN-γ ( P <0.05) and TNF-α ( P <0.05), activate lymphocyte T, decrease regulatory T cells, reduce M2 macrophage (CD45⁺CD11b⁺F4/ 80⁺CD206high) polarization, and decrease the number of myeloid -derived suppressor cells (MDSCs) (Gr1⁺CD11b⁺) ( 72 ) . In conclusion , this combination therapy can induce antitumoral activities by activating immune responses.

In another study, an experiment was conducted on the delivery of protamine-liposome system ( CLPP)/IL-15 mRNA complex (20 µg protamine /20 µg liposome /10 µg IL-15 mRNA per mouse ) to female BALB/c mice bearing colon tumor and resulted in increasing level of IL-15 when compared with the control group ( P <0.05). This outcome can modulate immune responses by increasing the population of NK cells and activating T CD8⁺, as well as inducing a high level of TNF-α, IFN- γ, and IL-12 ( P <0.05) ( 73 ) . Undoubtedly, this method is capable of inhibiting tumor cell growth through immunomodulation.

Other cancer types

According to recent studies, other types of tumors can be also modulated by the performance of immunogene therapy.

Hypoxia-inducible factor 1-α (HiF-1α) is a key transcription factor in tumor development, which is also known to play a fundamental role in hypoxia-mediated apoptosis ( 94 ). In a related study, Zhao et al . considered the exertion considered the exertion of lipid-polymer hybrid NPs for the co-delivery of HiF-1α siRNA (1.33 mg/kg) and gemcitabine (4 mg/kg) in female BALB/c mice with pancreatic cancer. Their findings indicated that the levels of IL-6 and IFN-γ were decreased to normal limits when compared with the group treated with free siRNA ( 69 ), which signified the possible modulation of immune responses that consequently reduced the inflammatory effects.

In another research, lipid NPs and chemokine receptor type 2 (CCR2) siRNA (0.5 mg/kg/day siCCR2) was administered for the treatment of colon cancer in a C57BL/6 and BALB/C mice, which resulted in silencing the CCR2 miRNA in inflammatory monocytes that consequently inhibited their migration to the inflammatory site. Moreover, it can be inferred that this method modulates the immune cell inducers of inflammation due to the existing CD14⁺CD16⁻ phenotype in the inflammatory monocytes, which can cause a rise in classical MQs that exacerbate inflammation ( 95 ).

A dual-targeted immune-gene therapy system was designed based on tumor-targeted lipid-dendrimer-calcium-phosphate (TT-LDCP) NPs for the in vitro and in vivo delivery of siRNA against immune checkpoint ligand PD-L1 and plasmid DNA encoding immunostimulatory IL-2 to hepatocellular carcinoma cell (HCC). This co-delivery system resulted in significantly decreasing PD-L1 and increasing IL-2 expression in liver tumor cell lines in comparison with the performed treatment with solitary PD-L1 siRNA or IL-2 pDNA in TT-LDCP NPs. Furthermore, the observations indicated that the significant enhancement of tumoral CD8+ T cell infiltration and activation led to suppression of HCC progression ( 67 ).

According to another finding, administration of a TGF-β2 antisense oligonucleotides and a CMV-β-gal plasmid attaching to polybutyl cyanoacrylate nanoparticles (9.34 nmol/Fisher rat) combined with vaccination with F98 cells infected by New Castle Virus in rats bearing brain tumor, was lead to elevated levels of T CD25+ cells, this indicates the proliferation of activated IL-2 receptor-positive lymphocytes. The TGF-β2 levels were also significantly reduced ( 74 ). This evidence could confirm that this method may increase the survival rate.  

It was reported that the delivery of plasmid encoding IL-12 gene in combination with PAMAM into mesenchymal stem cells (MSC), which resulted in increasing the expression of IL-12 from MSCs (68); increased levels of IL-12 can cause inducement of stronger tumor responses ( 96 ).

Viral and microbial diseases

According to the literature, immunogene therapy-based nano-carriers can be effective in the treatment of infectious diseases. Table 2 demonstrates the application of nano-carriers against microbial and viral diseases in immunogene therapy.

Table 2.

Nanoparticle-based immunogene delivery used in treatment of viral and microbial diseases

Microbial disease Gene therapeutic agents Nanoparticles Functional mechanisms Ref.
Paratyphoid IL-12 Chitosan Increased IL-2,4,6
T cell activation, and recruitment to the disease site
Th2 cell proliferation
Increased APC proliferation
Increased GM-CSF and IFN-γ secretion
Stimulated B cell proliferation
Increased Ig G, A, M secretion
(32)
Pneumococcal disease PsaA Chitosan Induced IgG1/IgG2a balance
Increased IL-17A and IFN-γ expression
(97)
Hepatic disease PDL1 siRNA Lipid NP PDL1 knockdown
Increased CD8⁺ effector T cells
Increased granzyme B release from NK and CTLs
No NK effector function changes
(98)
Mycobacterium Tuberculosis HLA Chitosan Human DC maturation
Increased CD 86,83,80 expression
Increased IFN-γ secretion
(99)
RSV infection NS1 Chitosan Decreased goblet cell hyperplasia
Decreased inflammatory cell infiltration
Decreased IFN-α and β levels
Increased IL-4 and IFN-γ secretion
Increased cellular immunity
Increased IRF-1 and STAT1 expression
(100)
RSV infection RSV antigen Chitosan Increased IgG
Increased nasal IgA
Increased IFN-γ levels in lung
High CTL generation
(101)

Piglet paratyphoid is recognized as one of the most severe epidemics caused by Salmonella infection (170). Plasmid-encoding IL-2 (6 pmol/mouse) were encapsulated in chitosan NPs for being exerted for the paratyphoid treatment of female BALB/c mice. According to the findings, the applied method increased the levels of IL-2, 4, 6 in peripheral blood when compared with the controls ( P <0.05) ( 32 ). In addition, IL-2 was abled to stimulate T cell proliferation and its clonal expansion ( 102 ), while the activated T cells secret ed Granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-γ, and IL-4, which leads to APC proliferation and recruitment of more specific T cells to the infection site. On the other hand, IL-2 proved to be capable of activating the proliferation of Th2 cells which causes the releasing of IL-4, 5, 6 and stimulated B cell differentiation into mature antibody-producing plasma cells and memory cells, which also promotes the production of Ig-G, A, M ( 32 ). Therefore, these data confirmed the effectiveness of this method in significantly promoting cellular and humoral immunity.

Streptococcus pneumonia is considered a leading cause of invasive pneumonia. Pneumococcal surface antigen A (PsaA) is a lipoprotein that is highly conserved across its serotype ( 103 ). In a study, conducted in (2010), chitosan NPs encapsulated with PsaA DNA (50 µg/mouse) were used for the pneumococcal treatment of Specific-pathogen-free (SPF) female BALB/c mice. According to the results, the ratio of IgG1/IgG2a was balanced and a significant increase was induced in the expression of IL-17A and IFN-γ ( P <0.01) ( 97 ). Recent related studies reported that IL-17 is mainly released from Th17 cells and has the ability to promote host immunity through the recruitment of neutrophils to mucosal sites and provoking their bactericidal potential ( 104 , 105 ). Therefore, this method can be applied to efficiently modulate the immune system.

The liver maintains a tolerogenic environment against persistent viral infections, such as the hepatitis C virus , and often takes advantage of hepatic tolerance. PDL1-siRNA-cationic lipoid NPs (containing 0.5 mg of siRNA/kg) were exerte for the treatment of hepatic disease in C57BL/6 mice , which resulted in the occurrence of PDL-1 surface expression knockdown. This inducement increased the effector function of CD8⁺ T cells and caused the release of granzyme B (GrB) from these cells that consequently improves their virus clearance potential. Furthermore, GrB secretion from the NK cells was enhanced without causing any changes in effector function ( 98 ). In viral hepatic infections, the high expression of PDL-1 can be observed on Kupffer cells and T cells, as well as NK, DC, and liver sinusoidal cells ( 106 , 107 ).

In another study, the application of DNA plasmid encoding 8 HLA Mycobacterium tuberculosis(Mtb) epitopes, which were formulated in chitosan NPs (containing 25 µg of DNA per mouse), was considered for the treatment of HLA-A*0201/Kb female transgenic mice infected with M. tuberculosis. According to the findings, this method caused an increase in the surface levels of CD86, CD83, and CD80 which led to maturing of human DC ( 99 ). It was indicated by other studies that CD80 and CD86 are labeled as T cell costimulatory molecules, while CD83 has the potential of affect ing the capacity of DC for stimulating T cells ( 108 ). In comparison with the outcomes of the intramuscular route, the application of this formulation was observed to increase the secretion of IFN-γ ( 99 ). Therefore, the capability of the proposed treatment method in modulating and activating immune responses was confirmed.

Respiratory syncytial virus (RSV) stands as the most common cause of lower respiratory tract viral infection that can result in inducement of bronchiolitis and otitis ( 109 ). According to the literature, nonstructural (NS) proteins, as well as F and G RSV proteins, have a fundamental role in disease progression ( 110 , 111 ).

Plasmid NS1 siRNA gene complex with chitosan NPs was used for the treatment of BALB/c mice with RSV infection. In the course of the disease, NS1 was observed to suppress STAT1 ( P <0.05), IRF-1 activation ( P <0.01), and type I IFN secretion from the DCs ( P <0.05), while NS1 proteins down-regulated the signaling of the IFN system by inactivating IRF-1 and STAT1. It can be concluded that this technique can decrease the goblet cell hyperplasia of bronchi and reduce the number of infiltrating inflammatory cells throughout the interstitial regions, while increasing IRF-1, STAT1, IFN-β, and IFN-α expression and causing the release of IL-4 and IFN-γ from CD4⁺ and CD8⁺ T cells; thereby, these occurrences result in enhanc ing the cellular immunity when compared with the control groups ( 100 ). In the mentioned study, the nano-formulation was observed to modulate the host immunity.

Chitosan nanospheres containing plasmid encoding all RSV antigen cDNAs were used for the treatment of BALB/c mice with RSV infection, which also resulted in increasing the neutralization of specific IgG and nasal IgA secretion, while increasing IFN-γ secretion in the pulmonary tissues and generating powerful CTL responses when compared with the controls ( P <0.01) ( 101 ). The literature confirmed the role of RSV proteins as suitable targets for CTLs along with affirming the direct antiviral effects of IFN-γ, which are essential for stimulating the cytolytic activity of NK cells and CTLs ( 112 , 113 ). These findings signify the potential of this method for being applicable in the treatment of RSV infections.

Allergies

Allergic diseases or type I hypersensitivity refer to the illnesses that are mediated by the IgE antibody that is produced by the atopic individual after being exposed to safe environmental allergens ( 114 ). The oral route is considered as a common track of gene delivery in allergy treatments ( 41 , 115 , 116 ), which provides poor efficacy due to the acidic pH of stomach and gastrointestinal tract enzymes ( 41 ). However, specific nano-carriers , such as cationic polymers, were utilized in an attempt to overcome these obstacles. Table 3 presents data on the application of immunomodulatory nano-formulation that contained genetic materials.

Table 3.

Nanoparticle-based immunogene delivery used in allergy treatment

Allergy Gene therapeutic agents Nanoparticles Functional mechanisms Ref.
House dust mite allergy Derp 1 Chitosan Increased immune response
Increased specific IgG2a and IgA
(41)
Allergic asthma IFNγ Chitosan Decreased AHR symptoms
Decreased eosinophils in BAL fluid
Decreased mucosal metaplasia
Decreased airway cellular infiltration
Decreased IL-4,5 and IgE secretion
Increased IFN-γ
Activated STAT4 signaling pathway
(42)
House dust mite allergy Derp1 Chitosan Moderate increase in IgG2a levels
Suppressed IgE secretion
Decreased IL-4 and increased IFN-γ secretion
(115)
Peanut allergy PCMVArah2 Chitosan Decreased IgE secretion
Decreased anaphylaxis symptoms
Increased IgG2a and IgA secretion
Deceased histamine levels
(116)
asthma Thymulin Poly β-Amino Ester
(PBAE NP)
Deceased CCL11 and CXCL1 levels
Increased CCL17 level
Deceased IL-4 and IL-13 levels
Decreased TGF-β level
Increased M1 macrophages
(117)

In the work of Chew et al. plasmid DNA containing Derp1 cDNA (50 µg) was encapsulated in chitosan NPs to be orally administered to female BALB/c mice that were allergic to house dust mite (Derp1). According to their outcomes, the immune responses against Derp1, as well as the production of specific IgG2a and IgA throughout the systemic circulation , was increased in the sample animals. They also reported the ability of chitosan to facilitate immunity induction by cross ing the mucosal epithelial barrier to cause the occurrence of chitosan-DNA complex uptake by M cells and provide translocation of molecules to the underlying lymphoid organs in which APCs are located ( 41 ). Therefore, the effectiveness of this method in oral gene delivery was confirmed.

In another study, researchers administered chitosan-IFN-γ pDNA NPs (CIN) (25 µg/mouse) to allergic BALB/c mice. Their results included the observance of attenuated airway hyper-responsiveness (AHR) ( P <0.01), significant reduction in the number of eosinophils in the Bronchoalveolar lavage fluid ( P <0.01), decreased mucus cell metaplasia, cellular infiltration into the airways, and significant reduction in IL-4, 5 and IgE ( P <0.05). However, the experiment also caused an increase in the secretion of IFN-γ, while the induced immune modulation via the CIN involved the Signal transducer and activator of transcription ( STAT) 4-dependent ( 42 ) . Moreover, the uptake of CIN occurred through the means of MQs and epithelial cells, both of which play a fundamental role in asthma immunomodulation ( 118 ). CIN therapy can facilitate the induction of IFN-γ expression in epithelial cells and lead to immunomodulation. It is also notable that other Th1 cytokines (e.g., IL-12) were also involved in this process and displayed the ability to activate STAT4 ( 42 ). Based on these findings, it is inferred that mucosal therapy can decrease the allergen that is induced and established by AHR.

In another research, chitosan NPs that containing plasmid DNA and encoded house dust mite allergen Derp1 (100 µg/mouse) was conducted in allergic BALB/c mice. Their findings reported the inducement of a moderate increase in IgG2a levels ( P <0.01), along with the suppression of IgE responses ( P <0.05), a decrease in IL-4 ( P <0.01), and an increase in IFN-γ ( P <0.01) when compared with the groups treated with the allergen alone (only NPs and phosphate-buffered saline). IgG2a and IgE isotypes reflect the type of T cell responses ( 115 ) and in conformity to previous discoveries, high levels of IgG2a in conjugation with low levels of IgE can propose the shift of immune responses from Th2 toward Th1 ( 119 ). Therefore, it is indicated that this method is effective for the modulation of immune responses towards allergies.

On the same note, chitosan NPs that were composed of plasmid DNA and encoded dominant peanut allergen gene (pCMVArah2; 50 µg of DNA per mouse) for the treatment of peanut allergy in AKR/J mice. According to their results, the IgE levels and anaphylaxis symptoms were decreased, while IgG2a and IgA levels faced an increase when compared with the control group. In addition, they observed a reduction in the levels of histamine plasma. This method can activate mucosal immunity and systemic immunity ( 116 ) since the generation of mucosal immunity can protect the antigens’ entry to the systemic immune system and also lead to unresponsive behaviors toward food allergens ( 120 ). Considering these data, the proposed method could be presumed as an effective technique for the modulation of anaphylactic responses ( 116 ).

Thymic nonapeptide (thymulin or serum thymus factor) is capable of preventing the cascades of inflammatory and fibrotic responses in a mouse model of allergic asthma ( 121 ). In this regard, PBAE NPs (Poly (ethylene oxide)-modified poly (beta-amino ester), consisted of thymulin expressing plasmids (50 μl of NP that contained 50 μg of plasmid per mouse) were applied for the treatment of asthmatic female BALB/c mice ( 117 ). In comparison with the control, the results of the target group displayed a decrease in the levels of CCL11 and CXCL1, which are responsible for recruitment of eosinophil and neutrophil ( P <0.05) ( 122 ). An increase in the level of CCL17 ( P <0.05) as a fundamental factor in T regulatory transmigration ( 123 ) causes a decrease in IL-4 and IL-13 ( P <0.05) levels that involves the suppression of Th2 responses, a decrease in TGF-β level ( P <0.05), and an increase in M1 macrophages ( P <0.05) ( 117 ). Therefore, this method can effectively suppress allergic responses through immunomodulation.

Inflammatory bowel disease (IBD)

Inflammatory bowel disease (IBD) is an inflammatory condition caused by dysregulation of mucosal immunity to the luminal antigens derived from the intestinal microflora ( 124 , 125 ). IBD has two main phenotypes including Crohn’s disease (CD) and ulcerative colitis (UC). The condition of CD mostly affects the end of the small intestine and the beginning of the colon, while UC only affects the colon ( 126 ). Table 4 exhibits the application of nano-carriers against IBD in immunogene therapy.

Table 4.

Nanoparticle-based immunogene delivery used in IBD treatment

IBD Gene therapeutic agents Nanoparticles Functional mechanisms Ref.
IBD IL-10 NiMOS Decreased IL-10,12,1β and TNFα and IFN-γ secretion
Decreased colitis inflammation
Decreased MCP-1 and MIP-1α expression
(127)
IBD TNF-α siRNA NiMOS Decreased TNF-α secretion
Decreased GM-CSF, MIP-1α, and MCP-1 expression
Down-regulated IL-1β and IL-5 secretion
Regulated cell infiltration to the inflammatory site
(126)
IBD TNF-α siRNA PEI Decreased TNF-α levels (128)
IBD TNF-α siRNA PLGA Decreased TNF-α levels (129)

In a related study, the application of nanoparticles-in-microsphere oral system (NiMOS) was reported for the delivery of pDNA-encoding IL-10 (100 µg/mouse) throughout the treatment of UC in BALB/c mice. The obtained results were indicative of increased IL-10 levels ( P <0.05) and decreased TNFα , IL-12, IL-1 β , and IFN - γ . Furthermore, colitis inflammation was detected in histological examinations when compared with the control group. About IBD, the levels of Th1 cytokines (e.g., TNFα, IL-12, IL-1β, and IFN-γ) were up-regulated in order to enable IL-10 to modulate Th1-derived cytokines. Their other findings reported reduced levels of monocyte chemoattractant protein-1 (MCP-1) and Macrophage inflammatory protein-1α (MIP-1α) without mentioning any observance of direct action in between these chemokines and IL-10 (127). Therefore, it can be inferred that this transfection and oral gene delivery is an effective option for IBD treatments.

In another study, the conjugation of NiMOS NPs with TNF-α siRNA were utilized for utilized the conjugation of NiMOS NPs with TNF-α siRNA for the treatment of UC in BALB/c mice. The obtained results were indicative of a decrease in TNF-α when compared with the control group ( P <0.05) ( 126 ). According to related evidence, this cytokine has a fundamental functionality throughout modulation of inflammatory conditions in IBD ( 130 ). Furthermore, this study reported achievement of a lower expression of GM-CSF, MIP-1α, and MCP-1, which are accountable for regulat ing cell infiltration into the disease site and seem to be directly correlated with inflammation. Moreover, the solitary usage of NiMOS can cause immunomodulation effects and moderately down-regulate IL-1 β ( P <0.001), IL-5 ( P <0.001), MIP-1α, and GMCSF ( 126 ). Based on these data, this formulation can be effective and applicable for the treatment of IBD.

Relatively, TNF - α siRNA-polyethyleneimine noncomplex (100 µg/kg) was used in C57BL/6 mice with IBD. The obtained results included the inducement of a significant decrease in TNF-α levels when compared with the control group (P<0.01) (128), and also displayed the ability of nano-complex in mediating inflammation in IBD.

In another study, researchers loaded TNF-α siRNA into the galactosylated PLGA NPs and co-delivered this formulation with IL-22, which was embedded in chitosan/alginate hydrogel (20 µg/kg of siTNF and 50 µg/kg of IL-22), to the FVB male mice with UC. The essential functionality of IL-22 in mucosal healing is undeniable. Considering the induced decrease in TNF-α expression as a result of this procedure ( P <0.05), it is evident that the blockade of TNF-α expression leads to inhibition of IL-22 production ( 129 ), which signifies the ability of this co-administration to facilitate the recovery of UC through immunomodulation.

Rheumatoid arthritis (RA)

Rheumatoid arthritis (RA) is a chronic inflammatory disease that is associated with joint destruction caused by the synovial infiltration of Th1 cells ( 131 ). As an important cytokine in RA, TNF-α is involved in inflammatory conditions ( 132 ) and therefore, its targeting at the RNA level can stand as an effective therapeutic approach for the treatment of RA ( 133 , 134 ). Table 5 represents the application of immunogene therapy by the usage of various nano-formulations in RA treatment.

Table 5.

Nanoparticle-based immunogene delivery used in RA treatment

Rheumatoid arthritis Gene therapeutic agents Nanoparticles Functional mechanisms Ref.
RA Anti-TNFα dicer substrate siRNA Chitosan Decreased TNF-α and IFN-I release from MQs
Non-induction of innate immune response
Decreased inflammatory cell infiltration
(135)
RA TNF-α siRNA Liposome Decreased TNF-α secretion
Decreased IL-6 and MCP-1 locally
Increased IL-10 secretion
Induced balance between Th1 and Th2 through increased IL-4 and decreased IFN-γ
(136)

In a related study, chitosan NPs that contained the unmodified anti-TNF - α dicer substrate siRNA (DsiRNA) (2.5 µg) were transfected to DBA/mouse with RA. The results of this method included a decrease in TNF-α (P<0.01) and IFN type I (P<0.05) from the MQs when compared with the untreated groups, which led to observing the lack of innate immune responses (135). During the acute and chronic phases of arthritis, the activated peritoneal MQs release inflammatory cytokines such as IL-1,6 and TNF- α (137). In this regard, the proposed method can be considered as a novel strategy for RA treatment due to succeeding in reducing joint destruction and inflammatory cell infiltration (135).

In conformity to another study, the application of cationic liposome, containing TNF-α siRNA, in DBA/1 mice with RA caused a decrease in TNF-α release throughout the knee joint and led to the local reduction of IL-6 and MCP-1 while increasing the secretion of anti-inflammatory cytokines (e.g., IL-10) when compared with the control group. Therefore, this method was confirmed to be effective in RA treatment since TNF-α suppression can significant ly balance the induced changes between Th1 and Th2 in the draining lymph node by causing an increase in IL-4 secretion while decreasing the IFN-γ levels ( 136 ).

Conclusion and future perspective

Immunogene therapy is potentially considered an effective approach for the treatment of various complex disorders due to being capable of targeting cells more specifically and providing control over the expression of therapeutic genes. On the other hand, the local and single-dose application of most gene therapies results in the exertion of high therapeutic doses with a low risk of systemic side effects. In this case, the U.S. Food and Drug Administration (FDA) has reviewed and confirmed all comments about clinical trial of immunogene therapy in combination with AstraZeneca’s Tagrisso ®  in patients with late-stage non-small cell lung cancer (NSCLC) whose disease progressed after treatment with Tagrisso in Genprex, Inc. (138). Various gene delivery systems have been developed to improve the delivery efficiency of genetic materials. Most of the applied carriers in human clinical trial phases for immunogene therapy include different types of viral vectors. The combination of AdCD40L (an adenovirus carrying CD40 ligand gene, a key activator of adaptive immunity) in phase I/II study (NCT01455259) with cyclophosphamide chemotherapy was exerted for patients with metastatic malignant melanoma (139). In addition, clinical-grade chimeric antigen receptor (CAR) T cells were engineered by viral vectors and established to mediate tumor rejection. There are important limitations due to uncontrolled responses as a consequence of constitutive expression of the CAR molecules on the surface of T cells (140). So, more standard manufacturing processes are required for immunogene T-cell therapies (141).

Viral vectors were also exerted for the development of clinical trials with critical inflammatory cytokine gene therapies for cancer treatments.

There are reports on the application of nano-systems in clinical or preclinical studies for the delivery of therapeutic genes. Nano-carriers offer prominent advantages such as safety, high targeting potential, low toxicity, cost-efficiency, and easy preparation. However, several factors can possibly affect the immune responses of NPs such as the composition, size, surface chemistry, transfection efficiency, cytotoxicity, and exposure route of carriers.

Most of the immunogene therapy studies by the usage of NPs were conducted with animal models , which provide a strong preclinical basis for human clinical approaches. Therefore, further clinical trials are required in order to clarify the therapeutic efficacy and possible toxic effects of exerted nanosystem s in immunogene therapy.

Authors’ Contributions

MH Supervised the research; SSH and FO Draft manuscript preparation; SSH and EH Critically revised the paper. SSH, FO, EH and MH Read and approved the the final version to be published.

Conflicts of Interest

The authors declare that there are no relevant financial or non-financial competing interests to report.

Acknowledgment

The authors are thankful to Mashhad University of Medical Science (MUMS). No funding to declare.

References

  • 1.Pusuluri A, Wu D, Mitragotri S. Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release. 2019;305:130–154. doi: 10.1016/j.jconrel.2019.04.020. [DOI] [PubMed] [Google Scholar]
  • 2.Smith AJ, Oertle J, Prato D. Immunotherapy in cancer treatment. Open J Med Microbiol. 2014;4:178. [Google Scholar]
  • 3.Oroojalian F, Charbgoo F, Hashemi M, Amani A, Yazdian-Robati R, Mokhtarzadeh A, et al. Recent advances in nanotechnology-based drug delivery systems for the kidney. J Control Release . 2020;321:442–462. doi: 10.1016/j.jconrel.2020.02.027. [DOI] [PubMed] [Google Scholar]
  • 4.Naldini L. Gene Therapy Returns to Centre Stage. 2015;526:351–360. doi: 10.1038/nature15818. [DOI] [PubMed] [Google Scholar]
  • 5.Zhou Y, Zhang C, Liang W. Development of RNAi technology for targeted therapy—a track of siRNA based agents to RNAi therapeutics. J Control Release. 2014;193:270–281. doi: 10.1016/j.jconrel.2014.04.044. [DOI] [PubMed] [Google Scholar]
  • 6.Zhang X, Godbey W. Viral vectors for gene delivery in tissue engineering. Adv Drug Deliv Rev. 2006;58:515–534. doi: 10.1016/j.addr.2006.03.006. [DOI] [PubMed] [Google Scholar]
  • 7.Cherkassky L, Grosser R, Adusumilli PS. Regional Gene Therapy for Cancer. J Cancer Ther. 2020:55–71. [Google Scholar]
  • 8.Heilbronn R, Weger S. Viral vectors for gene transfer: current status of gene therapeutics. Drug Deliv. 2010:143–170. doi: 10.1007/978-3-642-00477-3_5. [DOI] [PubMed] [Google Scholar]
  • 9.Fischer A, Hacein-Bey-Abina S. Gene therapy for severe combined immunodeficiencies and beyond. Exp Med. 2020;217:e20190607. doi: 10.1084/jem.20190607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ugwu GC, Egbuji JVI, Okanya LC, Omeje JN, Eyo JE. Gene therapy, physiological applications, problems and prospects-a review. Anim Res Int. 2019;16:3367–3392. [Google Scholar]
  • 11.Li C, Samulski RJ. Engineering adeno-associated virus vectors for gene therapy. Nat Rev Genet. 2020;21:255–272. doi: 10.1038/s41576-019-0205-4. [DOI] [PubMed] [Google Scholar]
  • 12.Shirley JL, de Jong YP, Terhorst C, Herzog RW. Immune Immune responses to viral gene therapy vectors. Mol Ther. 2020;28:709–722. doi: 10.1016/j.ymthe.2020.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Deng W, Fu M, Cao Y, Cao X, Wang M, Yang Y, et al. Angelica sinensis polysaccharide nanoparticles as novel non-viral carriers for gene delivery to mesenchymal stem cells. Nanomed: Nanotechnol Biol Med. 2013;9:1181–1191. doi: 10.1016/j.nano.2013.05.008. [DOI] [PubMed] [Google Scholar]
  • 14.Kommareddy S, Amiji M. Poly(ethylene glycol)–modified thiolated gelatin nanoparticles for glutathione-responsive intracellular DNA delivery. Nanomed: Nanotechnol Biol Med. 2007;3:32–42. doi: 10.1016/j.nano.2006.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Pathak A, Patnaik S, Gupta KC. Recent trends in non-viral vector-mediated gene delivery. Biotechnol J. 2009;4:1559–1572. doi: 10.1002/biot.200900161. [DOI] [PubMed] [Google Scholar]
  • 16.Chuan D, Jin T, Fan R, Zhou L, Guo G. Chitosan for gene delivery: Methods for improvement and applications. Adv Colloid Interface Sci. 2019;268:25–38. doi: 10.1016/j.cis.2019.03.007. [DOI] [PubMed] [Google Scholar]
  • 17.Pridgen EM, Langer R, Farokhzad OC. Biodegradable, polymeric nanoparticle delivery systems for cancer therapy. Nanomedicine. 2007;2:669–680. doi: 10.2217/17435889.2.5.669. [DOI] [PubMed] [Google Scholar]
  • 18.Ozpolat B, Sood A, Lopez‐Berestein G. Nanomedicine based approaches for the delivery of siRNA in cancer. J Intern Med. 2010;267:44–53. doi: 10.1111/j.1365-2796.2009.02191.x. [DOI] [PubMed] [Google Scholar]
  • 19.Bayat S, Rabbani Zabihi A, Amel Farzad S, Movaffagh J, Hashemi E, Arabzadeh S, Hahsemi M. Evaluation of debridement effects of bromelain-loaded sodium alginate nanoparticles incorporated into chitosan hydrogel in animal models. Iran J Basic Med Sci. 2021;24:1404–1412. doi: 10.22038/IJBMS.2021.58798.13060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rezaei M, Hosseini SN, Khavari-Nejad RA, Najafi F, Mahdavi M. Fast antibody responses by immuno-targeting and nanotechnology strategies versus HBsAg vaccine. Iran J Basic Med Sci. 2021;24:545–555. doi: 10.22038/IJBMS.2021.52715.11896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fontana F, Liu D, Hirvonen J, Santos HA. Delivery of therapeutics with nanoparticles: what’s new in cancer immunotherapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017;9:e1421. doi: 10.1002/wnan.1421. [DOI] [PubMed] [Google Scholar]
  • 22.Rashidi A, Omidi M, Choolaei M, Nazarzadeh M, Yadegari A, Haghierosadat F, et al. , editors. Electromechanical properties of vertically aligned carbon nanotube. Adv Mat Res. 2013:332–336. [Google Scholar]
  • 23.Singh MS, Bhaskar S. Nanocarrier-based immunotherapy in cancer management and research. ImmunoTargets and Therapy. 2014;3:121–134. doi: 10.2147/ITT.S62471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Klinman DM. Immunotherapeutic uses of CpG oligodeoxynucleotides. Nat Rev Immunol. 2004;4 doi: 10.1038/nri1329. [DOI] [PubMed] [Google Scholar]
  • 25.Fu J, Cai J, Ling G, Li A, Zhao J, Guo X, et al. Cationic polymers for enhancing CpG oligodeoxynucleotides-mediated cancer immunotherapy. Eur. 2019;113:115–132. [Google Scholar]
  • 26.De Jong S, Chikh G, Sekirov L, Raney S, Semple S, Klimuk S, et al. Encapsulation in liposomal nanoparticles enhances the immunostimulatory, adjuvant and anti-tumor activity of subcutaneously administered CpG ODN. Cancer Immunol Immunother. 2007;56:1251–1264. doi: 10.1007/s00262-006-0276-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gao Q, Hong J, Xiao X, Cao H, Yuan R, Liu Z, et al. T cell epitope of arginine kinase with CpG co-encapsulated nanoparticles attenuates a shrimp allergen-induced Th2-bias food allergy. Biosci Biotechnol Biochem. 2019;84:804–814. doi: 10.1080/09168451.2019.1699395. [DOI] [PubMed] [Google Scholar]
  • 28.Gunawardana T, Ahmed KA, Goonewardene K, Popowich S, Kurukulasuriya S, Karunarathana R, et al. CpG-ODN induces a dose-dependent enrichment of immunological niches in the spleen and lungs of neonatal chicks that correlates with the protectivei mmunity against Escherichia coli. J Immunol Res . 2020;2020:2704–728. doi: 10.1155/2020/2704728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Jahanban‐Esfahlan R, Seidi K, Majidinia M, Karimian A, Yousefi B, Nabavi SM, et al. Toll‐like receptors as novel therapeutic targets for herpes simplex virus infection. Rev Med Virol. 2019;29:e2048. doi: 10.1002/rmv.2048. [DOI] [PubMed] [Google Scholar]
  • 30.Krieg AM. CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. 2002;20:709–760. doi: 10.1146/annurev.immunol.20.100301.064842. [DOI] [PubMed] [Google Scholar]
  • 31.Mavi SA, Modarressi MH, Mohebali M, Shojaee S, Zeraati H. Assessment of the immunogenicity and protective efficiency of a novel dual-promoter DNA vaccine, harboring SAG1 and GRA7 genes, from RH strain of Toxoplasma gondii in BALB/c mice. Infect Drug Resist. 2019;12:2519. doi: 10.2147/IDR.S209270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yang Y, Chen J, Li H, Wang Y, Xie Z, Wu M, et al. Porcine interleukin-2 gene encapsulated in chitosan nanoparticles enhances immune response of mice to piglet paratyphoid vaccine. Comp Immunol Microbiol Infect Dis. 2007;30:19–32. doi: 10.1016/j.cimid.2006.09.006. [DOI] [PubMed] [Google Scholar]
  • 33.Bourquin C, Anz D, Zwiorek K, Lanz AL, Fuchs S, Weigel S, et al. Targeting CpG oligonucleotides to the lymph node by nanoparticles elicits efficient antitumoral immunity. J Immunol. 2008;181:2990–2998. doi: 10.4049/jimmunol.181.5.2990. [DOI] [PubMed] [Google Scholar]
  • 34.Kawakami S, Higuchi Y, Hashida M. Nonviral approaches for targeted delivery of plasmid DNA and oligonucleotide. J Pharm Sci. 2008;97:726–745. doi: 10.1002/jps.21024. [DOI] [PubMed] [Google Scholar]
  • 35.Xu Y, Yuen P-W, Lam J. Intranasal DNA vaccine for protection against respiratory infectious diseases: the delivery perspectives. Pharmaceutics. 2014;6:378–415. doi: 10.3390/pharmaceutics6030378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pishdadian A, Varasteh A, Gholamin M, Roozbeh Nasiraie L, Hosseinpour M, Moghadam M, Sankian M. Lung‐derived innate cytokines: New epigenetic targets of allergen‐specific sublingual immunotherapy. Iran J Basic Med Sci. 2016;19:64–71. [PMC free article] [PubMed] [Google Scholar]
  • 37.Havenar-Daughton C, Carnathan DG, Boopathy AV, Upadhyay AA, Murrell B, Reiss SM, et al. Rapid germinal center and antibody responses in non-human primates after a single nanoparticle vaccinei mmunization. Cell Rep. 2019;29:1756–1766. doi: 10.1016/j.celrep.2019.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liu MA. A comparison of plasmid DNA and mrna as vaccine technologies. Vaccines. 2019;7:37–57. doi: 10.3390/vaccines7020037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gupta J, Pathak M, Misra S, Misra-Bhattacharya S. CpG enhances the immunogenicity of heterologous DNA-prime/protein-boost vaccination with the heavy chain myosin of Brugia malayi in BALB/c mice. Parasitol Res. 2019;118:1943–1952. doi: 10.1007/s00436-019-06318-6. [DOI] [PubMed] [Google Scholar]
  • 40.Wu H-M, Xie Q-M, Zhao C-C, Xu J, Fan X-Y, Fei G-H. Melatonin biosynthesis restored by CpG oligodeoxynucleotides attenuates allergic airway inflammation via regulating NLRP3 inflammasome. Life Sci. 2019;239:117067. doi: 10.1016/j.lfs.2019.117067. [DOI] [PubMed] [Google Scholar]
  • 41.Chew JL, Wolfowicz CB, Mao H-Q, Leong KW, Chua KY. Chitosan nanoparticles containing plasmid DNA encoding house dust mite allergen, Der p 1 for oral vaccination in mice. Vaccine. 2003;21:2720–2729. doi: 10.1016/s0264-410x(03)00228-7. [DOI] [PubMed] [Google Scholar]
  • 42.Kumar M, Kong X, Behera AK, Hellermann GR, Lockey RF, Mohapatra SS. Chitosan IFN-γ-pDNA nanoparticle (CIN) therapy for allergic asthma. Genet Vaccines Ther. 2003;1:3–13. doi: 10.1186/1479-0556-1-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hou Z, Han Q, Zhang C, Zhang J. Perspectives on RNA Interference in Immunopharmacology and Immunotherapy. J RNAi Gene Silenc . 2016:285–309. [Google Scholar]
  • 44.Kawasaki H, Taira K. Induction of DNA methylation and gene silencing by short interfering RNAs in human cells. Nature. 2004;431 doi: 10.1038/nature02889. [DOI] [PubMed] [Google Scholar]
  • 45.Morris KV, Chan SW-L, Jacobsen SE, Looney DJ. Small interfering RNA-induced transcriptional gene silencing in human cells. Science. 2004;305:1289–1292. doi: 10.1126/science.1101372. [DOI] [PubMed] [Google Scholar]
  • 46.Bowers K. RNA interference-mediated inhibition of ESCRT in mammalian cells. The ESCRT Complexes. 2019;1998:305–318. doi: 10.1007/978-1-4939-9492-2_22. [DOI] [PubMed] [Google Scholar]
  • 47.Lichtenberg SS, Tsyusko OV, Palli SR, Unrine JM. Uptake and bioactivity of chitosan/double-stranded RNA polyplex nanoparticles in Caenorhabditis elegans. Environ Sci Technol. 2019;53:3832–3840. doi: 10.1021/acs.est.8b06560. [DOI] [PubMed] [Google Scholar]
  • 48.Mansoori B, Mohammadi A, Shir Jang S, Baradaran B. Mechanisms of immune system activation in mammalians by small interfering RNA (siRNA) Artif Cells Nanomed Biotechnol. 2016;44:1589–1596. doi: 10.3109/21691401.2015.1102738. [DOI] [PubMed] [Google Scholar]
  • 49.Wittrup A, Lieberman J. Knocking down disease: a progress report on siRNA therapeutics. Nat Rev Genet. 2015;16:543–552. doi: 10.1038/nrg3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Cubillos-Ruiz JR, Engle X, Scarlett UK, Martinez D, Barber A, Elgueta R, et al. Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J Clin Invest. 2009;119:2231–2244. doi: 10.1172/JCI37716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Teo PY, Yang C, Whilding LM, Parente-Pereira AC, Maher J, George AJ, et al. Ovarian cancer immunotherapy using PD-L1 siRNA targeted delivery from folic acid-functionalized polyethylenimine: strategies to enhance T cell killing. Adv Healthc Mater. 2015;4:1180–1189. doi: 10.1002/adhm.201500089. [DOI] [PubMed] [Google Scholar]
  • 52.Jerome V, Graser A, Muller R, Kontermann RE, Konur A. Cytotoxic T lymphocytes responding to low dose TRP2 antigen are induced against B16 melanoma by liposome-encapsulated TRP2 peptide and CpG DNA adjuvant. J Immunother. 2006;29:294–305. doi: 10.1097/01.cji.0000199195.97845.18. [DOI] [PubMed] [Google Scholar]
  • 53.Xu Z, Ramishetti S, Tseng Y-C, Guo S, Wang Y, Huang L. Multifunctional nanoparticles co-delivering Trp2 peptide and CpG adjuvant induce potent cytotoxic T-lymphocyte response against melanoma and its lung metastasis. J Control Release . 2013;172:259–265. doi: 10.1016/j.jconrel.2013.08.021. [DOI] [PubMed] [Google Scholar]
  • 54.Ito A, Matsuoka F, Honda H, Kobayashi T. Heat shock protein 70 gene therapy combined with hyperthermia using magnetic nanoparticles. Cancer Gene Ther. 2003;10:918–925. doi: 10.1038/sj.cgt.7700648. [DOI] [PubMed] [Google Scholar]
  • 55.Oberli MA, Reichmuth AM, Dorkin JR, Mitchell MJ, Fenton OS, Jaklenec A, et al. Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy. Nano Lett. 2017;17:1326–1335. doi: 10.1021/acs.nanolett.6b03329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yan S, Rolfe BE, Zhang B, Mohammed YH, Gu W, Xu ZP. Polarized immune responses modulated by layered double hydroxides nanoparticle conjugated with CpG. Biomaterials. 2014;35:9508–9516. doi: 10.1016/j.biomaterials.2014.07.055. [DOI] [PubMed] [Google Scholar]
  • 57.Hobo W, Novobrantseva TI, Fredrix H, Wong J, Milstein S, Epstein-Barash H, et al. Improving dendritic cell vaccine immunogenicity by silencing PD-1 ligands using siRNA-lipid nanoparticles combined with antigen mRNA electroporation. Cancer Immunol Immunother. 2013;62:285–297. doi: 10.1007/s00262-012-1334-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chellat F, Grandjean-Laquerriere A, Le Naour R, Fernandes J, Yahia L, Guenounou M, et al. Metalloproteinase and cytokine production by THP-1 macrophages following exposure to chitosan-DNA nanoparticles. Biomaterials. 2005;26:961–970. doi: 10.1016/j.biomaterials.2004.04.006. [DOI] [PubMed] [Google Scholar]
  • 59.Kim TH, Nah JW, Cho MH, Park TG, Cho CS. Receptor-mediated gene delivery into antigen presenting cells using mannosylated chitosan/DNA nanoparticles. J Nanosci Nanotechnol. 2006;6:2796–2803. doi: 10.1166/jnn.2006.434. [DOI] [PubMed] [Google Scholar]
  • 60.Li Y, Su Z, Zhao W, Zhang X, Momin N, Zhang C, et al. Multifunctional oncolytic nanoparticles deliver self-replicating IL-12 RNA to eliminate established tumors and prime systemic immunity. Nat Rev Cancer. 2020;1:882–893. doi: 10.1038/s43018-020-0095-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cubillos-Ruiz JR, Baird JR, Tesone AJ, Rutkowski MR, Scarlett UK, Camposeco-Jacobs AL, et al. Reprogramming tumor-associated dendritic cells in vivo using miRNA mimetics triggers protective immunity against ovarian cancer. Cancer Res. 2012;72:1683–1693. doi: 10.1158/0008-5472.CAN-11-3160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Suzuki R, Namai E, Oda Y, Nishiie N, Otake S, Koshima R, et al. Cancer gene therapy by IL-12 gene delivery using liposomal bubbles and tumoral ultrasound exposure. J Control Release. 2010;142:245–250. doi: 10.1016/j.jconrel.2009.10.027. [DOI] [PubMed] [Google Scholar]
  • 63.Conde J, Bao C, Tan Y, Cui D, Edelman ER, Azevedo HS, et al. Dual targeted immunotherapy via in vivo delivery of biohybrid RNAi-peptide nanoparticles to tumor-associated macrophages and cancer cells. Adv Funct Mater. 2015;25:4183–4194. doi: 10.1002/adfm.201501283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Rodrigo-Garzon M, Berraondo P, Ochoa L, Zulueta JJ, Gonzalez-Aseguinolaza G. Antitumoral efficacy of DNA nanoparticles in murine models of lung cancer and pulmonary metastasis. Cancer Gene Ther. 2010;17:20–27. doi: 10.1038/cgt.2009.45. [DOI] [PubMed] [Google Scholar]
  • 65.Sun Y, Yang J, Yang T, Li Y, Zhu R, Hou Y, et al. Co-delivery of IL-12 cytokine gene and cisplatin prodrug by a polymetformin-conjugated nanosystem for lung cancer chemo-gene treatment through chemotherapy sensitization and tumor microenvironment modulation. Acta Biomater. 2021;128:447–461. doi: 10.1016/j.actbio.2021.04.034. [DOI] [PubMed] [Google Scholar]
  • 66.Diez S, Navarro G, de ICT. In vivo targeted gene delivery by cationic nanoparticles for treatment of hepatocellular carcinoma. J Gene Med. 2009;11:38–45. doi: 10.1002/jgm.1273. [DOI] [PubMed] [Google Scholar]
  • 67.Huang K-W, Hsu F-F, Qiu JT, Chern G-J, Lee Y-A, Chang C-C, et al. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci Adv. 2020;6 doi: 10.1126/sciadv.aax5032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Azimifar MA, Salmasi Z, Doosti A, Babaei N, Hashemi M. Evaluation of the efficiency of modified PAMAM dendrimer with low molecular weight protamine peptide to deliver IL‐12 plasmid into stem cells as cancer therapy vehicles. Biotechnol Prog. 2021;37:e3175. doi: 10.1002/btpr.3175. [DOI] [PubMed] [Google Scholar]
  • 69.Zhao X, Li F, Li Y, Wang H, Ren H, Chen J, et al. Co-delivery of HIF1alpha siRNA and gemcitabine via biocompatible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials. 2015;46:13–25. doi: 10.1016/j.biomaterials.2014.12.028. [DOI] [PubMed] [Google Scholar]
  • 70.Men K, Huang R, Zhang X, Zhang R, Zhang Y, He M, et al. Local and systemic delivery of interleukin-12 gene by cationic micelles for cancer immunogene therapy. J Biomed Nanotech. 2018;14:1719–1730. doi: 10.1166/jbn.2018.2593. [DOI] [PubMed] [Google Scholar]
  • 71.Pishavar E, Oroojalian F, Ramezani M, Hashemi M. Cholesterol‐conjugated PEGylated PAMAM as an efficient nanocarrier for plasmid encoding interleukin‐12 immunogene delivery toward colon cancer cells. Biotechnol Prog. 2020;36:e2952. doi: 10.1002/btpr.2952. [DOI] [PubMed] [Google Scholar]
  • 72.Liu X, Wang B, Li Y, Hu Y, Li X, Yu T, et al. Powerful anticolon tumor effect of targeted gene immunotherapy using folate-modified nanoparticle delivery of CCL19 to activate the immune system. ACS Cent Sci. 2019;5:277–289. doi: 10.1021/acscentsci.8b00688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lei S, Zhang X, Men K, Gao Y, Yang X, Wu S, et al. Efficient colorectal cancer gene therapy with IL-15 mRNA nanoformulation. Mol Pharm. 2020;17:3378–3391. doi: 10.1021/acs.molpharmaceut.0c00451. [DOI] [PubMed] [Google Scholar]
  • 74.Schneider T, Becker A, Ringe K, Reinhold A, Firsching R, Sabel BA. Brain tumor therapy by combined vaccination and antisense oligonucleotide delivery with nanoparticles. J Neuroimmunol. 2008;195:21–27. doi: 10.1016/j.jneuroim.2007.12.005. [DOI] [PubMed] [Google Scholar]
  • 75.Zoller M, Strubel A, Hammerling G, Andrighetto G, Raz A, Ben-Ze’ev A. Interferon-gamma treatment of B16 melanoma cells: opposing effects for non-adaptive and adaptive immune defense and its reflection by metastatic spread. Int J Cancer. 1988;41:256–266. doi: 10.1002/ijc.2910410217. [DOI] [PubMed] [Google Scholar]
  • 76.Melief CJ. Tumor eradication by adoptive transfer of cytotoxic T lymphocytes. Adv Cancer Res. 1992;58:143–175. doi: 10.1016/s0065-230x(08)60294-8. [DOI] [PubMed] [Google Scholar]
  • 77.Melief CJ, Van Der Burg SH, Toes RE, Ossendorp F, Offringa R. Effective therapeutic anticancer vaccines based on precision guiding of cytolytic T lymphocytes. Immunol Rev. 2002;188:177–182. doi: 10.1034/j.1600-065x.2002.18816.x. [DOI] [PubMed] [Google Scholar]
  • 78.Hulina-Tomašković A, Somborac-Bačura A, Rajković MG, Bosnar M, Samaržija M, Rumora L. Effects of extracellular Hsp70 and cigarette smoke on differentiated THP-1 cells and human monocyte-derived macrophages. Mol Immunol. 2019;111:53–63. doi: 10.1016/j.molimm.2019.04.002. [DOI] [PubMed] [Google Scholar]
  • 79.Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006;13:816–825. doi: 10.1038/sj.cdd.4401850. [DOI] [PubMed] [Google Scholar]
  • 80.Abiko K, Mandai M, Hamanishi J, Yoshioka Y, Matsumura N, Baba T, et al. PD-L1 on tumor cells is induced in ascites and promotes peritoneal dissemination of ovarian cancer through CTL dysfunction. Clin Cancer Res. 2013;19:1363–1374. doi: 10.1158/1078-0432.CCR-12-2199. [DOI] [PubMed] [Google Scholar]
  • 81.Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res. 2013;73:6900–6912. doi: 10.1158/0008-5472.CAN-13-1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:3360–3365. doi: 10.1073/pnas.0611533104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Maine CJ, Aziz NH, Chatterjee J, Hayford C, Brewig N, Whilding L, et al. Programmed death ligand-1 over-expression correlates with malignancy and contributes to immune regulation in ovarian cancer. Cancer Immunol Immunother. 2014;63:215–224. doi: 10.1007/s00262-013-1503-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G, et al. MiR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–161. doi: 10.1016/j.cell.2007.03.008. [DOI] [PubMed] [Google Scholar]
  • 85.Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR, et al. Requirement of bic/microRNA-155 for normal immune function. Science. 2007;316:608–611. doi: 10.1126/science.1139253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Thai TH, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y, et al. Regulation of the germinal center response by microRNA-155. Science. 2007;316:604–608. doi: 10.1126/science.1141229. [DOI] [PubMed] [Google Scholar]
  • 87.O’Connell RM, Rao DS, Chaudhuri AA, Boldin MP, Taganov KD, Nicoll J, et al. Sustained expression of microRNA-155 in hematopoietic stem cells causes a myeloproliferative disorder. J Exp Med. 2008;205:5855–5894. doi: 10.1084/jem.20072108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci U S A. 2007;104:1604–1609. doi: 10.1073/pnas.0610731104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
  • 90.Cook J, Hagemann T. Tumour-associated macrophages and cancer. Curr Opin Pharmacol. 2013;13:595–601. doi: 10.1016/j.coph.2013.05.017. [DOI] [PubMed] [Google Scholar]
  • 91.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 92.Vignali DA, Kuchroo VK. IL-12 family cytokines: immunological playmakers. Nat Immunol. 2012;13:722–728. doi: 10.1038/ni.2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Roupakia E, Markopoulos GS, Kolettas E. IL-12-mediated transcriptional regulation of matrix metalloproteinases. Biosci Rep. 2018;38:BSR20171420. doi: 10.1042/BSR20171420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Luongo M, Brigida AL, Mascolo L, Gaudino G. Possible therapeutic effects of ozone mixture on hypoxia in tumor development. Anticancer Res. 2017;37:425–435. doi: 10.21873/anticanres.11334. [DOI] [PubMed] [Google Scholar]
  • 95.Leuschner F, Dutta P, Gorbatov R, Novobrantseva TI, Donahoe JS, Courties G, et al. Therapeutic siRNA silencing in inflammatory monocytes in mice. Nat Biotechnol. 2011;29:1005–1010. doi: 10.1038/nbt.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hu Y-L, Miao P-H, Huang B, Zhang T-Y, Hu Z-J, Tabata Y, et al. Reversal of tumor growth by gene modification of mesenchymal stem cells using spermine-pullulan/DNA nanoparticles. J Biomed Nanotech. 2014;10:299–308. doi: 10.1166/jbn.2014.1712. [DOI] [PubMed] [Google Scholar]
  • 97.Xu J, Dai W, Wang Z, Chen B, Li Z, Fan X. Intranasal vaccination with chitosan-DNA nanoparticles expressing pneumococcal surface antigen a protects mice against nasopharyngeal colonization by Streptococcus pneumoniae. Clin Vaccine Immunol. 2011;18:75–81. doi: 10.1128/CVI.00263-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Dolina JS, Sung SS, Novobrantseva TI, Nguyen TM, Hahn YS. Lipidoid nanoparticles containing PD-L1 siRNA delivered in vivo enter kupffer cells and enhance NK and CD8(+) T Cell-mediated hepatic antiviral immunity. Mol Ther Nucleic Acids. 2013;2:e72. doi: 10.1038/mtna.2012.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bivas-Benita M, van Meijgaarden KE, Franken KL, Junginger HE, Borchard G, Ottenhoff TH, et al. Pulmonary delivery of chitosan-DNA nanoparticles enhances the immunogenicity of a DNA vaccine encoding HLA-A*0201-restricted T-cell epitopes of Mycobacterium tuberculosis. Vaccine. 2004;22:1609–1615. doi: 10.1016/j.vaccine.2003.09.044. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang W, Yang H, Kong X, Mohapatra S, San Juan-Vergara H, Hellermann G, et al. Inhibition of respiratory syncytial virus infection with intranasal siRNA nanoparticles targeting the viral NS1 gene. Nat Med. 2005;11:56–62. doi: 10.1038/nm1174. [DOI] [PubMed] [Google Scholar]
  • 101.Kumar M, Behera AK, Lockey RF, Zhang J, Bhullar G, de la Cruz CP, et al. Intranasal gene transfer by chitosan–DNA nanospheres protects BALB/c mice against acute respiratory syncytial virus infection. Hum Gene Ther. 2002;13:1415–1425. doi: 10.1089/10430340260185058. [DOI] [PubMed] [Google Scholar]
  • 102.Spolski R, Li P, Leonard WJ. Biology and regulation of IL-2: from molecular mechanisms to human therapy. Nat Rev Immunol. 2018;18:648–659. doi: 10.1038/s41577-018-0046-y. [DOI] [PubMed] [Google Scholar]
  • 103.Morrison KE, Lake D, Crook J, Carlone GM, Ades E, Facklam R, et al. Confirmation of psaA in all 90 serotypes of Streptococcus pneumoniae by PCR and potential of this assay for identification and diagnosis. J Clin Microbiol. 2000;38:434–437. doi: 10.1128/jcm.38.1.434-437.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Curtis MM, Way SS. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology. 2009;126:177–185. doi: 10.1111/j.1365-2567.2008.03017.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhang Z, Clarke TB, Weiser JN. Cellular effectors mediating Th17-dependent clearance of pneumococcal colonization in mice. J Clin Invest. 2009;119:1899–1909. doi: 10.1172/JCI36731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med. 2003;198:39–50. doi: 10.1084/jem.20022235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Loke P, Allison JP. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc Natl Acad Sci U S A. 2003;100:5336–5341. doi: 10.1073/pnas.0931259100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lechmann M, Krooshoop DJ, Dudziak D, Kremmer E, Kuhnt C, Figdor CG, et al. The extracellular domain of CD83 inhibits dendritic cell-mediated T cell stimulation and binds to a ligand on dendritic cells. J Exp Med. 2001;194:1813–1821. doi: 10.1084/jem.194.12.1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Abreo A, Wu P, Donovan BM, Ding T, Gebretsadik T, Huang X, et al. Infant respiratory syncytial virus bronchiolitis and subsequent risk of pneumonia, otitis media, and antibiotic utilization. Clin Infect Dis. 2019;71:211–214. doi: 10.1093/cid/ciz1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Li X, Sambhara S, Li CX, Ettorre L, Switzer I, Cates G, et al. Plasmid DNA encoding the respiratory syncytial virus G protein is a promising vaccine candidate. Virology. 2000;269:54–65. doi: 10.1006/viro.2000.0186. [DOI] [PubMed] [Google Scholar]
  • 111.Murphy BR, Collins PL. Live-attenuated virus vaccines for respiratory syncytial and parainfluenza viruses: applications of reverse genetics. J Clin Investig. 2002;110:21–27. doi: 10.1172/JCI16077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Schwarz B, Morabito KM, Ruckwardt TJ, Patterson DP, Avera J, Miettinen HM, et al. Viruslike particles encapsidating respiratory syncytial virus M and M2 proteins induce robust T cell responses. ACS Biomater Sci Eng. 2016;2:2324–2332. doi: 10.1021/acsbiomaterials.6b00532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Song X, Xiao H-T, Liao C-H, Li L, Kang Q-R, Jiang Y-C, et al. Natural products: the master regulators of antiviral cytokines. Curr Org Chem. 2017;21:1805–1823. [Google Scholar]
  • 114.Kay AB. Allergy and allergic diseases. NEJM. 2001;344:30–37. doi: 10.1056/NEJM200101043440106. [DOI] [PubMed] [Google Scholar]
  • 115.Li G, Liu Z, Liao B, Zhong N. Induction of Th1-type immune response by chitosan nanoparticles containing plasmid DNA encoding house dust mite allergen Der p 2 for oral vaccination in mice. Cell Mol Immunol. 2009;6:45–50. doi: 10.1038/cmi.2009.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Roy K, Mao H-Q, Huang S-K, Leong KW. Oral gene delivery with chitosan–DNA nanoparticles generates immunologic protection in a murine model of peanut allergy. Nat Med. 1999;5:387–391. doi: 10.1038/7385. [DOI] [PubMed] [Google Scholar]
  • 117.Da Silva AL, de Oliveira GP, Kim N, Cruz FF, Kitoko JZ, Blanco NG, et al. Nanoparticle-based thymulin gene therapy therapeutically reverses key pathology of experimental allergic asthma. Sci Adv. 2020;6:eaay7973. doi: 10.1126/sciadv.aay7973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tang C, Inman MD, van Rooijen N, Yang P, Shen H, Matsumoto K, et al. Th type 1-stimulating activity of lung macrophages inhibits Th2-mediated allergic airway inflammation by an IFN-γ-dependent mechanism. J Immunol. 2001;166:1471–1481. doi: 10.4049/jimmunol.166.3.1471. [DOI] [PubMed] [Google Scholar]
  • 119.Hajavi J, Hashemi M, Sankian M. Evaluation of size and dose effects of rChe a 3 allergen loaded PLGA nanoparticles on modulation of Th2 immune responses by sublingual immunotherapy in mouse model of rhinitis allergic. Int J Pharm. 2019;563:282–292. doi: 10.1016/j.ijpharm.2019.03.040. [DOI] [PubMed] [Google Scholar]
  • 120.Lehrer RI, Ganz T. Biochemistry and function of monocytes and macrophages. Hematol. 2001;6:865–869. [Google Scholar]
  • 121.Da Silva AL, Martini SV, Abreu SC, Samary CdS, Diaz BL, Fernezlian S, et al. DNA nanoparticle-mediated thymulin gene therapy prevents airway remodeling in experimental allergic asthma. J Control Release. 2014;180:125–133. doi: 10.1016/j.jconrel.2014.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Isgro M, Bianchetti L, Marini M, Bellini A, Schmidt M, Mattoli S. The CC motif chemokine ligands CCL5, CCL11, and CCL24 induce the migration of circulating fibrocytes from patients with severe asthma. Mucosal Immunol. 2013;6:718–727. doi: 10.1038/mi.2012.109. [DOI] [PubMed] [Google Scholar]
  • 123.Lloyd CM, Hawrylowicz CM. Regulatory T cells in asthma. Immunity. 2009;31:438–449. doi: 10.1016/j.immuni.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Isaacs KL, Lewis JD, Sandborn WJ, Sands BE, Targan SR. State of the art: IBD therapy and clinical trials in IBD. Inflamm Bowel Dis. 2005;11:S3–S12. doi: 10.1097/01.mib.0000184852.84558.b2. [DOI] [PubMed] [Google Scholar]
  • 125.Korzenik JR, Podolsky DK. Evolving knowledge and therapy of inflammatory bowel disease. Nat Rev Drug Discov. 2006;5:197–209. doi: 10.1038/nrd1986. [DOI] [PubMed] [Google Scholar]
  • 126.Kriegel C, Amiji M. Oral TNF-α gene silencing using a polymeric microsphere-based delivery system for the treatment of inflammatory bowel disease. J Control Release. 2011;150:77–86. doi: 10.1016/j.jconrel.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Bhavsar M, Amiji M. Oral IL-10 gene delivery in a microsphere-based formulation for local transfection and therapeutic efficacy in inflammatory bowel disease. Gene Ther. 2008;15:1200–1209. doi: 10.1038/gt.2008.67. [DOI] [PubMed] [Google Scholar]
  • 128.Laroui H, Theiss AL, Yan Y, Dalmasso G, Nguyen HT, Sitaraman SV, et al. Functional TNFα gene silencing mediated by polyethyleneimine/TNFα siRNA nanocomplexes in inflamed colon. Biomaterials. 2011;32:1218–1228. doi: 10.1016/j.biomaterials.2010.09.062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Xiao B, Chen Q, Zhang Z, Wang L, Kang Y, Denning T, et al. TNFα gene silencing mediated by orally targeted nanoparticles combined with interleukin-22 for synergistic combination therapy of ulcerative colitis. J Control Release . 2018;287:235–246. doi: 10.1016/j.jconrel.2018.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mueller C. Tumour necrosis factor in mouse models of chronic intestinal inflammation. Immunology. 2002;105:1–8. doi: 10.1046/j.1365-2567.2002.01329.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Arend WP. Physiology of cytokine pathways in rheumatoid arthritis. Arthritis Rheumatol. 2001;45:101–106. doi: 10.1002/1529-0131(200102)45:1<101::AID-ANR90>3.0.CO;2-7. [DOI] [PubMed] [Google Scholar]
  • 132.Park S, Le TT, Slejko JF, Villalonga-Olives E, Onukwugha E. Changes in opioid utilization following tumor necrosis factor inhibitor initiation in patients with rheumatoid arthritis. Rheumatol Ther. 2019;6:611–616. doi: 10.1007/s40744-019-00175-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kumar R, Dammai V, Yadava P, Kleinau S. Gene targeting by ribozyme against TNF-α mRNA inhibits autoimmune arthritis. Gene Ther. 2005;12:1486–1493. doi: 10.1038/sj.gt.3302583. [DOI] [PubMed] [Google Scholar]
  • 134.Schiffelers RM, Xu J, Storm G, Woodle MC, Scaria PV. Effects of treatment with small interfering RNA on joint inflammation in mice with collagen‐induced arthritis. Arthritis Rheumatol. 2005;52:1314–1318. doi: 10.1002/art.20975. [DOI] [PubMed] [Google Scholar]
  • 135.Howard KA, Paludan SR, Behlke MA, Besenbacher F, Deleuran B, Kjems J. Chitosan/siRNA nanoparticle–mediated TNF-α knockdown in peritoneal macrophages for anti-inflammatory treatment in a murine arthritis model. Mol Ther. 2009;17:162–168. doi: 10.1038/mt.2008.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Khoury M, Louis‐Plence P, Escriou V, Noel D, Largeau C, Cantos C, et al. Efficient new cationic liposome formulation for systemic delivery of small interfering RNA silencing tumor necrosis factor α in experimental arthritis. Arthritis Rheumatol. 2006;54:1867–1877. doi: 10.1002/art.21876. [DOI] [PubMed] [Google Scholar]
  • 137.Nissler K, Pohlers D, Hückel M, Simon J, Bräuer R, Kinne R. Anti-CD4 monoclonal antibody treatment in acute and early chronic antigen induced arthritis: influence on macrophage activation. ARD. 2004;63:1470–1477. doi: 10.1136/ard.2003.013060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Genprex Announces Initiation of its Phase 1/2 Acclaim-1 Clinical Trial for REQORSA™ Immunogene Therapy in Combination with Tagrisso® to Treat Non-Small Cell Lung Cancer Following FDA Review Yahoo finance 2021. [Available from: https://finance.yahoo.com/news/genprex-announces-initiation-phase-1-123000637]
  • 139.Malmström P-U, Loskog AS, Lindqvist CA, Mangsbo SM, Fransson M, Wanders A, et al. AdCD40L immunogene therapy for bladder carcinoma—the first phase I/IIa trial. Clin Cancer Res. 2010;16:3279–3287. doi: 10.1158/1078-0432.CCR-10-0385. [DOI] [PubMed] [Google Scholar]
  • 140.Tristán-Manzano M, Justicia-Lirio P, Maldonado-Pérez N, Cortijo-Gutiérrez M, Benabdellah K, Martin F. Externally-controlled systems for immunotherapy: from bench to bedside. Front immunol. 2020;11:2044–2061. doi: 10.3389/fimmu.2020.02044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Xu J, Melenhorst JJ, Fraietta JA. Toward precision manufacturing of immunogene T-cell therapies. Cytotherapy. 2018;20:623–638. doi: 10.1016/j.jcyt.2017.12.007. [DOI] [PubMed] [Google Scholar]

Articles from Iranian Journal of Basic Medical Sciences are provided here courtesy of Mashhad University of Medical Sciences

RESOURCES