Abstract
Immunotherapy has recently emerged as a powerful tool for cancer treatment. Early clinical successes from cancer immunotherapy have led to a growing list of FDA approvals, and many new therapies are in clinical and preclinical development. Nucleic acid therapeutics, including DNA, mRNA, and genome editing systems, hold significant potential as a form of immunotherapy due to its robust uses in cancer vaccines, adoptive T-cell therapies, and gene regulation towards the goal of treating cancer. However, these therapeutics must overcome numerous delivery obstacles to be successful, including rapid in vivo degradation, poor uptake in target cells, required nuclear entry, and potential in vivo toxicity in healthy cells and tissues. Nanoparticle delivery systems have been engineered to overcome several of these barriers as a means to safely and effectively deliver nucleic acid therapeutics to immune cells. In this Review, we discuss the applications of nucleic acid therapeutics in cancer immunotherapy, and we detail how nanoparticle platforms can be designed to deliver mRNA, DNA, and genome editing systems to enhance the potency and safety of these therapeutics.
Keywords: CRISPR, mRNA, DNA, gene editing, drug delivery, oncology, nanotechnology
Introduction
Despite advances in understanding the underlying mechanisms of cancer progression, chemotherapy, radiation, and surgery remain the current standards-of-care for many cancers [1]. The use of these strategies has become more focused and personalized based on the type and stage of disease, which has led to a decline in cancer-related mortality over the past three decades [2]. However, these therapies are often highly invasive, have substantial adverse side effects, and therapeutic results are variable [3–5]. Thus, there is a dire need to develop non-invasive, minimally toxic, and highly specific alternatives. Towards this goal, cancer immunotherapy has emerged as a powerful alternative to conventional therapies, and substantial research efforts are ongoing to improve upon their delivery and success, many of which we discuss below.
The overarching goal of cancer immunotherapy is to introduce the necessary molecular tools to harness the immune system to halt disease progression. Thus, immunotherapy can be personalized for specific types and stages of cancer, with higher safety profiles and longer therapeutic windows compared to traditional cancer therapies [6]. The field encompasses several classes of immunotherapy, including gene therapy, cellular vaccines, checkpoint inhibitors, agonistic antibodies, and cytokines [7]. Of these, checkpoint inhibitors and cytokines are the most widely studied to date, and multiple therapies are currently used in the clinic [7]. More recently, nucleic acid therapeutics including DNA, mRNA, and CRISPR/Cas9 gene editing systems have emerged as an important branch of cancer immunotherapy. The vast potential of nucleic acids for treating cancer is demonstrated by the use of CRISPR/Cas9 to inactivate PD-1/PD-L1 interactions between cancer cells and T-cells [8]. In one example of this, CRISPR/Cas9 genome editing was used to generate PD-1 deficient anti-CD19 chimeric antigen receptor (CAR) T-cells, resulting in enhanced killing of PD-L1+ tumor xenografts [8]. Similarly, gene therapy approaches are highly prevalent in adoptive T-cell immunotherapy to induce T-cells to express CARs. In 2017, Novartis gained the first FDA approval for cell-based gene therapy with Kymriah, which utilizes CAR T-cells to treat leukemia [9,10]. The early success of Kymriah and the ability for CRISPR/Cas9 to enhance T-cell-mediated killing forms the basis for the development of other types of gene therapies to treat cancer with reduced adverse effects and higher success rates than traditional approaches [9,10].
Although the examples described above demonstrate the therapeutic potential of nucleic acid therapeutics, their translation into the clinic is hindered by several delivery challenges for both ex vivo and in vivo applications. Nucleic acids are highly unstable, and they degrade quickly in the presence of nucleases before reaching the desired tissues [11]. Further, nucleic acids are unable to enter cells alone, requiring the use of transfection reagents or physical techniques (such as electroporation) that are highly toxic to cells ex vivo and are not feasible for in vivo use [12,13]. Several nucleic acid therapeutics, such as gene editing components and DNA, are faced with another delivery barrier of crossing the nuclear membrane to be transcribed in the nucleus [14]. Thus, there is great interest in developing novel delivery platforms that can encapsulate and protect nucleic acids, as well as mediate their delivery into the desired tissues and cells, in order to exploit the powerful therapeutic potential of these molecules.
Nanoparticles (NPs), which are typically defined as ultra-small particles that are 1–1000 nm in diameter, are being developed to overcome the delivery barriers faced by nucleic acids. NPs can be comprised of a range of materials such as lipids, polymers, or metals, all of which offer unique delivery advantages that have been thoroughly reviewed elsewhere [11,15,16]. Importantly, NP features, such as their material composition, size, and surface chemistry, can be carefully engineered for nucleic acid delivery. NPs can encapsulate or bind to nucleic acid therapeutics based on electrostatic interactions or chemical conjugation to overcome the therapeutic challenges faced by unbound nucleic acids [6,7]. First, NPs can reduce the toxicity of the therapeutics by promoting site-specific accumulation and reducing off target effects. Further, NPs offer protection over the therapeutic cargo to avoid nuclease degradation in circulation and to extend circulation half-life. In addition to protecting the nucleic acids, these NP carriers can be engineered to respond to acidic environments, such as in solid tumors or within endosomes inside cells, to degrade and release their cargo on-demand [17]. By enabling control over nucleic acid delivery, NPs can minimize toxicities to healthy tissues while maximizing delivery to cancer cells, which may be highly beneficial for solid tumor immunotherapy [18]. Lastly, NPs can be modified with targeting ligands and other molecules to promote both cellular and nuclear uptake and biodistribution to the targeted tissues that overexpress the targeted protein [19].
Here, we review the design of NP platforms for nucleic acid delivery - including mRNA, DNA, and genome editing therapies – and their applications in cancer immunotherapy. Several NP platforms have demonstrated preclinical success in delivering nucleic acids to target cells, and significant efforts are now underway to translate these technologies into the clinic. Of note, a lipid NP (LNP) complexed with mRNA is currently being evaluated in clinical trials of melanoma ( NCT02410733) [20]. Further, Alnylam Pharmaceuticals received the first FDA approval of an RNA therapeutic for their lipid-siRNA NP, Onpattro, in 2018 [7,21]. Below, we overview applications of NPs for delivering DNA, mRNA, and genome editing systems for cancer immunotherapy, and we discuss future directions of gene therapy towards the goal of clinical translation.
Nanoparticles for DNA Delivery
DNA vaccine-based cancer immunotherapy, in which cells are transfected with plasmid or chemically synthesized DNA to elicit immune responses against the encoded antigen, is a powerful tool to engage the immune system to attack cancer cells [22]. Early studies in mice demonstrated the ability of DNA plasmids to drive immune responses against transgene products related to influenza, human immunodeficiency virus-1, and cancer, which established DNA as a promising immunization platform [23]. However, initial clinical applications of DNA vaccines revealed only low levels of immunity, indicating that naked DNA was not feasible as an independent vaccination strategy, which is largely due to delivery barriers (Fig. 1) [23]. First, nucleic acids are degraded in the presence of nucleases and, without additional modifications, they are rapidly cleared from circulation [24]. Further, the negatively charged DNA typically cannot cross the anionic cell membrane without an exogenous transfection reagent or delivery vehicle [24]. Once within cells, DNA needs to surpass the nuclear membrane and enter the nucleus [24]. Lastly, it is critical that DNA is transfected into the desired cells with minimal off-target expression [25–28]. Several physical techniques to improve DNA delivery including gene guns, electroporation, and sonoporation are commonly used ex vivo and in small animals [29,30], but they are either not feasible in vivo, or they are limited to local delivery [24]. Utilizing NPs as DNA delivery vehicles can overcome the aforementioned limitations, and several unique applications are described below [24].
Figure 1.
The role of non-viral vectors in overcoming extracellular and intracellular barriers for nucleic acid delivery. In circulation, non-viral vectors need to protect nucleic acids from serum endo- and exonucleases, evade immune detection, and avoid non-specific protein interactions within the blood. Further, vectors must avoid renal clearance (achieved through size modulation), while also promoting extravasation from the blood and into target tissues, upon which they promote cellular uptake and localization into the cytosol or nucleus. Adapted from [24].
Several types of LNPs, including liposomes, ionizable lipids, and polymer-lipid NPs, have been developed to deliver DNA to target cells. Liposomes were among the first DNA delivery systems and are the furthest in clinical development, as they are currently used clinically to treat cancer [31]. Liposomes are composed of materials with polar head groups and non-polar tails, and they spontaneously self-assemble into vesicles at low concentrations [32,33]. Cationic lipids, such as DOTMA, DOTAP and zwitterionic DOPE, are commonly used to form cationic liposomes by exploiting electrostatic interactions between the lipids and the negatively charged nucleic acids (Fig. 2a). When used to encapsulate DNA or other drugs, these cationic liposomes induce stronger therapeutic effects than free drug, which has led to many cationic liposomal drug formulations advancing into clinical trials [34–38]. However, the use of cationic liposomes is limited due to toxicity at the site of administration [39–41], undesired immune responses [42], and clot formation [43], all of which can limit the allowable administered dose [41,44–48].
Figure 2.
Chemical structures of common non-viral vectors used for nucleic acid delivery. A. Common lipids used for liposomal formulations including DOTMA, DOSPA, DOTAP, DMRIE and DC-cholesterol, which are used to condense and encapsulate nucleic acids. Structurally, cationic lipids are defined as having a cationic head group, linker region, and hydrophobic tails. B. Ionizable LNP formulations are comprised of four components: ionizable lipids, such as C12– 200, phospholipids (DOPE, DSPC), cholesterol, and lipid-anchored PEG. C. Cationic polymers and biopolymers used as vectors for nucleic acid delivery. PEI and PLL were two of the initial vectors used for DNA delivery but are faced with safety (PEI) and efficacy (PLL) concerns. PBAEs and pDMAEMA are newer polymer vectors developed for nucleic acid delivery with improved safety and efficacy. Panels A and B are adapted from [24].
As an alternative to traditional cationic liposomes, ionizable lipids that are neutral at physiologic pH (~7.4) but ionize under mild acidic conditions, such as those found within endosomes, have been developed for nucleic acid delivery [49–51]. The ability of these lipids to buffer endosomal compartments by taking on positive charges can promote endosomal escape to enable processing of the nucleic acids in the cytosol [51,52]. Ionizable LNPs typically have 3 components in addition to the ionizable lipid itself; a fusogenic helper phospholipid (DSPC, DOPE, DOTC, DOTMA, POPC) [53,54], cholesterol for increased stability and membrane fusion [55,56], and a lipid-anchored poly(ethylene glycol) (PEG) to extend their circulatory half-life and decrease non-specific protein adsorption (Fig. 2a,b) [54]. Ionizable LNPs have been used for cancer immunotherapy by encapsulating CpG (a TLR-9 agonist) oligodeoxynucleotides (ODNs). In this application CpG-NPs were subcutaneously co-administered with tumor associated antigens in murine models of thymoma and melanoma [57]. NPs exhibited preferential accumulation and uptake by immune cells in lymph nodes and augmented antigen-specific immune cell and cytokine/chemokine responses, ultimately leading to greater tumor rejection in a murine EG7-OVA tumor model [57]. Although ionizable LNPs have been shown to effectively load nucleic acids of relatively small size (e.g. short synthetic DNA, siRNA, and microRNA), encapsulating large cargo (e.g. pDNA) is challenging [58–60]. Thus, new classes of polymer-based NP carriers, such as polyplexes [61–63], chitosan-based NPs [64,65], and poly(beta-amino esters (PBAEs) [66–68], can effectively condense pDNA into NPs and enhance transgene expression as described below.
Cationic polymeric NPs can be engineered to possess specific physicochemical properties, such as hydrophobicity and charge, due to the diverse range of available polymers and chemical modifications [27]. This chemical diversity allows researchers to utilize polymeric NPs for delivery to a wide array of cell types [69]. Poly(l-lysine) (PLL) is a homopolymer of the amino acid lysine that has been shown to effectively condense DNA (Fig. 2c) [70]. Studies indicate that PLL generally has low transfection success, likely due to its low rate of endosomal escape [24]. However, one cancer immunotherapy application used a PLL coating onto polystyrene NPs to deliver pDNA encoding OVA antigen as a model for a DNA-based prophylactic cancer vaccine against EG7 tumor cells [70]. Two vaccinations with these NPs inhibited tumor growth following challenge with EG7 tumor cells in mice [70]. Notably, immature dendritic cells (DCs) had higher levels of NP uptake compared to mature DCs [70]. This is likely due to the reduced endocytic and phagocytic rates in mature DCs, which lowers their capacity to internalize and process antigens [70].
Similar to PLL, polyethylenimine (PEI) is another cationic polymer that is often used as the “gold-standard” for transfection efficacy (Fig. 2c) [24,71]. PEI exerts a high charge density at low pH, which enhances endosomal escape and makes it a potent transfection reagent, but it also confers high cytotoxicity [72–76]. Longer chain and higher charge density PEIs tend to have damaging interactions with cellular membranes that lead to potent cytotoxicity, and several strategies have emerged to address this including branched architectures, biodegradability, and PEG-grafting [74,75,77]. In one instance, modified branched PEI was synthesized to improve upon the cytocompatibility and transfection efficiency of the unmodified PEI [74]. Of note, succinylated PEI induced better siRNA-mediated knockdown and 10-fold lower polymer toxicity compared to unmodified PEI [74]. This demonstrates the importance of balancing transfection efficiency and biocompatibility when designing PEI-based delivery vehicles. The high transfection of PEI was exploited for cancer immunotherapy by condensing IL-12-encoding pDNA [78]. This therapy was administered as an aerosol to mice bearing SAOS-LM7 tumors in a murine model of osteosarcoma lung metastases [78]. Mice receiving aerosolized PEI-IL-12 gene therapy exhibited IL-12 expression only in the lungs and had significantly fewer lung metastases than untreated controls [78]. The ability of PEI to condense DNA is also applicable to newer polymer-based delivery platforms, such as PBAEs, described below.
PBAEs are straightforward to synthesize and they provide an additional benefit of having tunable biodegradation (Fig. 2c) [79]. A major advantage of their simple, parallelizable synthesis is the ability to generate diverse libraries of PBAE structures that can then be screened for DNA delivery, which aids in the identification of key structures for potent gene delivery for immunotherapy [80]. Towards this goal, PBAE NPs functionalized with an anti-CD3e T-cell-targeting antibody fragment were used to deliver leukemia specific CD194–1BBz CAR pDNA to T-cells in situ in a murine leukemia model (Fig. 3a) [81]. These NPs also contained microtubule-associated sequence (MTAS) and nuclear localization signal (NLS) peptides to mediate nuclear translocation of the therapeutic pDNA cargo [81]. NP-programmed CAR T-cells generated tumor regressions similar to that of traditionally prepared CAR T-cells, with only a small portion of NPs transducing phagocytic cells, potentially due to successful antibody targeting to T-cells [81]. PBAEs have also been used to deliver cyclic dinucleotides (a STING agonist) or CpG nucleic acid adjuvants (Fig. 3b) [82,83]. Notably, results indicate that the PBAE:DNA ratio is a critical factor for NP stability and in vivo functionality. Specifically, PBAE-NPs with higher PBAE:DNA ratios yield better protection of the CpG cargo. However, lower ratios exhibit better CpG uptake and activation of tumor-specific T-cells, resulting in improved survival in a mouse melanoma model [82,83].
Figure 3.
NP delivery platforms used for gene therapies. A. PBAE polymer functionalized with an MTAS-NLS peptide was used to condense CAR-encoding plasmid DNA. In this application, an anti-CD3e-poly(glutamic acid) (PGA) conjugate was adsorbed to the surface of the PBAE core to enable T-cell targeting and in situ generation of CAR T-cells. Adapted from [81]. B. PBAE polymer used to deliver a Stimulator of Interferon Receptor Genes (STING) antagonizing cyclic dinucleotide (CDN) intratumorally in combination with a PD-1 blocking antibody and demonstrated potent inhibition of tumor growth. Adapted from [83]. C. A biodegradable ionizable lipid was used to co-deliver a modified sgRNA and Cas9 mRNA that achieved potent gene editing in the liver for 12 weeks. sgRNA was modified with phosphothiorate bonds at both ends of the strand (indicated by *) and 2’-O-methylation of nucleotides (shown in red). Adapted from [126]. D. Multilamellar ionizable lipid NPs generated potent CD8 T-cell activation upon antigen delivery and were used to deliver tumor antigens gp100 and TRP2 that led to tumor shrinkage and elongated survival in a B16F10 melanoma mouse model. Adapted from [49].
With the aid of NP delivery systems, DNA based therapeutics have shown great promise in the field of cancer immunotherapy [84]. Although using pDNA as an antigen source has shown encouraging outcomes in many preclinical studies, the same success has not been found in human clinical trials, and interest in using DNA as antigen sources has decreased [84,85]. However, there is substantial ongoing work to develop DNA NPs in immunotherapy for CAR T-cells or as adjuvants [81,82,86]. More recently, mRNA has emerged as a potent tool for gene immunotherapy for cancer, and several unique applications are described below.
Nanoparticles for mRNA Delivery
Early interest in mRNA stemmed from its use as an alternative to conventional and DNA-based vaccines [87]. mRNA therapeutics are a promising alternative to DNA owing to their lower mutational risk, fewer intracellular delivery barriers, and transient expression [24,88,89]. Further, mRNA only needs to cross the cell membrane and reach the cytosol – in contrast to DNA which requires nuclear entry - to induce protein translation [88–90]. Finally, protein expression induced by mRNA is transient and does not require integration into the genome, thereby avoiding the risk of insertional mutagenesis that can occur from DNA [24,88,89]. When used as a vaccine, mRNAs encoding for antigens are delivered to antigen presenting cells, either through ex vivo transfection or under systemic administration. Antigen presenting cells then translate mRNA into its encoded cancer associated antigen that is presented to T-cells for activation and induction of cytotoxic T lymphocyte responses [87]. However, the large size (105-106 base pairs), negative charge, and hydrophilicity of mRNA, combined with its susceptibility to nucleases, hinder the ability of naked mRNA to reach and enter target cells upon systemic administration [14,24,88–90]. NPs can overcome these barriers and facilitate its intracellular delivery, and several NP platforms for mRNA delivery are described below.
Similar to DNA delivery, ionizable LNPs have also been used for mRNA delivery. In one example of this, LNPs comprised of an ionizable lipid, a helper phospholipid, cholesterol, lipid-anchored polyethylene glycol (PEG), and mRNA were designed to induce expression of luciferase and erythropoietin following systemic injection in BALB/c mice [91]. The study utilized Design of Experiment (DoE) methodologies used in combinatorial chemistry to adapt a top-performing LNP for delivery of mRNA, demonstrating that LNPs may hold potential for delivering mRNA vaccines for cancer immunotherapy [91]. More recently, multilamellar ionizable LNPs were used to deliver tyrosine-related protein 2 (TRP2) and glycoprotein 100 (gp100) tumor self-antigen mRNAs to antigen presenting cells (Fig. 3d) to induce a cytotoxic CD 8 T-cell response. Subcutaneous administration of these LNPs led to reductions in tumor volume, extended survival in a B16F10 tumor model, and potent CD8+ activation [49]. Interestingly, these LNPs were able to transfect neutrophils, macrophages, and dendritic cells, demonstrating that they may be useful to deliver mRNA to a range of immune cells.
Similar to DNA delivery, the chemical diversity of polymers and polymer-lipid systems allows for identification and incorporation of structures that can improve biocompatibility and bioavailability of the encapsulated materials [27]. Polymers used for mRNA delivery are positively charged and can condense mRNA into nanometer sized electrostatic complexes [88]. PBAEs are a major class of pH-responsive and bioreducible polymers known for their biocompatibility, but they have had limited in vivo success due to their poor serum stability [92,93]. Recent PBAE work has explored the incorporation of PEG-lipids and new PBAE architectures to improve uptake, stability, and biodistribution [94–96]. In one approach, PBAEs were modified to include internal alkyl tails to enable formulation with PEG-lipids and improve their stability under physiologic conditions [96]. mRNA-loaded PBAE terpolymers incorporating PEG-lipids demonstrated greater serum stability in vitro compared to the terpolymers without PEG-lipids, and they achieved near-exclusive luciferase expression in the lungs following intravenous injection in healthy mice [96]. These have also been used for mucosal immunization, where lipid-enveloped NPs with pH-responsive PBAE cores delivered mRNA to immune cells [97]. Mucosal immunization has drawn interest for two reasons: 1) many pathogens invade through mucosal surfaces and 2) mucosal immunizations can elicit both systemic and mucosal immunity [98,99]. These NPs successfully delivered GFP mRNA to difficult-to-transfect dendritic cells in vitro as well as luciferase mRNA to mucosal tissue compartments following intranasal administration in vivo [97].
Vaccination is a central application of mRNA for cancer immunotherapy. In these therapies, electroporation is typically used to introduce mRNA into T-cells ex vivo, which can induce cellular toxicity [13]. Further, this ex vivo cell engineering process is time, labor, and cost intensive, which creates significant challenges towards broader clinical translation [100]. Thus, NPs are ideal to deliver mRNA to T-cells without the need for electroporation. An early mRNA cancer vaccine approach utilized PEGylated histidine-rich polylysines mixed with L-histidine- (N,N-di-n-hexadecylamine)ethylamide (HDHE) and cholesterol liposomes, referred to as histidylated lipopolyplexes, to deliver mRNA encoding human melanoma antigen MART1 to T-cells. Immunization with MART1 histidylated lipoplexes induced priming of B16-specific CD4+ and CD8+ T-cells, leading to a ~10-fold reduction in tumor volume and a 75% reduction in detectable lung metastases compared to control mice in a B16/F10 melanoma model [101]. More recently, PBAE NPs were coated with CD3 or CD8 antibodies and used to target T-cells in order to induce receptor-mediated endocytosis. Antibody-coated NPs improved ex vivo T-cell transfection 10-fold compared to non-targeted PBAE NPs [12]. Building upon this success, this robust platform has been used to deliver two different mRNAs. In one study, these NPs delivered megaTAL nuclease mRNA to knockout endogenous T-cell receptors that may cause graft-versus-host disease [12]. Separately, NPs loaded with an mRNA encoding the Foxo13A transcription factor were used to guide CD62L+ T-cells away from terminal differentiation and senescence and towards a central memory phenotype [12]. When taken together, these results indicate that this robust platform can be adapted to generate several distinct immune responses.
Recently, mRNA vaccines have begun testing in clinical trials. The clinical translation of mRNA vaccines is being led, in part, by Curevac ( NCT03291002) with an RNA-based adjuvant that is being tested in patients with melanoma and squamous cell carcinoma, among others. With the introduction of these vaccines into the clinic, researchers are increasingly working towards introducing mRNA delivery systems as well. LNPs complexed with NY-ESO-1, MAGE-A3, tyrosinase, and TPTE mRNA are currently being evaluated in clinical trials, and early phase 1 dose escalation data has demonstrated that neutral or negatively charged LNP-mRNA complexes are well tolerated and there were dose dependent IFN- and antigen-specific T-cell responses in three melanoma patients [20]. Together with the preclinical studies described above, this demonstrates that NPs can aid in overcoming challenges associated with mRNA delivery and enable potential use for multiple forms of cancer immunotherapy.
Nanoparticles for Gene Editing
Since its first use for genome editing in 2013, CRISPR/Cas9 has emerged as a powerful tool in understanding and treating the genetic cause of various diseases (Fig. 4) [102,103]. In the context of cancer immunotherapy, CRISPR has been applied to disease modeling [104–106], target identification [107], and immune cell engineering [108–111]. CRISPR/Cas9 can be delivered as a nucleic acid-loaded protein (ribonucleoprotein, or RNP) or purely as nucleic acids [112,113]. Protein delivery presents several challenges, and strategies for overcoming these barriers have been reviewed elsewhere [114–116]. In contrast to nucleic acids, the chemical diversity and size of proteins often necessitates modifications to enable delivery with a vector [117,118]. For example, a successful approach is to “supercharge” proteins through the addition of densely charged moieties to enable electrostatic complexation with carriers [117,118], and this approach has been particularly effective in the localized delivery of Cas9 RNP NPs [119–121].
Figure 4.
Methods for genome editing systems. Zinc-finger nucleases (ZFN), transcription activator-like nuclease (TALEN), or CRISPR-Cas systems can be delivered with non-viral delivery platforms. These gene editing systems can edit mammalian genomes by introducing double stranded breaks in a highly specific, sequence-dependent manner. Repair occurs through non-homologous end-joining or by homology-directed repair. Adapted from [112].
Delivery of Cas9 protein and mRNA offer transient protein expression, which is potentially beneficial because constitutive expression increases the risk for off target editing and stimulation of preexisting adaptive immune responses to the Cas9 protein [122–124]. However, the most successful attempts at formulating Cas9 RNP into NPs have been limited to localized delivery, whereas Cas9 mRNA has been successfully delivered via systemic administration [119–121,124–126]. Since CRISPR/Cas9 technology is still relatively new, there have been few attempts to deliver Cas9 components using NPs for the purpose of cancer immunotherapy. Here, we highlight NP delivery systems that have delivered CRISPR/Cas9 to treat non-cancer diseases, as well as viral delivery mechanisms for cancer immunotherapy. Moving forward, we anticipate that the critical insights gained from the gene editing studies discussed below will form the basis for NP-mediated gene editing for cancer immunotherapy.
One of the first successful approaches utilized viral and non-viral NP delivery to achieve homology directed repair in hepatocytes in a mouse model of hereditary tyrosinemia type 1 [124]. A lipidoid-based LNP was used to encapsulate Cas9 mRNA and, due to size constraints, the sgRNA expression cassette and homology directed repair template were delivered using an adeno-associated virus (AAV) [124]. After optimizing the timing of LNP and AAV administration to maximize the overlap between peak Cas9 and sgRNA expression, this system achieved gene editing in 6% of hepatocytes at a 24.1% indel rate measured by deep sequencing of the target locus in total liver genomic DNA [124]. In a different proof-of-concept study, ionizable lipids and helper lipids were used to co-deliver Cas9 mRNA and modified sgRNA to reduce serum concentrations of transthyretin and achieved 70% gene editing and >97% knockdown in hepatocytes following a single intravenous injection in mice (Fig. 3c) [126]. In another example of NP-mediated Cas9 delivery, Miller et al synthesized a library of zwitterionic amino lipids to co-deliver Cas9 mRNA and Lox sgRNA to mice expressing a Lox-Stop-Lox tdTomato cassette, and demonstrated stable gene editing two months after NP administration [125]. ZALs demonstrated potent protein expression with luciferase mRNA at doses <600 pM in vitro and l mg/kg in vivo [125]. Together, these early examples of NP delivery platforms for genome editing preface their use for CRISPR/Cas9 nucleic acid delivery for cancer immunotherapy.
In the preceding text, we highlighted successful preclinical applications of NPs for the delivery of Cas9 components. Here, we describe the use of CRISPR/Cas9 for cancer immunotherapy mediated by viral delivery. One important use of CRISPR/Cas9 is towards more robust T-cell engineering. Allogeneic CAR T-cells are an attractive alternative to traditional autologous CAR T-cells because they can be distributed “off the shelf” to patients [127,128]. However, allogeneic T-cell transplants’ TCRs can be reactive to host antigens in healthy tissues, leading to graft-versus-host disease (GVHD) [128,129]. Additionally, alloantigens present on transplanted cells, such as human leukocyte antigen-1 (HLA-1), can elicit unwanted host immune responses [128,129]. CRISPR/Cas9 could be employed to knock-out surface molecules to improve the compatibility of allogeneic CAR T-cells [128–131]. An early study utilized a combination of CRISPR mRNA and gRNA to target the T-cell Receptor alpha-constant (TRAC) locus and knock out T-cell receptors (TCRs). Subsequent transfection with an AAV encoding CAR cDNA was used to induce expression of CD19-specific CAR under transcriptional control of the TRAC promoter. These T-cells were more resistant to tonic signaling and had delayed differentiation and exhaustion, ultimately leading to greater tumor rejection when compared to retrovirally transduced CARs, both with and without TCR knockout [108]. CRISPR/Cas9 targeting enabled the identification of a specific transcriptional regulator that modulated CAR expression to maximize therapeutic benefit.
While single gene editing has several applications, the simplicity of CRISPR/Cas9 gives rise to the capability of multiplexed gene editing to simultaneously knock-out several genes. In one example, Cas9 mRNA and gRNAs were delivered to primary T-cells via electroporation and used to knock out PD-1, a suppressor of CD8 T-cell activity, in addition to TCR and HLA-1 [111,132]. A lentivirus was used to transduce CD19 or prostate-stem cell antigen (PSCA) CARs. Double disrupted (TCR- and HLA-1-) CAR T-cells yielded reduced alloreactivity compared to single knockout (TCR-) CAR T-cells while maintaining potent antitumor activity, measured by enhanced survival in a Nalm6 mouse tumor model [111]. Triple disrupted (TCR-, HLA-1-, and PD-1-) CAR T-cells demonstrated quicker and complete elimination of tumor cells compared to double ablated CAR T-cells in an aggressive Nalm6-PDL1 leukemia model [111]. These results highlight the utility for CRISPR/Cas9 technologies for improving CAR T-cell therapy and their potential for clinical application.
CRISPR/Cas9 is potent new tool for treating and understanding disease, but effective intracellular delivery remains a challenge. The applications of CRISPR/Cas9 in cancer immunotherapy previously mentioned rely on combinations of physical and viral delivery methods to achieve gene editing [108,111]. Additionally, Cas9 transfection was done ex vivo and in homogenous cell populations, further simplifying the challenge of delivery [108,111]. However, there has been successful NP mediated delivery of Cas9 components to immune cells. In one example of targeted CRISPR/Cas9 expression, CRISPR/Cas9 pDNA was delivered using cationic lipid assisted polymeric NPs (CLANs) and administered intravenously to type 2 diabetic mice. The CRISPR/Cas9 gene was designed to be under transcriptional control of the CD68 promoter unique to monocytes and macrophages in order to decrease off-target Cas9 expression [133]. These particles were used knock out Ntn1, a guidance cue that blocks macrophage migration in adipose tissue and leads to insulin sensitivity and inflammation, and they achieved gene editing in vivo primarily in macrophages and monocytes, with little editing occurring in neutrophils or other off target immune cells [133,134]. In another example of NP-mediated Cas9 delivery, Li et. al screened a library of PEG-PLGA, PLGA, BHEM-Chol, and DOTAP NPs for delivery of Cas9 plasmids to B cells in vivo. After identifying a lead candidate based on accumulation in the spleen and lymph node, the lead formulation was used to correct B cell dysfunction in a rheumatoid arthritis model [135]. Although NPs are still being developed to deliver CRISPR/Cas9 components to immune cells, these early examples of successful in vivo delivery preclude their use for gene editing in cancer.
As previously mentioned, the field of NPs to deliver CRISPR/Cas9 for cancer immunotherapy is nascent. The early research into the role of gene editing in cancer immunotherapy, especially the examples of T-cell engineering mentioned above, has yielded promising results as well as insight into the future directions in immunotherapy [108,111]. Concurrently, NPs are actively being developed to deliver Cas9 components, and have demonstrated efficacy in doing so in both immune and non-immune cells [124–126,133,135].
Future Directions and Conclusions
Currently, most forms of immunotherapy and their NP-based delivery systems have been effective at targeting hematological cancers or melanoma [7]. Moving forward, there is great opportunity to study how NPs can be utilized to treat solid tumors by exploiting their unique physicochemical properties. This engineering challenge can be faced by carefully choosing materials that are known to efficiently transfect cells (such as cationic lipids and polymers) [11,50], have highly controllable sizes and surface chemistry [16,24,136], and those that can be easily coated with targeting ligands to promote uptake by the tissues and cells of interest. In an elegant example of NP targeting for solid tumor immunotherapy, LNPs loaded with a DNA plasmid and cationic protamines were coated with targeting ligands to activate dendritic cells [137]. These targeted NPs were intravenously injected into mice with orthotopic colorectal tumors, and mice treated with the NPs and chemotherapy experienced greater tumor inhibition compared to mice treated with chemotherapy alone [137]. The improvements gained by attaching targeting ligands to NPs can be attributed to enhanced target binding strength, biodistribution, and uptake that may decrease the doses required to produce therapeutic effects [138].
The improvements afforded by attaching targeting ligands to NPs are particularly attractive for transfecting various subtypes of lymphocytes directly in vivo for adoptive T-cell therapies. For example, PEG-PEI targeted to T-cells using CD3 antibodies have been shown to induce transfection and functional regulation of T-cells in vivo in a rat allogeneic heart transplantation model [139]. In the context of cancer immunotherapy, antibody-targeted PLGA NPs have been engineered to bind PD-1+ T-cells in blood, lymphoid tissues, and tumors to deliver a TGFβ inhibitor [136]. These complexes were shown to improve survival of melanoma-bearing mice compared to administration of free drug, an effect attributed to targeting this specific population of T-cells. This study demonstrates the concept that NPs can be used to isolate specific types of lymphocytes in vivo, which has the potential to expand the field of cancer immunotherapy in several ways. First, targeting T-cells in vivo can initiate specific anti-tumor immune responses for solid tumor immunotherapy, as described in the study above [136]. Second, this technique can be used to generate CAR T-cells directly in vivo to overcome the many manufacturing limitations of in vitro CAR T-cell development, including high costs and production time [12]. Lastly, the ability to activate specific subpopulations of immune cells in circulation may be exploited to treat metastatic secondary tumors in addition to primary tumors, as these engineered cells can target any cancer cells throughout the body that express the targeted protein [101].
Another means of utilizing nanotechnology to deliver immunotherapy for solid tumor treatment is by alleviating immunosuppressive signaling within the microenvironment to improve native T-cell responses (Fig. 5) [140]. For example, delivering siRNA against Snail, a critical transcription factor that accelerates cancer metastasis by inducing immunosuppression, to tumor cells was shown to promote the infiltration of tumor-specific lymphocytes into melanoma tumors [140]. This increase in lymphocyte infiltration led to both inhibited primary and metastatic tumor growth [140]. By utilizing NPs to enhance anti-tumor immune responses against solid tumors, researchers can exploit the inherent function of immune cells to attack cancer cells, which may be more effective for treating solid tumors compared to treating tumor cells themselves [141]. Moving forward, this technique to potentially make solid tumors more susceptible to native immune activity could be combined with in vivo targeting of immune cells for a multi-pronged approach to solid tumor immunotherapy [141].
Figure 5.
Sites of therapeutic intervention for NPs to generate anti-tumor immune responses in solid tumors. Anti-tumor immune responses result from the presentation of tumor-associated antigens (TAAs), stimulating protective T-cell responses, and overcoming the immunosuppressive tumor microenvironment (TME). NPs can be used to activate these pathways to successfully deliver immunotherapeutics to solid tumors by: (1) enhancing delivery of nucleic acids encoding TAAs to improve delivery to antigen presenting cells for immune activation; (2) delivering nucleic acids to T-cells to promote their survival, proliferation, and anti-tumor phenotypes; and (3) alleviating the immunosuppressive signaling within the tumor microenvironment. Figure adapted from [141]
In addition to developing delivery systems to enable solid tumor immunotherapy, it is also critical to engineer NPs to deliver gene editing tools [24]. Previously, we discussed the use of NPs to deliver CRISPR/Cas9 gene editing material to immune cells to treat diabetes and rheumatoid arthritis, as well as the viral delivery of gene editing technology for cancer immunotherapy [108,111,133,135]. There are several challenges to address as researchers develop nanotechnology to deliver gene editing components in vivo for cancer immunotherapy. A critical consideration is developing NPs that offer precise control over timing of nucleic acid delivery and release, as successful gene editing requires delivery of both guide RNA as well as the Cas9 protein or mRNA [124]. This challenge can be met by developing NPs using materials with highly tunable degradation profiles to release the nucleic acid cargo on-demand [18]. Additionally, NPs carrying gene editing material need to successfully edit a sufficient number of cells to mediate the desired therapeutic result, which can be difficult in vivo where immune cells are circulating throughout the body. This requirement can be met by attaching targeting ligands to NPs to promote their binding and uptake to the targeted cells [19,136]. Ultimately, specially designed NPs that can deliver gene editing technology for cancer immunotherapy can exploit the delivery capabilities of the carriers as well as the highly specific editing afforded by CRISPR/Cas9 systems.
The NP delivery platforms in this article above represent novel and recent developments in nucleic acid delivery, with several applications in cancer immunotherapy. However, challenges remain before these systems can be used broadly in the clinic. For example, several of the examples discussed above utilize materials that have not yet been used in clinical trials or are not yet approved by the FDA. NPs comprised of FDA approved materials may have a simpler and expedited path towards clinical translation. Importantly, several NP-based gene therapies are in clinical trials or have been recently approved by the FDA. For example, in 2018 Alnylam Pharmaceuticals gained FDA approval for their lipid-siRNA NP Onpattro to treat polyneuropathy caused by transthyretin amyloidosis [142]. This introduction of NP-nucleic acid complexes into the clinic is a critical milestone for the entry of other NP-based immunotherapies into clinical trials and ultimately for FDA approval. As new NP-based therapies appear in the clinic, physicians and scientists can begin to shift the current paradigm of cancer therapy towards potent and biocompatible nucleic acid delivery systems for immunotherapy.
Highlights.
Immunotherapy has emerged as a powerful tool for the treatment of cancer.
Gene therapy approaches (DNA, mRNA, genome editing) are a promising class of immunotherapy.
Nucleic acid therapeutic success requires overcoming multiple extracellular and intracellular barriers.
Nanoparticles have been engineered to deliver nucleic acid therapeutics to immune cells.
These delivery systems can potentially enhance cancer vaccination and CAR-T cell therapies.
Acknowledgements
The authors acknowledge support from a Burroughs Wellcome Fund Career Award at the Scientific Interface (CASI), a US National Institutes of Health (NIH) Director’s New Innovator Award (DP2 TR002776), a grant from the American Cancer Society (129784-IRG-16–188-38-IRG), and a 2018 AACR-Bayer Innovation and Discovery Grant, Grant Number 18–80-44-MITC (to M.J.M.). R.S.R. is supported by an NIH T32 multidisciplinary training grant. A.J.M. is supported by an NSF Graduate Research Fellowship.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interests
The authors have no conflicts of interest to declare.
References
- [1].Burstein HJ, Krilov L, Aragon-Ching JB, Baxter NN, Chiorean EG, Chow WA, De Groot JF, Devine SM, DuBois SG, and El-Deiry WS, Clinical cancer advances 2017: Annual report on progress against cancer from the american society of clinical oncology, Journal of Clinical Oncology 35 (2017) 1341–1367. [DOI] [PubMed] [Google Scholar]
- [2].Siegel RL, Miller KD, and Jemal A, Cancer statistics, 2018, CA: A Cancer Journal for Clinicians 68 (2018) 7–30. [DOI] [PubMed] [Google Scholar]
- [3].Lameire N, Nephrotoxicity of recent anti-cancer agents, Clinical Kidney Journal 7 (2014) 11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Suter TM and Ewer MS, Cancer drugs and the heart: Importance and management, Eurheartj 34 (2012) 1102–1111. [DOI] [PubMed] [Google Scholar]
- [5].Baskar R, Dai J, Wenlong N, Yeo R, and Yeoh K, Biological response of cancer cells to radiation treatment, Frontiers in Molecular Biosciences 1 (2014) 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Milling L, Zhang Y, and Irvine DJ, Delivering safer immunotherapies for cancer, Adv. Drug Deliv. Rev 114 (2017) 79–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Riley RS, June CH, Langer R, and Mitchell MJ, Delivery technologies for cancer immunotherapy, Nature Reviews Drug Discovery (2019) 175–196. [DOI] [PMC free article] [PubMed]
- [8].Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, and Marson A, CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells, Scientific Reports 7 (2017) 737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Drug and device news, P & T : A Peer-Reviewed Journal for Formulary Management 42 (2017) 608–651. [PMC free article] [PubMed] [Google Scholar]
- [10].Fisher A and Stark A, FDA approval brings first gene therapy to the united states, (2017).
- [11].Kauffman KJ, Webber MJ, and Anderson DG, Materials for non-viral intracellular delivery of messenger RNA therapeutics, J. Controlled Release 240 (2016) 227–234. [DOI] [PubMed] [Google Scholar]
- [12].Moffett HF, Coon ME, Radtke S, Stephan SB, McKnight L, Lambert A, Stoddard BL, Kiem HP, and Stephan MT, Hit-and-run programming of therapeutic cytoreagents using mRNA nanocarriers, Nature Communications 8 (2017) 389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Stewart MP, Sharei A, Ding X, Sahay G, Langer R, and Jensen KF, In vitro and ex vivo strategies for intracellular delivery, Nature 538 (2016) 183–192. [DOI] [PubMed] [Google Scholar]
- [14].McNamara MA, Nair SK, and Holl EK, RNA-based vaccines in cancer immunotherapy, Journal of Immunology Research 2015 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Wang C, Ye Y, Hu Q, Bellotti A, and Gu Z, Tailoring biomaterials for cancer immunotherapy: Emerging trends and future outlook, Adv Mater 29 (2017) 1606036. [DOI] [PubMed] [Google Scholar]
- [16].Riley RS and Day ES, Gold nanoparticle-mediated photothermal therapy: Applications and opportunities for multimodal cancer treatment, WIREs Nanomed Nanobiotechnol 9 (2017) e1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Whitehead KA, Dorkin JR, Vegas AJ, et al. , Degradable lipid nanoparticles with predictable in vivo siRNA delivery activity, Nature Communications 5 (2014) 4277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Kamaly N, Yameen B, Wu J, and Farokhzad OC, Degradable controlled-release polymers and polymeric nanoparticles: Mechanisms of controlling drug release, Chem. Rev 116 (2016) 2602–2663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Large DE, Soucy JR, Hebert J, and Auguste DT, Advances in receptor-mediated, tumor-targeted drug delivery, Adv. Therap 2 (2019) 1800091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Kranz LM, Diken M, Haas H, et al. , Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy, Nature 534 (2016) 396–401. [DOI] [PubMed] [Google Scholar]
- [21].Mitchell MJ, Jain RK, and Langer R, Engineering and physical sciences in oncology: Challenges and opportunities, Nature Reviews Cancer 17 (2017) 659–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Tiptiri-Kourpeti A, Spyridopoulou K, Pappa A, and Chlichlia K, DNA vaccines to attack cancer: Strategies for improving immunogenicity and efficacy, Pharmacol. Ther 165 (2016) 32–49. [DOI] [PubMed] [Google Scholar]
- [23].Kutzler MA and Weiner DB, DNA vaccines: Ready for prime time? Nature Reviews Genetics 9 (2008) 776–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, and Anderson DG, Non-viral vectors for gene-based therapy, Nature Reviews Genetics 15 (2014) 541–555. [DOI] [PubMed] [Google Scholar]
- [25].Chen Y, Groves B, Muscat RA, and Seelig G, DNA nanotechnology from the test tube to the cell, Nature Nanotechnology 10 (2015) 748–760. [DOI] [PubMed] [Google Scholar]
- [26].Shen X, Rajapakse A, Gallazzi F, Junnotula V, Fuchs-Knotts T, Glaser R, and Gates KS, Isotopic labeling experiments that elucidate the mechanism of DNA strand cleavage by the hypoxia-selective antitumor agent 1,2,4-benzotriazine 1,4-di-N-oxide, Chem. Res. Toxicol 27 (2014) 111–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Pack DW, Hoffman AS, Pun S, and Stayton PS, Design and development of polymers for gene delivery, Nature Reviews Drug Discovery 4 (2005) 581–593. [DOI] [PubMed] [Google Scholar]
- [28].Zhang R and Ulery BD, Synthetic vaccine characterization and design, Journal of Bionanoscience 12 (2018) 1–11. [Google Scholar]
- [29].Pan L, Wang Z, Li Y, Xu F, Zhang Q, and Zhang C, Nicking enzyme-controlled toehold regulation for DNA logic circuits, Nanoscale 9 (2017) 18223–18228. [DOI] [PubMed] [Google Scholar]
- [30].Ibraheem D, Elaissari A, and Fessi H, Gene therapy and DNA delivery systems, Int. J. Pharm 459 (2014) 70–83. [DOI] [PubMed] [Google Scholar]
- [31].Park JW, Liposome-based drug delivery in breast cancer treatment, Breast Cancer Research 4 (2002) 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Malam Y, Loizidou M, and Seifalian AM, Liposomes and nanoparticles: Nanosized vehicles for drug delivery in cancer, Trends in Pharmacological Sciences 30 (2009) 592–599. [DOI] [PubMed] [Google Scholar]
- [33].Antonietti M and Förster S, Vesicles and liposomes: A self-assembly principle beyond lipids, Adv Mater 15 (2003) 1323–1333. [Google Scholar]
- [34].Christensen D, Korsholm KS, Andersen P, and Agger EM, Cationic liposomes as vaccine adjuvants, Expert Review of Vaccines 10 (2011) 513–521. [DOI] [PubMed] [Google Scholar]
- [35].Schwendener RA, Liposomes as vaccine delivery systems: A review of the recent advances, Therapeutic Advances in Vaccines 2 (2014) 159–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Allen TM and Cullis PR, Liposomal drug delivery systems: From concept to clinical applications, Adv. Drug Deliv. Rev 65 (2013) 36–48. [DOI] [PubMed] [Google Scholar]
- [37].Chang H and Yeh M, Clinical development of liposome-based drugs: Formulation, characterization, and therapeutic efficacy, International Journal of Nanomedicine 7 (2012) 49–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Bennett CF, Chiang MY, Chan H, Shoemaker JE, and Mirabelli CK, Cationic lipids enhance cellular uptake and activity of phosphorothioate antisense oligonucleotides. Molecular Pharmacology 41 (1992) 1023–1033. [PubMed] [Google Scholar]
- [39].Miller CR, Bondurant B, McLean SD, McGovern KA, and O’Brien DF, Liposome-Cell interactions in vitro: Effect of liposome surface charge on the binding and endocytosis of conventional and sterically stabilized liposomes, Biochemistry 37 (1998) 12875–12883. [DOI] [PubMed] [Google Scholar]
- [40].Fröhlich E, The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles, International Journal of Nanomedicine 7 (2012) 5577–5591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Lv H, Zhang S, Wang B, Cui S, and Yan J, Toxicity of cationic lipids and cationic polymers in gene delivery, Journal of Controlled Release 114 (2006) 100–109. [DOI] [PubMed] [Google Scholar]
- [42].Ma Z, Li J, He F, Wilson A, Pitt B, and Li S, Cationic lipids enhance siRNA-mediated interferon response in mice, Biochemical and Biophysical Research Communications 330 (2005) 755–759. [DOI] [PubMed] [Google Scholar]
- [43].Senior JH, Trimble KR, and Maskiewicz R, Interaction of positively-charged liposomes with blood: Implications for their application in vivo, Biochimica Et Biophysica Acta (BBA) - Biomembranes 1070 (1991) 173–179. [DOI] [PubMed] [Google Scholar]
- [44].Filion MC and Phillips NC, Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells, Biochimica Et Biophysica Acta (BBA) - Biomembranes 1329 (1997) 345–356. [DOI] [PubMed] [Google Scholar]
- [45].Knudsen KB, Northeved H, Kumar EK P, et al. , In vivo toxicity of cationic micelles and liposomes, Nanomedicine: Nanotechnology, Biology and Medicine 11 (2015) 467–477. [DOI] [PubMed] [Google Scholar]
- [46].Jahnová E, Ferenčík M, Nyulassy Š, Devínsky F, and Lacko I, Amphiphilic detergents inhibit production of IgG and IgM by human peripheral blood mononuclear cells, Immunology Letters 39 (1993) 71–75. [DOI] [PubMed] [Google Scholar]
- [47].Dass CR, Biochemical and biophysical characteristics of lipoplexes pertinent to solid tumour gene therapy, International Journal of Pharmaceutics 241 (2002) 1–25. [DOI] [PubMed] [Google Scholar]
- [48].Tousignant JD, Gates AL, Ingram LA, Johnson CL, Nietupski JB, Cheng SH, Eastman SJ, and Scheule RK, Comprehensive analysis of the acute toxicities induced by systemic administration of cationic lipid:Plasmid DNA complexes in mice, Hum. Gene Ther 11 (2000) 2493–2513. [DOI] [PubMed] [Google Scholar]
- [49].Oberli MA, Reichmuth AM, Dorkin JR, Mitchell MJ, Fenton OS, Jaklenec A, Anderson DG, Langer R, and Blankschtein D, Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy, Nano Letters 17 (2017) 1326–1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Love KT, Mahon KP, Levins CG, et al. , Lipid-like materials for low-dose, in vivo gene silencing, Proc. Natl. Acad. Sci. USA 107 (2010) 1864–1869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Akinc A, Zumbuehl A, Goldberg M, et al. A combinatorial library of lipid-like materials for delivery of RNAi therapeutics, Nature Biotechnology 26 (2008) 561–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Schiffelers RM, Woodle MC, and Scaria P, Pharmaceutical prospects for RNA interference, Pharmaceutical Research 21 (2004) 1–7. [DOI] [PubMed] [Google Scholar]
- [53].Siegel DP and Epand RM, The mechanism of lamellar-to-inverted hexagonal phase transitions in phosphatidylethanolamine: Implications for membrane fusion mechanisms, Biophysical Journal 73 (1997) 3089–3111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Li W and Szoka FC, Lipid-based nanoparticles for nucleic acid delivery, Pharm. Res 24 (2007) 438–449. [DOI] [PubMed] [Google Scholar]
- [55].Cheng X and Lee RJ, The role of helper lipids in lipid nanoparticles (LNPs) designed for oligonucleotide delivery, Advanced Drug Delivery Reviews 99 (2016) 129–137. [DOI] [PubMed] [Google Scholar]
- [56].Allen TM and Cullis PR, Liposomal drug delivery systems: From concept to clinical applications, Advanced Drug Delivery Reviews 65 (2013) 36–48. [DOI] [PubMed] [Google Scholar]
- [57].de Jong S, Chikh G, Sekirov L, Raney S, Semple S, Klimuk S, Yuan N, Hope M, Cullis P, and Tam Y, Encapsulation in liposomal nanoparticles enhances the immunostimulatory, adjuvant and anti-tumor activity of subcutaneously administered CpG ODN, Cancer Immunology, Immunotherapy 56 (2007) 1251–1264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Whitehead KA, Langer R, and Anderson DG, Knocking down barriers: Advances in siRNA delivery, Nature Reviews Drug Discovery 8 (2009) 129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Shen X and Corey DR, Chemistry, mechanism and clinical status of antisense oligonucleotides and duplex RNAs, Nucleic Acids Res 46 (2018) 1584–1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].McLendon JM, Joshi SR, Sparks J, Matar M, Fewell JG, Abe K, Oka M, McMurtry IF, and Gerthoffer WT, Lipid nanoparticle delivery of a microRNA-145 inhibitor improves experimental pulmonary hypertension, J. Controlled Release 210 (2015) 67–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Tolstyka ZP, Phillips H, Cortez M, Wu Y, Ingle N, Bell JB, Hackett PB, and Reineke TM, Trehalose-based block copolycations promote polyplex stabilization for lyophilization and in vivo pDNA delivery, ACS Biomater. Sci. Eng 2 (2016) 43–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Olden BR, Cheng Y, Yu JL, and Pun SH, Cationic polymers for non-viral gene delivery to human T cells, J. Controlled Release 282 (2018) 140–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Feng G, Chen H, Li J, Huang Q, Gupte MJ, Liu H, Song Y, and Ge Z, Gene therapy for nucleus pulposus regeneration by heme oxygenase-1 plasmid DNA carried by mixed polyplex micelles with thermo-responsive heterogeneous coronas, Biomaterials 52 (2015) 1–13. [DOI] [PubMed] [Google Scholar]
- [64].Yu J, Xie X, Zheng M, Yu L, Zhang L, Zhao J, Jiang D, and Che X, Fabrication and characterization of nuclear localization signal-conjugated glycol chitosan micelles for improving the nuclear delivery of doxorubicin, International Journal of Nanomedicine 7 (2012) 5079–5090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Tammam SN, Azzazy HME, Breitinger HG, and Lamprecht A, Chitosan nanoparticles for nuclear targeting: The effect of nanoparticle size and nuclear localization sequence density, Mol. Pharmaceutics 12 (2015) 4277–4289. [DOI] [PubMed] [Google Scholar]
- [66].Guerrero-Cázares H, Tzeng SY, Young NP, Abutaleb AO, Quiñones-Hinojosa A, and Green JJ, Biodegradable polymeric nanoparticles show high efficacy and specificity at DNA delivery to human glioblastoma in vitro and in vivo, ACS Nano 8 (2014) 5141–5153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Bishop CJ, Majewski RL, Guiriba TM, Wilson DR, Bhise NS, Quiñones-Hinojosa A, and Green JJ, Quantification of cellular and nuclear uptake rates of polymeric gene delivery nanoparticles and DNA plasmids via flow cytometry, Acta Biomaterialia 37 (2016) 120–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Mangraviti A, Tzeng SY, Kozielski KL, et al. , Polymeric nanoparticles for nonviral gene therapy extend brain tumor survival in vivo, ACS Nano 9 (2015) 1236–1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Xiang SD, Selomulya C, Ho J, Apostolopoulos V, and Plebanski M, Delivery of DNA vaccines: An overview on the use of biodegradable polymeric and magnetic nanoparticles, WIREs Nanomed Nanobiotechnol 2 (2010) 205–218. [DOI] [PubMed] [Google Scholar]
- [70].Minigo G, Scholzen A, Tang CK, Hanley JC, Kalkanidis M, Pietersz GA, Apostolopoulos V, and Plebanski M, Poly-l-lysine-coated nanoparticles: A potent delivery system to enhance DNA vaccine efficacy, Vaccine 25 (2007) 1316–1327. [DOI] [PubMed] [Google Scholar]
- [71].Boussif O, Lezoualc’h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, and Behr JP, A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: Polyethylenimine, Proc. Natl. Acad. Sci. USA 92 (1995) 7297–7301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Varkouhi AK, Scholte M, Storm G, and Haisma HJ, Endosomal escape pathways for delivery of biologicals, J. Controlled Release 151 (2011) 220–228. [DOI] [PubMed] [Google Scholar]
- [73].Seow WY, Liang K, Kurisawa M, and Hauser CAE, Oxidation as a facile strategy to reduce the surface charge and toxicity of polyethyleneimine gene carriers, Biomacromolecules 14 (2013) 2340–2346. [DOI] [PubMed] [Google Scholar]
- [74].Zintchenko A, Philipp A, Dehshahri A, and Wagner E, Simple modifications of branched PEI lead to highly efficient siRNA carriers with low toxicity, Bioconjugate Chem 19 (2008) 1448–1455. [DOI] [PubMed] [Google Scholar]
- [75].Yue Y, Jin F, Deng R, Cai J, Dai Z, Lin MCM, Kung H, Mattebjerg MA, Andresen TL, and Wu C, Revisit complexation between DNA and polyethylenimine — effect of length of free polycationic chains on gene transfection, Journal of Controlled Release 152 (2011) 143–151. [DOI] [PubMed] [Google Scholar]
- [76].Tzeng SY and Green JJ, Polymeric nucleic acid delivery for immunoengineering, Current Opinion in Biomedical Engineering; Molecular and Cellular Engineering: Gene Therapy 7 (2018) 42–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Melamed JR, Kreuzberger NL, Goyal R, and Day ES, Spherical nucleic acid architecture can improve the efficacy of polycation-mediated siRNA delivery, Molecular Therapy - Nucleic Acids 12 (2018) 207–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Jia S, Worth LL, Densmore CL, Xu B, Duan X, and Kleinerman ES, Aerosol gene therapy with PEI: IL-12 eradicates osteosarcoma lung metastases, Clinical Cancer Research 9 (2003) 3462–3468. [PubMed] [Google Scholar]
- [79].Karlsson J, Vaughan HJ, and Green JJ, Biodegradable polymeric nanoparticles for therapeutic cancer treatments, Annu. Rev. Chem. Biomol. Eng 9 (2018) 105–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Green JJ, Langer R, and Anderson DG, A combinatorial polymer library approach yields insight into nonviral gene delivery, Acc. Chem. Res 41 (2008) 749–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Smith TT, Stephan SB, Moffett HF, McKnight LE, Ji W, Reiman D, Bonagofski E, Wohlfahrt ME, Pillai SPS, and Stephan MT, In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers, Nature Nanotechnology 12 (2017) 813–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Tsai SJ, Andorko JI, Zeng X, Gammon JM, and Jewell CM, Polyplex interaction strength as a driver of potency during cancer immunotherapy, Nano Research 11 (2018) 5642–5656. [Google Scholar]
- [83].Wilson DR, Sen R, Sunshine JC, Pardoll DM, Green JJ, and Kim YJ, Biodegradable STING agonist nanoparticles for enhanced cancer immunotherapy, Nanomedicine: Nanotechnology, Biology and Medicine 14 (2018) 237–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Zhang R, Billingsley MM, and Mitchell MJ, Biomaterials for vaccine-based cancer immunotherapy, J. Controlled Release 292 (2018) 256–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Yang B, Jeang J, Yang A, Wu TC, and Hung C, DNA vaccine for cancer immunotherapy, Human Vaccines & Immunotherapeutics 10 (2014) 3153–3164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Zhao G, Steinhagen F, Kinjo T, and Klinman DM, CpG DNA as a vaccine adjuvant AU - bode, christian, Expert Review of Vaccines 10 (2011) 499–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Pardi N, Hogan MJ, Porter FW, and Weissman D, mRNA vaccines — a new era in vaccinology, Nature Reviews Drug Discovery 17 (2018) 261–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Hajj KA and Whitehead KA, Tools for translation: Non-viral materials for therapeutic mRNA delivery, Nature Reviews Materials 2 (2017) 17056. [Google Scholar]
- [89].Weissman D, mRNA transcript therapy, Expert Review of Vaccines 14 (2015) 265–281. [DOI] [PubMed] [Google Scholar]
- [90].Kay MA, State-of-the-art gene-based therapies: The road ahead, Nature Reviews Genetics 12 (2011) 316–328. [DOI] [PubMed] [Google Scholar]
- [91].Kauffman KJ, Dorkin JR, Yang JH, Heartlein MW, DeRosa F, Mir FF, Fenton OS, and Anderson DG, Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs, Nano Letters 15 (2015) 7300–7306. [DOI] [PubMed] [Google Scholar]
- [92].Green JJ, Langer R, and Anderson DG, A combinatorial polymer library approach yields insight into nonviral gene delivery, Acc. Chem. Res 41 (2008) 749–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Zugates GT, Peng W, Zumbuehl A, Jhunjhunwala S, Huang Y, Langer R, Sawicki JA, and Anderson DG, Rapid optimization of gene delivery by parallel end-modification of poly(β-amino ester)s, Molecular Therapy 15 (2007) 1306–1312. [DOI] [PubMed] [Google Scholar]
- [94].Patel AK, Kaczmarek JC, Bose S, Kauffman KJ, Mir F, Heartlein MW, DeRosa F, Langer R, and Anderson DG, Inhaled nanoformulated mRNA polyplexes for protein production in lung epithelium, Adv Mater 0 (2019) 1805116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Kaczmarek JC, Kauffman KJ, Fenton OS, Sadtler K, Patel AK, Heartlein MW, DeRosa F, and Anderson DG, Optimization of a degradable Polymer–Lipid nanoparticle for potent systemic delivery of mRNA to the lung endothelium and immune cells, Nano Lett 18 (2018) 6449–6454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Kaczmarek JC, Patel AK, Kauffman KJ, Fenton OS, Webber MJ, Heartlein MW, DeRosa F, and Anderson DG, Polymer–Lipid nanoparticles for systemic delivery of mRNA to the lungs, Angewandte Chemie International Edition 55 (2016) 13808–13812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Su X, Fricke J, Kavanagh DG, and Irvine DJ, In vitro and in vivo mRNA delivery using lipid-enveloped pH-responsive polymer nanoparticles, Molecular Pharmaceutics 8 (2011) 774–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Nasal vaccination: A non-invasive vaccine delivery method that holds great promise for the future, Advanced Drug Delivery Reviews 51 (2001) 1–3. [PubMed] [Google Scholar]
- [99].Holmgren J and Czerkinsky C, Mucosal immunity and vaccines, Nat. Med 11 (2005) 45. [DOI] [PubMed] [Google Scholar]
- [100].June CH, Riddell SR, and Schumacher TN, Adoptive cellular therapy: A race to the finish line, Science Translational Medicine 7 (2015) 280ps7. [DOI] [PubMed] [Google Scholar]
- [101].Mockey M, Bourseau E, Chandrashekhar V, Chaudhuri A, Lafosse S, Le Cam E, Quesniaux VFJ, Ryffel B, Pichon C, and Midoux P, mRNA-based cancer vaccine: Prevention of B16 melanoma progression and metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes, Cancer Gene Therapy 14 (2007) 802–814. [DOI] [PubMed] [Google Scholar]
- [102].Wu Y, Liang D, Wang Y, Bai M, Tang W, Bao S, Yan Z, Li D, and Li J, Correction of a genetic disease in mouse via use of CRISPR-Cas9, Cell Stem Cell 13 (2013) 659–662. [DOI] [PubMed] [Google Scholar]
- [103].Hsu P, Lander E, and Zhang F, Development and applications of CRISPR-Cas9 for genome engineering, Cell 157 (2014) 1262–1278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Heckl D, Kowalczyk MS, Yudovich D, Belizaire R, Puram RV, McConkey ME, Thielke A, Aster JC, Regev A, and Ebert BL, Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing, Nat. Biotechnol 32 (2014) 941–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, Watanabe T, Kanai T, and Sato T, Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids, Nat. Med 21 (2015) 256–262. [DOI] [PubMed] [Google Scholar]
- [106].Platt R, Chen S, Zhou Y, et al. , CRISPR-Cas9 knockin mice for genome editing and cancer modeling, Cell 159 (2014) 440–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Manguso RT, Pope HW, Zimmer MD, et al. , In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target, Nature 547 (2017) 413–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Eyquem J, Mansilla-Soto J, Giavridis T, d. S. van, M. Hamieh, K. M. Cunanan, A. Odak, M. Gönen, and M. Sadelain, Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection, Nature 543 (2017) 113–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Su S, Hu B, Shao J, et al. , CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients, Scientific Reports 6 (2016) 20070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Schumann K, Lin S, Boyer E, et al. , Generation of knock-in primary human T cells using Cas9 ribonucleoproteins, Proc. Natl. Acad. Sci. USA 112 (2015) 10437–10442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Ren J, Liu X, Fang C, Jiang S, June CH, and Zhao Y, Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition, Clin. Cancer Res 23 (2017) 2255–2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Yin H, Kauffman KJ, and Anderson DG, Delivery technologies for genome editing, Nature Reviews Drug Discovery 16 (2017) 387–399. [DOI] [PubMed] [Google Scholar]
- [113].Liu C, Zhang L, Liu H, and Cheng K, Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications, Journal of Controlled Release 266 (2017) 17–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Langer R and Peppas NA, Advances in biomaterials, drug delivery, and bionanotechnology, AIChE J 49 (2003) 2990–3006. [Google Scholar]
- [115].Almeida AJ and Souto E, Solid lipid nanoparticles as a drug delivery system for peptides and proteins, Adv. Drug Deliv. Rev 59 (2007) 478–490. [DOI] [PubMed] [Google Scholar]
- [116].Torchilin VP and Lukyanov AN, Peptide and protein drug delivery to and into tumors: Challenges and solutions, Drug Discov. Today 8 (2003) 259–266. [DOI] [PubMed] [Google Scholar]
- [117].Cronican JJ, Thompson DB, Beier KT, McNaughton BR, Cepko CL, and Liu DR, Potent delivery of functional proteins into mammalian cells in vitro and in vivo using a supercharged protein, ACS Chem. Biol 5 (2010) 747–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Thompson D, Villaseñor R, Dorr B, Zerial M, and Liu D, Cellular uptake mechanisms and endosomal trafficking of supercharged proteins, Chemistry & Biology 19 (2012) 831–843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Zuris JA, Thompson DB, Shu Y, Guilinger JP, Bessen JL, Hu JH, Maeder ML, Joung JK, Chen Z, and Liu DR, Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo, Nat. Biotechnol 33 (2014) 73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Gao X, Tao Y, Lamas V, et al. , Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents, Nature 553 (2017) 217–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Wang M, Zuris JA, Meng F, et al. , Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles, Proc. Natl. Acad. Sci. USA 113 (2016) 2868–2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Charlesworth CT, Deshpande PS, Dever DP, et al. , Identification of preexisting adaptive immunity to Cas9 proteins in humans, Nat. Med 25 (2019) 249–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [123].Wang H, Li M, Lee CM, Chakraborty S, Kim H, Bao G, and Leong KW, CRISPR/Cas9-based genome editing for disease modeling and therapy: Challenges and opportunities for nonviral delivery, Chem. Rev 117 (2017) 9874–9906. [DOI] [PubMed] [Google Scholar]
- [124].Yin H, Song C, Dorkin JR, et al. , Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo, Nat. Biotechnol 34 (2016) 328–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [125].Miller JB, Zhang S, Kos P, Xiong H, Zhou K, Perelman SS, Zhu H, and Siegwart DJ, Non-viral CRISPR/cas gene editing in vitro and in vivo enabled by synthetic nanoparticle co-delivery of Cas9 mRNA and sgRNA, Angew. Chem 129 (2017) 1079–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Finn JD, Smith AR, Patel MC, et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent In Vivo genome editing, Cell Reports 22 (2018) 2227–2235. [DOI] [PubMed] [Google Scholar]
- [127].Bonifant CL, Jackson HJ, Brentjens RJ, and Curran KJ, Toxicity and management in CAR T-cell therapy, Molecular Therapy - Oncolytics 3 (2016) 16011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [128].Graham C, Jozwik A, Pepper A, and Benjamin R, Allogeneic CAR-T cells: More than ease of access? Cells 7 (2018) e155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Lim WA and June CH, The principles of engineering immune cells to treat cancer, Cell 168 (2017) 724–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Petersdorf EW, Hansen JA, Martin PJ, Woolfrey A, Malkki M, Gooley T, Storer B, Mickelson E, Smith A, and Anasetti C, Major-histocompatibility-complex class I alleles and antigens in hematopoietic-cell transplantation, N. Engl. J. Med 345 (2001) 1794–1800. [DOI] [PubMed] [Google Scholar]
- [131].Liu X and Zhao Y, CRISPR/Cas9 genome editing: Fueling the revolution in cancer immunotherapy, Current Research in Translational Medicine 66 (2018) 39–42. [DOI] [PubMed] [Google Scholar]
- [132].Keir ME, Butte MJ, Freeman GJ, and Sharpe AH, PD-1 and its ligands in tolerance and immunity, Annu. Rev. Immunol 26 (2008) 677–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [133].Luo Y, Xu C, Li H, Cao Z, Liu J, Wang J, Du X, Yang X, Gu Z, and Wang J, Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles, ACS Nano 12 (2018) 994–1005. [DOI] [PubMed] [Google Scholar]
- [134].Ramkhelawon B, Hennessy EJ, Ménager M, et al. , Netrin-1 promotes adipose tissue macrophage retention and insulin resistance in obesity, Nat. Med 20 (2014) 377–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [135].Li M, Fan Y, Chen Z, Luo Y, Wang Y, Lian Z, Xu C, and Wang J, Optimized nanoparticle-mediated delivery of CRISPR-Cas9 system for B cell intervention, Nano Research 11 (2018) 6270–6282. [Google Scholar]
- [136].Schmid D, Park CG, Hartl CA, et al. , T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity, Nature Communications 8 (2017) 1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [137].Song W, Shen L, Wang Y, Liu Q, Goodwin TJ, Li J, Dorosheva O, Liu T, Liu R, and Huang L, Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap, Nature Communications 9 (2018) 2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [138].Akinc A, Querbes W, De S, et al. , Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms, Molecular Therapy 18 (2010) 1357–1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Guo Y, Chen W, Wang W, Shen J, Guo R, Gong F, Lin S, Cheng D, Chen G, and Shuai X, Simultaneous diagnosis and gene therapy of immuno-rejection in rat allogeneic heart transplantation model using a T-cell-targeted theranostic nanosystem, ACS Nano 6 (2012) 10646–10657. [DOI] [PubMed] [Google Scholar]
- [140].Kudo-Saito C, Shirako H, Takeuchi T, and Kawakami Y, Cancer metastasis is accelerated through immunosuppression during snail-induced EMT of cancer cells, Cancer Cell 15 (2009) 195–206. [DOI] [PubMed] [Google Scholar]
- [141].Mellman I, Coukos G, and Dranoff G, Cancer immunotherapy comes of age, Nature 480 (2011) 480–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Garber K, Alnylam launches era of RNAi drugs, (2018). [DOI] [PubMed]