Next Article in Journal
Genetic and Epigenetic Biomarkers Linking Alzheimer’s Disease and Age-Related Macular Degeneration
Previous Article in Journal
Endothelial Protection by Sodium-Glucose Cotransporter 2 Inhibitors: A Literature Review of In Vitro and In Vivo Studies
Previous Article in Special Issue
Glycan Structures in Osteosarcoma as Targets for Lectin-Based Chimeric Antigen Receptor Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Coding RNA in Tumor Cells and Tumor-Associated Myeloid Cells—Function and Therapeutic Potential

by
Amanda Katharina Binder
1,2,3,4,
Franziska Bremm
1,2,3,4,
Jan Dörrie
1,2,3,4,† and
Niels Schaft
1,2,3,4,*,†
1
Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
2
Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
3
Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
4
Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(13), 7275; https://doi.org/10.3390/ijms25137275 (registering DOI)
Submission received: 6 May 2024 / Revised: 19 June 2024 / Accepted: 29 June 2024 / Published: 2 July 2024
(This article belongs to the Special Issue Molecular Advances in Cancer Immunotherapy)

Abstract

:
The RNA world is wide, and besides mRNA, there is a variety of other RNA types, such as non-coding (nc)RNAs, which harbor various intracellular regulatory functions. This review focuses on small interfering (si)RNA and micro (mi)RNA, which form a complex network regulating mRNA translation and, consequently, gene expression. In fact, these RNAs are critically involved in the function and phenotype of all cells in the human body, including malignant cells. In cancer, the two main targets for therapy are dysregulated cancer cells and dysfunctional immune cells. To exploit the potential of mi- or siRNA therapeutics in cancer therapy, a profound understanding of the regulatory mechanisms of RNAs and following targeted intervention is needed to re-program cancer cells and immune cell functions in vivo. The first part focuses on the function of less well-known RNAs, including siRNA and miRNA, and presents RNA-based technologies. In the second part, the therapeutic potential of these technologies in treating cancer is discussed, with particular attention on manipulating tumor-associated immune cells, especially tumor-associated myeloid cells.

1. Introduction

1.1. RNA Is More Than a Blueprint for Proteins

Proteins are encoded by DNA, yet DNA is not directly translated into polypeptides. First, the information is transcribed into RNA, which functions as a vehicle to transport the information from the nucleus to the production site. The importance of RNA culminates in the RNA world hypothesis, which suggests that RNA precedes DNA as genetic material and proteins as biological enzymes, thus forming the archetype molecule of life [1]. In recent years, the therapeutical application of (m)RNA became feasible since biochemical modifications and new shuttling systems allowed higher stability of the RNA, lower immunogenicity, and a protected delivery within the human body. The field of mRNA-based immunization became very popular due to the coronavirus pandemic.
However, besides mRNA and its functions, several other RNA types have been identified in plants, bacteria, and animals [2]. RNA-interactome studies revealed that these RNAs are able to interact with other RNAs, DNA, proteins, and peptides and exert crucial functions in the regulation of replication, transcription, and translation [3]. In addition to these ubiquitous functions, the non-coding RNAs are central in multiple cellular defense systems against viruses (e.g., double-stranded RNA of viral origin) and mobile genetic elements (e.g., transposons, plasmids, and phages) [3,4]. Hence, many functional principles involving non-coding (nc)RNA were first observed and characterized in the context of pathogen defense but can be exploited for new therapeutic approaches. A part of these defense systems utilizes small guide RNAs that recognize aberrant transcripts and mediate their inactivation [5]. This mechanism of action is termed RNA interference (RNAi) and subsumes several molecular mechanisms that use small guide RNA fragments to target specific nucleic acid sequences. The recognition of the target is mediated via complementary base pairing, which requires a reverse complementary target sequence and is therefore referred to as antisense regulation [5,6].

1.2. A Brief History of RNA-Based Regulation

Historically, gene regulation by RNAs was already proposed by Francois Jacob and Jacques Monod in 1961 [7] but could not be proven experimentally at this time. In the 1980s, antisense RNA control was thoroughly studied in bacteria and phages [8]. In the 1990s, a key experiment with eukaryotic RNAi was conducted by Fire and Mellow in Caenorhabditis elegans, showing that artificially introduced short antisense RNA was able to down-regulate the expression of developmental genes on the translational level by binding to the mRNA of the target gene [9,10,11]. This work won the Nobel prize in 2006, and RNAi was judged to be “a fundamental mechanism for controlling the flow of genetic information in cells” [12].
In 2020, another system that utilizes short non-coding RNA in a similar fashion to RNAi won the Nobel prize: the nucleic acid-guided defense systems of the bacterial adaptive immune system called CRISPR/Cas [13,14]. In prokaryotes, the clusters of regulatory interspersed short palindromic repeats (CRISPR) are elements in the original bacterial DNA, and the sequences between the repeats represent genetic information from previous invaders that integrated into the bacterial DNA. Upon transcription of the CRISPR DNA elements, CRISPR-RNA is formed, which is complementary to the original pathogenic genetic material, and is thereby able to recognize the pathogens with this sequence upon repeated intrusion. Since this system is a defense mechanism, the binding of the CRISPR-RNA initiates the cleavage of the foreign genetic material by endonucleases of the Cas family [15,16,17,18].
Evolutionarily, the adaptive immunity in eukaryotes was mainly taken over by protein-based recognition molecules, while RNAi still remains a central gene regulatory mechanism in eukaryotes [5,19]. In the following section, a brief overview of eukaryotic RNA forms and their functions is given, with a special focus on endogenous sequence-specific gene silencing on the post-transcriptional level.

2. RNAs in Eukaryotes

Endogenously expressed RNAs in eukaryotes can be divided into different subclasses based on their biogenesis, general characteristics, and functions. The most prominent subdivision distinguishes protein-coding RNAs (mRNAs) and non-protein coding RNAs (ncRNAs) [20,21].
mRNAs with sizes up to several kilobases (kb) are structurally well-defined by being composed of the protein-coding region for translation as well as 3′ and 5′ untranslated regions (UTRs). First, a 7-methylguanosin is added as a cap at the 5′UTR that protects the mRNA from degradation. At the end of the 3′UTR, the mRNA is polyadenylated, thus forming a so-called Poly(A) tail that enhances the stability and is involved in the transport of the mRNA [22]. mRNAs are processed in the nucleus, after which the matured mRNAs are shuttled into the cytoplasm. Finally, the translation into a protein occurs at ribosomes [23].
Ribosomal RNA (rRNA) and transfer RNA (tRNA) are functional RNAs that are highly abundant and form their own subclasses. Both are constitutively expressed and harbor housekeeping functions involved in general protein synthesis at the ribosomes. Although they do not encode proteins, they are not considered part of the group of ncRNAs. Their biogenesis and functions are reviewed extensively elsewhere [24,25,26].
In contrast to mRNA, rRNA, and tRNA, ncRNAs comprise a diverse group with less well-defined subgroups and continuously new discoveries of RNA types. However, the ncRNAs can be broadly grouped by their size [27,28]. The long non-coding RNAs (lncRNAs) are, per definition, larger than 200 nucleotides (nt), often, but not always, capped and polyadenylated, and are present in the nucleus and the cytoplasm [29,30,31]. This group includes, for example, enhancer RNAs (eRNA) which mediate mRNA transcription [32,33]. Their biological function is not yet fully agreed upon and is extensively reviewed elsewhere [34,35]. Another example of this size group is circular RNAs (circRNA), which are a product of non-canonical splicing and have different regulatory functions, including the regulation of other RNAs and proteins [36].
The second group is small ncRNAs with sizes smaller than 200 nt. One representative of this group is P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNA). Especially in germ-line cells, this functional complex of protein and RNA protects the genome from transposons by chromatin modifications that repress transcription [37,38]. Other small ncRNAs, such as small nuclear RNA (snRNA), act in the cell nucleus or nucleoli that are involved in the splicing of pre-mRNA [39,40], and small nucleolar RNA (snoRNA) that mediate the processing of other RNAs, e.g., of rRNA [41]. Lastly, micro-RNAs (miRNA) and small-interfering RNAs (siRNA), both 19–25 nt long, act on the translation of mRNA and are therefore the two main RNA types involved in post-transcriptional gene silencing [19,42,43,44]. Since they play a prominent role in gene regulation and their regulatory function holds great potential in therapeutic application, both are discussed in detail below.

2.1. Biogenesis of Micro-RNAs and Small-Interfering RNAs

The discrimination between miRNA and siRNA is difficult since both have similar mechanisms of action. So far, the main distinction is made by their origin and biogenesis. miRNAs, as endogenous regulators of mRNA translation, are always genome-derived, originating from introns of protein-coding genome sequences, from introns or exons of ncRNA genome sequences, or from independent transcriptional units [45]. The biogenesis of miRNAs via the canonical pathway starts with the transcription of the above-mentioned gene loci by RNA polymerase II/III, producing the so-called primary miRNA (pri-miRNA) present in the nucleus [46]. Structurally, this pri-miRNA forms a hairpin-shaped RNA construct with complementary-bound double-stranded RNA (dsRNA) parts and an apical loop [47]. Still in the nucleus, the pri-miRNA is cleaved into one or more precursor miRNA (pre-miRNA) by the microprocessor complex composed of the enzymes Drosha and the co-factor Pasha [48,49] (Figure 1). Thereafter, the pre-miRNA is shuttled into the cytoplasm via Exportin 5 [50]. In the cytoplasm, the double-stranded part of the pre-miRNA binds to a ribonuclease III enzyme called Dicer [51], which, in turn, cleaves the pre-miRNA into a miRNA duplex composed of the guide miRNA single-strand and a complementary bound passenger strand termed miRNA* [52,53]. The non-canonical pathway utilizes different combinations of enzymes apart from Drosha and Dicer, which is reviewed by Ha et al. [46].
In contrast to miRNAs, siRNAs are primarily of exogenous origin, deriving from double-stranded RNA transcripts of viruses or transgenes. Those can originate from exogenous sequences already integrated into the host genome, termed endo-siRNA, or from ex-genomic sources (exo-siRNA), including pathogens or artificial exogenously transferred dsRNA in experimental settings [45,54,55,56]. For the endo-siRNA, the resulting RNA transcripts contain complementary double-stranded sequences and do not undergo extensive processing steps in the nucleus but are rapidly shuttled into the cytoplasm [11,45,57]. For the exo-genomic siRNA, the dsRNA strands are solely present in the cytoplasm. Without regard to their origin, these dsRNAs are recognized by Dicer and enter the RNAi pathway [45,58]. The resulting 21–25 nt long RNA molecules are termed siRNA duplexes. Similar to the miRNA, a siRNA duplex consists of a guide strand and a passenger strand. It should be mentioned that the term siRNA often refers only to synthetic components since it is prominently used in experimental settings, although, as explained above, this must not always be the case.
Figure 1. Overview of the mechanisms of action of naturally occurring RNAi pathways and therapeutic approaches intervening with the endogenous RNA interference (RNAi) pathway. (a) micro-RNA (miRNA) pathway. In the nucleus, the primary miRNA (pri-miRNA) is transcribed and processed by the enzyme Drosha and its co-factor Pasha into precursor miRNA (pre-miRNA). The pre-miRNA is shuttled into the cytoplasm and recognized by Dicer as double-stranded RNA (dsRNA) transcript. Dicer cleaves the pre-miRNA into the short miRNA duplex, which is recognized by Argonaute 2 (Ago2), which directs the binding to the mRNA targets and mediates the cleavage. (b) endo-siRNA. The dsRNA transcript originates from the genome in the nucleus and is shuttled into the cytoplasm, where it is recognized by Dicer. Dicer cleaves the long RNA transcript into the short small-interfering RNA (siRNA) duplex, which enters the RNAi pathways as described for miRNAs. (c) exo-siRNA pathway. The dsRNA transcript can be pathogen-derived (e.g., viruses) or therapeutically introduced into the cells. The dsRNA remains in the cytoplasm and is directly recognized by Dicer. (d) miRNA mimics. The miRNA mimics with the same sequence as the endogenous miRNA are recognized by Ago2 and direct the cleavage of the referring target mRNA. (e) Anti-miRs complementarily bind to the guide strand of endogenous miRNA, thus preventing binding to the target mRNA and subsequent cleavage. (f) miRNA sponges catch miRNAs by binding the seed region, also preventing the cleavage of the target mRNA. (g) ASOs. Anti-sense oligos (ASOs) have different mechanisms of action. In the cytoplasm, they can direct the cleavage of target mRNA via RNAi but can also inhibit the mRNA translation via steric hindrance at the ribosomes. The alternative mechanisms of action of ASOs within the nucleus are depicted in the following Figure in Section 3.1.
Figure 1. Overview of the mechanisms of action of naturally occurring RNAi pathways and therapeutic approaches intervening with the endogenous RNA interference (RNAi) pathway. (a) micro-RNA (miRNA) pathway. In the nucleus, the primary miRNA (pri-miRNA) is transcribed and processed by the enzyme Drosha and its co-factor Pasha into precursor miRNA (pre-miRNA). The pre-miRNA is shuttled into the cytoplasm and recognized by Dicer as double-stranded RNA (dsRNA) transcript. Dicer cleaves the pre-miRNA into the short miRNA duplex, which is recognized by Argonaute 2 (Ago2), which directs the binding to the mRNA targets and mediates the cleavage. (b) endo-siRNA. The dsRNA transcript originates from the genome in the nucleus and is shuttled into the cytoplasm, where it is recognized by Dicer. Dicer cleaves the long RNA transcript into the short small-interfering RNA (siRNA) duplex, which enters the RNAi pathways as described for miRNAs. (c) exo-siRNA pathway. The dsRNA transcript can be pathogen-derived (e.g., viruses) or therapeutically introduced into the cells. The dsRNA remains in the cytoplasm and is directly recognized by Dicer. (d) miRNA mimics. The miRNA mimics with the same sequence as the endogenous miRNA are recognized by Ago2 and direct the cleavage of the referring target mRNA. (e) Anti-miRs complementarily bind to the guide strand of endogenous miRNA, thus preventing binding to the target mRNA and subsequent cleavage. (f) miRNA sponges catch miRNAs by binding the seed region, also preventing the cleavage of the target mRNA. (g) ASOs. Anti-sense oligos (ASOs) have different mechanisms of action. In the cytoplasm, they can direct the cleavage of target mRNA via RNAi but can also inhibit the mRNA translation via steric hindrance at the ribosomes. The alternative mechanisms of action of ASOs within the nucleus are depicted in the following Figure in Section 3.1.
Ijms 25 07275 g001

2.2. Micro-RNA and Small-Interfering RNA-Mediated Gene Silencing

miRNAs and siRNAs suppress gene expression via the RNAi pathway, which requires an effector protein complex termed RNA-induced silencing complex (RISC) [59,60] (Figure 1). In the process, the short miRNA or siRNA duplex binds to the RNase H enzyme called Argonaute 2 (Ago2), the duplex gets unwound, and only the guide strand remains at the Ago protein, forming a miRISC or siRISC, while the respective passenger strand is degraded. In both cases, the retained miRNA or siRNA single-strands act as specific guide elements of the complex. Structurally, the recognition of the target sequence is mediated by the seed region, a heptameric (7 nt) sequence within the miRNA or siRNA [61,62,63]. The actual binding occurs via Watson–Crick base pairing to complementary mRNA sequences, mostly located in the 3′UTR of target mRNAs [52]. Upon a high level of complementarity, the target mRNA is cleaved directly [64,65,66], while loose binding, resulting from a partial mismatch, only leads to repression of mRNA translation [67,68,69,70]. miRNAs are able to act via both binding modalities, while siRNAs carry out their function upon complete complementary binding only [70,71].

2.3. Therapeutic Potential of Micro-RNA and Small-Interfering RNA

As outlined above, miRNAs and siRNAs are critical regulators of eukaryotic gene expression, thereby directing developmental processes, cell functions, and physiological processes [72]. In line with that, approximately 60% of the human protein-coding genes contain conserved miRNA-binding sites, implying that those miRNAs can control the expression of the majority of protein-coding genes [73,74]. Up to now, nearly 2700 identified human miRNAs are registered in the open data base miRBase (accessed March 2024). Moreover, dysregulated miRNAs or whole miRNA families are associated with the pathogenesis of human diseases, including CNS disorders, cardiovascular and metabolic diseases, and cancer [72]. Consequently, therapeutics that intervene in the miRNA regulation network or exploit siRNA functions are a powerful tool in several diseases.

3. RNA Constructs in Therapeutic Approaches

Upon using RNAs as therapeutic agents in vivo, three major challenges arise: (i) poor in vivo stability of RNA, (ii) delivery into the right tissue and through the cell membrane, and (iii) off-tissue effects caused by the binding to the target mRNA or to mRNAs with similar sequences in other cells and tissues. However, with ongoing improvement in all three fields, RNAs as posttranscriptional regulators are the subject of basic and clinical research [75,76,77].

3.1. Anti-Sense Oligonucleotides

Anti-sense-oligonucleotides (ASOs), also known as anti-sense therapy, were the first therapeutic approach exploiting complementary nucleic acid strands to interfere with gene expression [78]. ASOs are 12–25 nucleotides long oligomers of single-stranded DNA or RNA [79] (Figure 1). These chemically synthesized oligomers are designed to bind a specific nucleic acid sequence, such as mRNA or its nuclear precursor (pre-mRNA), via complementary base-pairing [80,81]. DNA-based ASOs are able to enter the nucleus of cells and form a DNA-RNA heteroduplex, leading to RNA degradation by activating RNase H, thereby preventing the expression of the respective protein [82] (Figure 2). Further, ASOs can induce altered splicing of pre-mRNA in the nucleus by mediating exon skipping or exon retention [83,84] (Figure 2). In the cytoplasm, DNA- and RNA-based ASOs act via steric hindrance, thus inhibiting translation of the mRNA at the ribosomes [85], as well as inducing mRNA degradation via RNAi [84] (Figure 1). However, a remaining problem of antisense therapy with single-stranded ASOs is chemical instability, low potency, and strong side effects due to the unwanted repression of mRNAs with similar sequences [79,86].

3.2. Small-Interfering RNA

Synthetic siRNA forms are a more specific and potent alternative to ASOs. The siRNA is introduced as a more stable double strand and exploits the cells’ intrinsic RNAi pathway, resulting in a sequence-specific inhibition of mRNA expression. Since siRNAs need to bind completely complementarily to exert mRNA degradation, the off-target effects are strongly diminished compared to ASOs [78] (Figure 1).

3.3. Micro-RNA

The above-described anti-sense therapy can be exploited within the complex miRNA network. By utilizing or targeting miRNAs therapeutically, the regulation of the mRNA expression can be altered in several manners. Firstly, miRNA replacement therapy can be used to enhance endogenous miRNA activity by the addition of synthetic miRNAs that mimic endogenous miRNAs (miRNA mimics). Consequently, the gene expression can be further inhibited, leading to a more effective knockdown in the expression of the affected protein. In contrast to that, synthetic single-strand RNAs acting as miRNA antagonists (termed antagomirs or anti-miRs) can be introduced. These bind to the endogenous mature miRNA and inhibit the miRNA-mediated repression of gene expression (Figure 1). Hence, gene expression can be restored [72,87].
The latter approach is exceeded with miRNA sponges [88]. The sponge RNA can be delivered via a transgene encoded by a viral vector. These transcripts are expressed from strong promoters and contain multiple complementary binding sites only to the heptameric seed regions of the miRNAs of interest (see Section 2.2). Since seed regions are conserved, whole miRNA seed region families can be functionally blocked; thereby, the gene expression of several genes can be rescued [88,89] (Figure 1).

3.4. Micro-RNAs and Small-Interfering RNAs in Clinical Use

In 2018, the FDA’s approval of the first siRNA drug called Patisiran marked a new era of RNAi therapeutics. Patisiran was approved for treating the rare and lethal genetic disease hereditary transthyretin (TTR) amyloidosis. From a mutated allele, defective, mis-folded TTR proteins form, causing severe polyneuropathy and cardiac symptoms. Patisiran binds to the mutated mRNA, causing its degradation and preventing the formation of defective proteins [90,91]. Clinical data showed that seventy percent of the patients receiving Patisiran reached a stable or improved neuropathological stage compared to the placebo group, while side effects were balanced between the groups. However, it should be mentioned that atrioventricular heart blockade as a severe side effect in the Patisiran group was observed [92]. For the treatment of genetic diseases, the FDA approved the three siRNA active agents, Givosiran, Lumasiran, and Inclisiran, in 2019, 2020, and 2021, respectively [93]. Givosiran is a medication for acute hepatic porphyria [94]. Lumasiran was developed as a treatment for primary hyperoxaluria type 1 [95]. Inclisiran was approved for the treatment of familial hypercholesterolemia and reduces the low-density lipoprotein cholesterol levels [96].
Next to these rare genetic diseases, several new siRNA and miRNA therapeutics are in clinical trials targeting a variety of diseases, including cancer. Table 1 subsumes all miRNA and siRNA therapeutics for cancer therapy recently tested in clinical trials (information from ClinicalTrials.gov, [97]). Of note, all approaches listed below have in common the fact that cancer cells are the primary target of miRNAs and siRNAs. Several trials use siRNA to target a tumorigenic driver directly. The drug siG12D-LODER™ targets the mutated form of Kirsten rat sarcoma virus protein (KRAS) called KRASD12G, which is a hallmark of several cancer types, including pancreatic cancer. KRAS naturally induces survival and proliferation of cells, which is exploited by cancer cells with KRAS mutations [98]. The therapeutic inhibition of KRAS expression abrogated this cancer-promoting effect. In a phase I study, it has been shown that the siG12D-LODER™ treatment led to stable disease or partial response in patients with locally advanced pancreatic cancer [99], which then proceeded to a phase II study with unknown status. The clinical phase I trial NCT03608631 also targets KRASD12G using siRNA shuttled in exosomes [100]. In preclinical studies conducted in mice, the treatment reduced KRASD12G mRNA levels, which resulted in suppressed cancer cell proliferation and an overall increased lifespan [100,101,102]. Another siRNA therapeutic, Atu027, targets the protein kinase N3 (PKN3), which is crucial for cell proliferation and migration. In liver cancer, the inhibition of PKN3 showed a significant reduction in cell growth and metastasis formation in non-human primates [103]. This was confirmed in a phase I study conducted in patients with advanced solid tumors who experienced either a stabilization of the disease or a complete or partial regression of metastases after treatment with Atu027 [104]. The siRNA therapeutic TKM-080301 influences the Polo-like kinase (PLK1), which has been shown to be overexpressed in hepatocellular carcinoma (HCC). The knockdown of PLK1 by TKM-080301 reduced cell proliferation in patients with HCC [105]. The drug STP705 is a two-component product containing siRNAs directed against transforming growth factor β (TGFβ) and cyclooxygenase-2 (COX-2). TGFβ released by cancer cells inhibits the function of T cells in the tumor microenvironment (TME), and COX-2 acts as a proliferative mediator. In a human cholangiocarcinoma xenograft tumor model in nude mice, STP705 treatment reduced the immune inhibitory effect of the cancer cells and consequently inhibited tumor growth [106]. Little information is available on the results of the phase I clinical study of STP705 (NCT04293679); however, a phase II clinical study is already ongoing (NCT04844983). The therapeutic agent siRNA-EphA2-DOPC (EPHARNA) targets the expression of the ephrin type-A receptor 2 (EphA2), which is overexpressed in human cancers, including breast, prostate, lung, ovarian, endometrial, and pancreatic carcinomas. It has been shown that this receptor is involved in proliferation, survival, migration, invasion, and angiogenesis and, thereby, is associated with adverse outcomes in cancer patients. In turn, the administration of siRNA against EphA2 shuttled in 1,2-Dioleoyl-sn-Glycero-3-Phosphatidylcholine (DOPC) nanoliposomal particles down-regulated the EphA2 expression and decreased tumor cell growth shown in a murine ovarian cancer model [107]. In 2017, preclinical mammalian safety studies were conducted with EPHARNA [108], followed by the start of a clinical study phase I (ongoing, NCT01591356). The latest clinical trial (NCT06424301) targets refractory retinoblastoma utilizing siRNA against nudix hydrolase 21 modulators (NUDT21). As central factor in alternative polyadenylation, NUDT21 induces 3′UTR shortening of mRNAs often observed in cancer cells to avoid miRNA repression of oncogenes. By inhibiting NUDT21, cancer cell proliferation is inhibited and apoptosis induced [109].
In contrast to siRNA, which usually has a clearly defined target, one miRNA can suppress several targets, and miRNAs are part of a complex regulatory network. While they are frequently utilized in clinical trials as prognostic factors or for disease monitoring, the list of trials using miRNA-based therapeutics is currently short. The first clinical trial with a miRNA-based therapeutic tested MRX34, which is a miR-34a mimic. miR-34a is a naturally occurring tumor suppressor that is lost or expressed at reduced levels in a broad range of tumor types. miR-34a is involved in the down-regulation of the expression of over 30 oncogenes, including myelocytomatosis oncogene (MYC), platelet derived growth factor receptor α (PDGFR-α), cyclin-dependent kinase (CDK) 4/6, and B-cell lymphoma 2 (BCL2) [110]. The first phase I study with MRX34 (NCT01829971) (Table 1) was terminated because of several drug-related severe adverse events, comprising systemic enterocolitis, colitis/pneumonitis, systemic inflammatory response syndrome, and cytokine release syndrome with respiratory failure, and hepatic failure, of which the latter four led to death. The following MRX34 study (NCT02862145) (Table 1) never recruited patients because of the severe side effects in the previous study [110]. Another approach is TargmiRs, which is a miR-16 mimic designed to counteract the loss of the miR-15 and miR-16 family miRNAs in cancer cells. In preclinical models, the expression of miR-16 was restored in malignant pleural mesothelioma tumor specimens as well as in xenograft-bearing nude mice, resulting in a dose-dependent tumor growth inhibition [111]. In the following clinical study phase I (NCT02369198), 1 out of 26 patients showed a partial response, and 15 patients showed stable disease. Regarding the side effects, five dose-limiting toxicities occurred, but no drug-related deaths [112]. The discussed siRNA and miRNA therapeutics targeting cancer cells are depicted in the figure in Section 4.
Table 1. Overview of clinical trials exploiting siRNA and miRNA therapeutics in cancer therapy.
Table 1. Overview of clinical trials exploiting siRNA and miRNA therapeutics in cancer therapy.
Name of the ProductCellular TargetDisease Phase and NCTRecruiting StatusSource
siRNAsiG12D LODERKRASG12DPancreatic cancerPhase II
NCT01676259
Unknown status[98,99]
iExosomesKRASG12DMetastatic pancreas cancerPhase I
NCT03608631
Active[100,101,102]
Atu027Protein kinase N3Advanced solid tumorsPhase I
NCT00938574
Completed[103,104]
TKM-080301PLK1Primary or secondary liver cancerPhase I
NCT01437007
Completed[105]
STP705COX-2 and TGFβCutaneous squamous cell carcinoma (in situ)Phase II
NCT04844983
Completed[106]
siRNA-EphA2-DOPCEphA2Advanced recurrent solid tumorsPhase I
NCT01591356
Recruiting [107,108]
-NUDT21Refractory retinoblastoma Phase I
NCT06424301
Not yet recruiting [109]
miRNAMRX34 miR-34 mimicPulmonary lymphangitic carcinomatosis, small cell lung cancer, lymphoma, melanoma, multiple myeloma, renal-cell carcinoma, non-small cell lung cancer Phase I
NCT01829971
Terminated[110]
MelanomaPhase I/II
NCT02862145
Withdrawn[110]
TargmiRsmiR16 mimicMalignant pleural mesothelioma and NSCLCPhase I
NCT02369198
Completed[111,112]

4. Exploiting the Immune System for Cancer Therapy

While all the above-mentioned RNA treatments are designed to manipulate cancer cells, the cells of the immune system could represent a potential alternative target (Figure 3). The term immune surveillance emphasizes that the host’s immune system is able to detect tumor cells and even has the capability to inhibit tumor growth [113,114,115]. However, the tumor counteracts the immune surveillance by various immune escape mechanisms. This antagonism is delineated in the concept of the three phases of immune editing: eradiation, equilibrium, and escape [113,115]. One escape mechanism is the development of a tumor-supportive, immune-suppressive TME as an ecological niche for the cancer cells, established via soluble factors such as cytokines, chemokines, and metabolites, as wells as the upregulation of inhibiting surface receptors by cancer cells (e.g., immune checkpoints) [114,116,117,118].
The TME consists not only of cancer cells and cancer-cell-derived factors but also of several non-tumoral cells, including various immune cells [119]. These are innate immune cells such as monocytes, macrophages, dendritic cells (DCs), and natural killer (NK) cells, as well as adaptive immune cells such as T cells and B cells [120,121,122]. However, it became evident that the presence of these immune cells is not sufficient for controlling the tumor since their altered phenotype within the TME acts in favor of the tumor. This includes the induction of immune suppressive cells, the functional inhibition of pro-inflammatory immune cells, and the attraction of tolerance-promoting immune cells [119,120]. Consequently, the tumor-mediated pro-tumoral immune suppression and the anti-tumoral immune effects are locked in a constant battle, with most immune cells being able to fight both sides. Today, it is widely accepted that the localization, number, and phenotype of the immune cells are decisive for the clinical outcome [123].
In this complex microcosm, pro-inflammatory immune cells such as M1-like macrophages, conventional dendritic cells type 1 (cDC1), and cytotoxic CD8+ T cells exert anti-tumoral functions and are associated with improved survival for cancer patients [124,125,126]. In contrast to that, anti-inflammatory immune cell phenotypes such as M2-like macrophages, the myeloid-derived suppressor cells (MDSC), mature DCs enriched in immunoregulatory genes (mreg-DCs), regulatory T cells (Treg), and B cells (Breg), as well as low numbers of cytotoxic CD8+ T cells and cDC1 have been linked to a poor survival prognosis [127,128]. Here, it must be emphasized that the antigen-presenting cells (APCs) that process and present tumor antigen and induce T cell activation, especially, play a central role in the decision for an anti- or pro-tumoral immune response. For efficient control of the tumor, different immune cells are required: those that identify abnormal host cells and those that exert effector functions for the elimination of these cells.

4.1. Rethinking Cancer Immunotherapy—The Potential of Small Non-Coding RNAs

Current cancer treatments include several immunotherapeutic approaches that include immune checkpoint inhibitors that block inhibitory receptors on immune effectors cells or their ligands and thus overcome functional impairment of anti-tumor immune reactions, adoptive cell transfer therapy with ex vivo expanded immune cells which can be transfected with chimeric antigen receptors (CARs) or T cell receptors, monoclonal antibodies, antibody–drug conjugates, treatment vaccines (such as the oncolytic virus T-VEC or monocyte-derived DCs) and small molecule drugs (reviewed by Liu et al. [129]). The main problems affecting all immunotherapies are immune-resistance mechanisms, including the initially low mutational burden of the tumor cells that results in the absence of tumor-specific antigens, adaptive changes of cancer cells causing induced therapy resistance, and an immune suppressive environment formed by infiltrating immune cells such as Tregs, MDSCs and tumor-associated macrophages (TAMs) [130,131,132]. sncRNAs in immunotherapy have two potential applications: they can serve as predictive biomarkers and as therapeutic targets. As predictive markers, specific miRNAs in the blood and tumor tissue of cancer patients can correlate with either positive or negative outcomes of CAR-T cell therapy, cancer vaccines, immune checkpoint inhibition, and small molecule treatment, as reviewed extensively by Alahdal et al. [133]. sncRNAs may serve as direct targets not only in tumor cells to address pro-tumoral alterations in their miRNA expression but also within immune cells in the TME to direct them toward an anti-tumoral phenotype [134,135]. The database RNA2immune lists today (accessed June 2024) over 3600 human miRNAs that are associated with cancer immunology, including cancer cells and adjacent immune cells [136]. Similar to immune cells, sncRNAs can act in favor or in detriment of the tumor and can participate in immune escape or immune response [135]. In general, therapeutic approaches targeting the myeloid cells within the TME, especially addressing the immune-suppressive phenotypes, are quite rare but could be beneficial in combination with the previously listed cancer therapies. The next section summarizes which myeloid cells are possible targets and how reprogramming on a molecular level could be achieved. The term ‘reprogramming’ in the following stands for shifting the balance within the TME by inhibiting immune suppressive signals and enhancing pro-inflammatory effects to recapture the immune cells’ own anti-tumoral functions.

4.2. Targeting Myeloid Cells to Rectify the Tumor Microenvironment

4.2.1. Macrophages

According to Mills et al., activated macrophages are divided into two phenotypes called classically activated and pro-inflammatory (M1) or alternatively activated and anti-inflammatory (M2), based on their polarization. Today, this clear division is widely discussed and shifted to the assumption that polarized macrophages in vivo form a spectrum. Since their polarization occurs as a response to a multitude of environmental influences, a variety of phenotypes in between the full M1 or M2 polarization are observed [137,138,139]. Despite this gradient, the phenotypical division is still made into M1 and M2 macrophages, with M1 macrophages expressing the surface markers CD86, CD80, CD68, and major histocompatibility complex class II (MHC-II), and the intracellular markers inducible nitric oxide synthetase (iNOS), interferon regulating factor 5 (IRF5), and signal transducer and activator of transcription (STAT) 1. On the contrary, M2 macrophages express the surface markers CD206, CD163, CD209, and the intracellular markers arginase 1, IRF4, and STAT3, while showing low expression levels of M1 markers [140,141].
In cancer research, TAMs represent macrophages in the TME, which are the main infiltrating immune cell population [142,143]. Regarding their phenotype, TAMs were initially classified as M2-like macrophages following Mills nomenclature [144]. Recently, it became evident that TAMs form at least two subgroups within the TME that are differently polarized into M1-like or M2-like phenotypes harboring different functionalities [145]. In this context, M1-like TAMs have the potential to kill tumor cells via antibody-dependent cellular cytotoxicity and phagocytosis and can induce anti-tumoral responses of innate and adaptive immune cells. However, in most established tumors, the environmental conditions formed by the tumor enhance the number of M2-like TAMs. This contributes to cancer progression and metastasis formation by several mechanisms, including promotion of tumor cell proliferation and survival, angiogenesis, and suppression of innate and adaptive immune responses [145,146].
Since environmental conditions drive the polarization of macrophages, the signaling molecules present in the TME as well as the receptor repertoire and the activity of internal signaling pathways of the macrophages, are substantially involved in the expression of either M1- or M2-related genes [147]. Table 2 summarizes key signaling pathways for macrophage polarization. For M1 polarization, tumor necrosis factor α (TNFα), CD40 ligand (CD40L), Toll-like receptors (TLR)-ligands, and granulocyte/macrophage colony-stimulating factor (GM-CSF) signaling converge in the induction of nuclear factor-κB (NFκB) downstream signaling as well as mitogen-activated protein kinase (MAPK), and serine/threonine-protein kinase (AKT) signaling. Apart from that, M1 polarization is induced via interferon γ (IFNγ) signaling that activates the interferon regulating factor 9 (IRF9), resulting in the expression of interferon-stimulated genes (ISGs). Further, M1 polarization is induced by IL12p40 and IL12p35 binding to IL-12R-β1 and β2, respectively. This leads to the translocation of STAT4 into the nucleus, where it binds to the promotor region of IFNγ targeted genes [148,149]. Another M1 driver is Angiopoietin 1 (ANG-1), which binds to the Tek tyrosine kinase receptor (TIE2). Upon ligand binding, the receptor induces signaling via p38 and extracellular signal-regulated kinas 1/2 (ERK1/2) independent of AKT activation [150].
The M2 polarization, which is driven by IL-4, IL-13, IL-10, IL-6, or TGFβ, uses different downstream signaling pathways, including STAT3 and STAT6 or the human mothers against decapentaplegic homologs 2, 3, and 4 (SMAD2/3/4). Further, M2 polarization is induced by colony-stimulating factor 1 (CSF-1), also known as macrophage colony-stimulating factor (M-CSF). Upon binding to the receptor CSF-R1, various singling pathways are induced, including phosphatidylinositol-3 kinase (PI3K)/AKT, jun N-terminal kinase (JNK)/ERK1/2, and phospholipase C γ 2 (PLC-γ2), which promote survival, proliferation, and differentiation of the macrophages [151,152]. Similar to ANG-1, ANG-2 binds to TIE2 but does not induce phosphorylation, thereby ANG-2 acts as a competitive inhibitor of ANG-1. This results in the upregulation of M2-associated genes, including IL-10, CCL17, and pro-angiogenic genes [153]. Additionally, the signaling axis of CD47 and the signal regulatory protein α (SIRPα) contribute to M2 polarization. Upon binding of the ligand CD47 to the receptor SIRPα, the immunoreceptor tyrosine-based inhibitory motif (ITIM) of SIRPα binds the cytoplasmatic Src homology 2 domain phosphatases 1 and 2 (SHP1/2), which are involved in several signal transduction process. The inhibition of SHP1/2 by SIRPα diminishes PI3K, NFκB, MAPK, and STAT1 activation and suppresses M1 polarization [154,155]. The same mechanism is observed for leukocyte immunoglobulin-like receptor (LILRB2)-binding HLA class I. The ITIMs bind SHP1/2 and thereby down-regulate MAPK and NFκB signaling [156,157].
For therapeutic reprogramming, enhancement of M1-driving signaling pathways and inhibiting M2-driving signaling pathways is desirable. Here, a therapeutic approach with non-coding RNAs could support the expression of M1-related genes and inhibit the expression of proteins that drive an M2 phenotype.
Table 2. Overview of macrophage signaling pathways involved in cell polarization.
Table 2. Overview of macrophage signaling pathways involved in cell polarization.
Signaling MoleculeReceptor on MacrophagesSignaling PathwaysSource
Pro-inflammatory,
M1-like
TNFαTNFR1/2NFκB and MAPK [158]
CD40LCD40NFκB and MAPK[159]
IFNγIFNRIRF9 [160]
TLR-ligandsTLR2/3/4/5/6MAPK and NFκB[161,162]
GM-CSFGM-CSF receptorAKT and NFκB [163]
IL-12p40
IL12p35
IL-12R-β1
IL-12R-β2
STAT4[148,149]
ANG-1TIE2P38/ERK1/2[150]
Anti-inflammatory,
M2-like
IL-4IL-4RSTAT6, PI3K/AKT[164]
IL-13IL-13RSTAT6[165]
IL-6IL-6RSTAT3, NFκB[166]
IL-10IL-10RSTAT3[167]
TGFβTGFβ receptorSMAD2/3/4[168]
CSF-1 (M-CSF)CSF-R1PI3K/AKT, JNK/ERK1/2, PLC-γ2[151,152]
ANG-2TIE2No signaling[153]
CD47SIRPαSHP1/2 fishing[154,155]
HLA class ILILRBSHP1/2 fishing[156,157]
Besides targeting internal signaling pathways that affect macrophage polarization, M1 and M2 macrophages can be functionally altered by inhibiting immune regulatory surface markers or by changing their cytokine expression. M1 macrophages secrete a variety of pro-inflammatory cytokines, including IL-6, IL-8, TNF, IL-1β, IL-12, and IL-23, nitric oxide (NO), reactive oxygen species (ROS), and down-regulate the expression of IL-10 [141]. M2 macrophages express high quantities of the cytokines IL-10 and TGFβ, the chemokines CCL17, CCL18, and CCL22, as well as the growth factors CSF-1 and vascular endothelial growth factor (VEGF) [141]. The role of M2 cytokines as central drivers of the immune escape underlines the critical role of all cytokines in the TME as key mediators of an anti-inflammatory or pro-tumoral immune response of several immune cells, not only of TAMs. Consequently, siRNA specifically targeting mRNA of these cytokines or receptors in the TME is a promising therapeutic strategy (Figure 3).

4.2.2. Dendritic Cells

DCs are divided into several subgroups depending on their ontogeny and phenotype, with emerging subgroups and a detectable heterogeneity within the subgroups. Consequently, this field is continuously developing, as reviewed by Segura et al. [169]. However, one main distinction is made between plasmacytoid DCs and conventional DCs. The latter are further divided into cDC1 and cDC2 [169]. This review focuses specifically on the DC subsets cDC1 and cDC2 that harbor different activating functions within the immune system. cDC1s, on the one hand, mediate immune responses against intracellular pathogens, drive T helper cell (Th) 1 activation, and are crucial for an anti-tumor immune response. cDC2, on the other hand, are central in extracellular pathogen defense by driving Th2 and Th17 responses and have a rather controversial role in anti-tumor immune responses [170]. Regarding ontogeny, both subsets derive from precursor cells from the bone marrow which are already partially committed to become cDC1 or cDC2 [171]. The underlying parameters of cDC differentiation are poorly understood. However, it is believed that at least a part of pre-cDCs differentiate within the tissue depending on environmental signals [172]. Within the tumor tissue, reduced chemokine-mediated recruitment of cDC1s [125], as well as dysfunctional cDCs have been observed [128,173].
In line with that, immature DCs (named iDCs) were identified in tumor tissue that was unable to stimulate T cells [174] and additionally expressed low levels of the co-stimulatory molecules CD80 and CD86, or none at all [175,176]. Ex vivo stimulation experiments of these iDCs failed since CD86 and CD80 were not inducible upon exogenous stimulation with TNF or CD40L [177]. This implies that these iDCs are not merely unstimulated but are functionally inhibited and have lost the ability to react adequately upon stimulation. The presentation of tumor antigens by these iDC in the absence of co-stimulatory molecules misleads T cell responses and induces peripheral T cell tolerance and anergy, which supports the tumor in immune evasion [177,178,179].
When examining mature DCs within the TME of human and mouse non-small cell lung cancer (NSCLC) samples, Maier et al. identified mregDCs that expressed an altered set of maturation genes. This regulatory cell program was identified in cDC1s and cDC2s upon tumor antigen uptake [128], indicating that DCs are altered by tumor cells within the TME. This alteration of DCs represents a mechanism that can potentially be targeted by cell reprogramming in new therapeutic approaches.
As described above, DCs are the central antigen-presenting cells that induce T cell responses via direct cell-to-cell contact. Hence, the surface molecule expression regarding co-stimulatory molecules and immune regulatory molecules is of vital importance. Table 3 subsumes tolerance-promoting signaling pathways that can lead to a down-regulation of DC-mediated CD8+ T cell responses or induce Treg generation [180]. One main tolerance-promoting effect on DCs is the down-regulation of CD80/86 as well as CD40. It is well known that this can be mediated via prostaglandin E2 (PGE2) and VEGF derived from tumor cells or other anti-inflammatory immune cells within the TME to suppress the maturation and function of DCs [181,182,183]. Using RNAi mechanisms to block the expression of the main receptors involved in the inhibition of DC functions, such as VEGF and PGE2 receptors, could be exploited as a therapeutic approach. In addition to that, non-coding RNA could be used to rescue the expression of co-stimulatory molecules and help to avoid the induction of T cell tolerance. To this end, therapies based on non-coding RNAs could target the negative regulators (e.g., miRNAs) of the mRNAs encoding these co-stimulatory molecules. However, since there is a variety of inhibitory signals that act simultaneously on DCs, the overall success of these strategies is questionable and needs to be tested. Alternatively, the overexpression of miR181-d could be a useful therapeutic tool since it has been shown that the overexpression of this miRNA alone is sufficient to induce DC maturation accompanied by the upregulation of CD80 and CD83 [184]. Su et al. suggested that, as a mechanism of action, miR-181-d targets a negative regulator of the NFκB signaling pathway, resulting in constitutive activation of the NFκB signaling pathway and cell maturation [184]. The direct induction of co-stimulatory molecule expression via NFκB signaling is also achievable via TLR ligands or by mRNA-mediated expression of constitutively active members of the NFκB-signaling cascade [185], but it has to be tested, which method is more useful.
Another promising target of RNAi-based therapy is checkpoint molecules on the DC surface, such as programmed cell death ligand 1 (PD-L1) and T cell immunoglobulin mucin receptor 3 (TIM3). TIM3 functions as a negative regulator of innate immune responses against nucleic acid in an HMGB1-dependent mechanism. The abrogated response of DCs towards nucleic acids diminishes the therapeutic efficacy of chemotherapy and DNA vaccination in cancer therapy [186]. Additionally, the expression of additional checkpoint molecules, including lymphocyte activation gene 3 protein (LAG3), CD200R, inducible T cell co-stimulator and its ligand (ICOS(L)), and glucocorticoid-induced tumor necrosis factor receptor related protein (GITR), have been reported for cDC1s [187]. For therapeutic use, the down-regulation of PD-L1 and other inhibitory receptors is immensely important, and, therefore, siRNA-mediated phenotypical knockdowns would be useful (Figure 3).
Cytokines that inhibit DC function are the anti-inflammatory cytokines IL-10 and TGFβ. Of note, these cytokines also drive M2 macrophage polarization (Table 3) [188,189,190]. Consequently, inhibiting the expression of the referring cytokine receptors on both immune cell populations could change the functionality of these cells within the TME tremendously. Further, Kudo-Saito et al. reported that tumor cell-derived CCL2 also promotes regulatory DCs, which in turn induce immunosuppressive CD4+FOXP3+ regulatory T cells [191]. Furthermore, Maier et al. demonstrated that mregDCs upregulate the IL-4R and that the IL-4 signaling down-regulates the IL-12 production by DCs, which is a central cytokine in T cell stimulation. The IL-12 production could be reinduced by a blockage of IL-4 [128]. Besides IL-4 blocking antibodies, RNAi that targets the downregulation of IL-4R or the upregulation of IL-12 could prevent DCs from acquiring a regulatory phenotype and, in turn, increase the stimulation of cytotoxic CD8+ T cells instead of regulatory T cells, but this needs to be tested.
Table 3. Signaling pathways inhibiting dendritic cell functions in the TME.
Table 3. Signaling pathways inhibiting dendritic cell functions in the TME.
Signaling MoleculeDC Marker ExpressionSource
PGE2MHCII↓ 1, CD40↓, PD-L1↑ 2[181]
VEGFMHCII↓, CD40↓, CD86↓, IL-12↓[182,183]
HMGB1TIM3↑[186,192]
CCL2HLA-DR↓, PD-L1↑[191]
IL-10CD86↓, CD80↓, CD40↓, CD83↓, PD-L1↑ [188,190]
TGFβMHCII↓, CD40↓, PD-L1↑
CD86↓, CD80↓, CD83↓, IL-12↓
[173,192,193]
IL-4IL-12↓[128]
1 ↓ downregulation, 2 ↑ upregulation.

4.2.3. Myeloid-Derived Suppressor Cells

In general, MDSCs are the subject of a controversial discussion. Originally, it was thought that MDSCs are migrated myeloid cells that are diverted within the TME toward an immature, non-specific immunosuppressive phenotype [194]. Recently, this shifted to the assumption of pathologically activated polymorphonuclear (PMN-MDSCs) and monocytic (M-MDSCs) MDSCs, both harboring immune suppressive functions [195,196]. The accumulation of MDSCs is induced by various factors in the TME, including cyclooxygenase 2, prostaglandins, GM-CSF, M-CSF, stem cell factor (SCF), VEGF, IL-1β, IL-6, IL-4, IL-10, and TGFβ [197,198]. However, it is still difficult to discriminate these cells from healthy cells by surface marker expression [199]. So far, human MDSCs were described as HLA-DRlow, CD33+, and CD11b+ while being negative for DC markers (except CD11b), as well as NK-cell and T cell markers [199]. Alternatively, Bronte et al. define PMN-MDSCs as CD11b+CD14CD15+ or CD11b+CD14CD66b+, and M-MDSCs as CD11b+CD14+HLA-DR−/lowCD15, with CD33 that can be used as a marker instead of CD11b [195]. Besides a characteristic surface marker expression, MDSCs showed changes in transcriptomic and metabolomic levels compared to normal polymorphonuclear cells, as well as the ability to inhibit other immune cells, including T cells, DCs, and NK cells [199]. In summary, MDSCs remain an interesting target within the TME, but since there is neither a clear definition nor a defined phenotype of these cells, there is not yet enough information available to target these cells. Hence, the current focus lies on macrophages and DCs as targets for reprogramming.
Figure 3. Therapeutical potential of small-interfering RNA and micro-RNA in cancer therapy. The therapeutical options are divided into two mechanisms of action: red hammers indicate the inhibition of pro-tumoral effects via small-interfering RNA (siRNA) or micro-RNA (miRNA) mimics of endogenous inhibitory miRNAs that knock down the referring mRNAs, blue arrows indicate the activation of anti-tumoral effects via anti-miRs that target miRNAs that are crucial in the negative regulation of these effects. An exception is the substitution therapy with miRNA mimics (violet arrow). For tumor cells, the inhibition of oncogenic drivers, immune suppressive cytokines, and pro-tumoral cell surface receptors are potential targets as well as the re-activation of oncogenic suppressor genes and pro-inflammatory cytokines. Thereby, the tumor growth is inhibited, cancer cell apoptosis is induced, and the suppressive microenvironment is shifted towards inflammation. For APCs like macrophages and dendritic cells, the inhibition of inhibitory cytokines and surface receptors (e.g., programmed cell-death ligand 1 (PD-L1)), as well as the activation of pro-inflammatory cytokines and the upregulation of co-stimulatory molecules, are therapeutical approaches to enhance inflammation and immune cell activation within the tumor microenvironment (TME). The reprogramming of inhibitory surface receptors and soluble factors of antigen-presenting cells (APCs) and cancer cells also affects other immune cells within the TME (e.g., natural killer (NK) cells and T cells), for example, via abrogating PD-L1/PD-1 interactions.
Figure 3. Therapeutical potential of small-interfering RNA and micro-RNA in cancer therapy. The therapeutical options are divided into two mechanisms of action: red hammers indicate the inhibition of pro-tumoral effects via small-interfering RNA (siRNA) or micro-RNA (miRNA) mimics of endogenous inhibitory miRNAs that knock down the referring mRNAs, blue arrows indicate the activation of anti-tumoral effects via anti-miRs that target miRNAs that are crucial in the negative regulation of these effects. An exception is the substitution therapy with miRNA mimics (violet arrow). For tumor cells, the inhibition of oncogenic drivers, immune suppressive cytokines, and pro-tumoral cell surface receptors are potential targets as well as the re-activation of oncogenic suppressor genes and pro-inflammatory cytokines. Thereby, the tumor growth is inhibited, cancer cell apoptosis is induced, and the suppressive microenvironment is shifted towards inflammation. For APCs like macrophages and dendritic cells, the inhibition of inhibitory cytokines and surface receptors (e.g., programmed cell-death ligand 1 (PD-L1)), as well as the activation of pro-inflammatory cytokines and the upregulation of co-stimulatory molecules, are therapeutical approaches to enhance inflammation and immune cell activation within the tumor microenvironment (TME). The reprogramming of inhibitory surface receptors and soluble factors of antigen-presenting cells (APCs) and cancer cells also affects other immune cells within the TME (e.g., natural killer (NK) cells and T cells), for example, via abrogating PD-L1/PD-1 interactions.
Ijms 25 07275 g003

4.3. Reprogramming Experiments Targeting Myeloid Cells

While immunotherapies have largely focused on increasing the functionality of cytotoxic T cells (e.g., immune checkpoint blockade or CAR-T cells), treatments that intent to reprogram immune cells from a pro-tumoral to a pro-inflammatory phenotype with the usage of non-coding RNAs as a therapeutic tool have not been studied intensively. Nevertheless, there has been some respective research evaluating RNAi as immunotherapy for cancer, which will be summarized in the following.
Table 4 summarizes therapeutical approaches to reprogramming myeloid cells in murine cancer models. In a colon cancer model, Rossowska et al. down-regulated the expression of IL-10 in DCs and tumor cells with short-hairpin RNA (shRNA) shuttled via a lentiviral vector. This resulted in decreased IL-10 concentration and enhanced DC activity in the TME. Furthermore, reduced numbers of MDSCs and Tregs within the TME, an enhanced Th1-type anti-tumoral immune response, and delayed tumor growth were observed [200]. Cubillos-Ruiz et al. used a murine ovarian cancer model to demonstrate that anti-PD-L1 siRNA, shuttled via nanoparticles, was able to reprogram tumor-adjacent regulatory DCs towards activated DCs in vivo. The re-activated immune response resulted in an increased survival span of the mice compared to anti-PD-L1 antibody treatment [201].
Focusing on the reprogramming of macrophages, the repolarization of TAMs with M2-phenotype was investigated in a murine 4T1-based metastatic breast cancer model. Here, CpG-DNA as a TLR-agonist combined with an anti-IL-10R antibody shifted M2 into M1 polarized macrophages. The treatment was associated with inhibited metastasis formation and prolonged survival compared to the control group [202]. Also targeting TAMs, Zhang et al. demonstrated in an ovarian cancer model that in vivo reprogramming of M2-like macrophages by enhancing the NFκB signaling was achieved. In this approach, mRNA encoding interferon regulating factor 5 and the kinase IKKβ, both components of the NFκB signaling pathway, were applied simultaneously and packaged in nanoparticles. This treatment induced anti-tumoral immunity and tumor regression [203]. Mimicking the upregulating effects of these mRNAs enhancing the NFκB signaling, siRNA targeting negative regulators of the NFκB pathway could induce more stable M2-like macrophages, similar to miR-181-d. In another approach, a prostate cancer model and nanoparticles equipped with β-Cyclodextrin were used to target specifically M2 macrophages. As a therapeutic agent, siRNA directed against CSF-R1, which was utilized to abrogate the CSF-R1 signaling, a main driver of M2 polarization within the TME. Upon treatment, M1-related cytokines increased while M2 macrophage numbers decreased. Further, an increased cytotoxic T cell infiltration and inhibited tumor cell growth was observed [204]. Shobaki et al. utilized a mouse model with xenografts of human renal cancer and lipid nanoparticles to specifically shuttle siRNA targeting STAT3 and hypoxia-inducible factor 1-alpha (HIF-1α) to TAMs. The treatment resulted in increased numbers of macrophages and a higher abundance of the M1 polarized cells within the TME. Additionally, a decrease in angiogenesis and tumor growth was observed [205]. Furthermore, Qian et al. used a nanoparticle-based delivery system to target M2 macrophages within a murine 4T1 breast cancer model. Here, the therapeutic agent consisted of a CpG oligonucleotide and a siRNA directed against PI3Kγ (phosphoinositid-3-kinase γ). The conjugate molecule was designed to be cleaved within the cells by Dicer, so upon cleavage, the CpG oligonucleotide activates TLR9 while the siRNA mediates the inhibition of PI3Kγ and induces proinflammatory NFκB signaling. The researchers observed repolarization of M2 to M1 macrophages, tumor growth inhibition, and prolonged survival of the mice [206].
Table 4. Therapeutic approaches reprogramming myeloid cells in murine cancer models.
Table 4. Therapeutic approaches reprogramming myeloid cells in murine cancer models.
ModelTherapeuticTargetEffectSource
Colon cancershRNAIL-10IL-10↓ 1, DC activity↑ 2, Th1↑, MDSCs↓, Treg↓
Tumor growth↓
[200]
Ovarian cancersiRNAPD-L1regDCs re-activated, survival↑[201]
Breast cancerCpG DNA + anti-IL-10R antibodyTLR, IL-10RM2 macrophages repolarized to M1, metastasis formation↓, survival↑[202]
Ovarian cancerIRF5 and IKKβ mRNANFκB signalingAnti-tumoral immunity↑, tumor growth↓[203]
Prostate cancersiRNACSF-1RM1 cytokines↑, M2 numbers↓, cytotoxic T cell infiltration↑, tumor growth↓[204]
Human xenograft in nude micesiRNASTAT3, HIF1-αMacrophage infiltration in TME↑, M1↑, angiogenesis↓ tumor growth↓[205]
Breast cancer xenograftCpG + siRNATLR, PI3KγM2 macrophages repolarized to M1, tumor growth↓, survival↑[206]
1 ↓ downregulation, 2 ↑ upregulation.
In 2014, Troegeler et al. demonstrated efficient siRNA-mediated gene silencing in ex vivo-generated primary human monocytes, DCs, and macrophages [207]. Table 5 summarizes the following therapeutical approaches in myeloid cells in humans. Cheng et al. showed in isolated MDSCs from renal cancer patients’ blood that these cells were reprogrammed via intervening with the Notch signaling using siRNA. They previously observed that down-regulated Notch signaling in myeloid cells within tumor tissue drives an abnormal cell differentiation of these myeloid cells. Here, the Notch signaling was down-regulated by the kinase CK2 that phosphorylates Notch. Consequently, they used a siRNA directed against CK2, leading to reinitiation of the Notch signaling and, thereby, to normal cell differentiation [208].
Further, a clinical phase Ib trial (NCT03087591) applied siRNA therapy for solid tumors that could not be removed by surgery. Here, peripheral blood mononuclear cells (PBMCs) from the patients were collected via leukapheresis and ex vivo electroporated with siRNA that specifically knocks down Cbl proto-oncogene B (Cbl-b). This product, named APN401, was intravenously infused back into the patient. This resulted in increased production of IFNγ and IL-2 by the PBMCs in response to stimulation with anti-CD3/28, which implies that the function of T cells included in the PBMCs was central in this study [209]. Lastly, the clinical trial NCT04995536 aimed to evaluate the effect of the product named CAS3/SS3, which is a conjugate molecule of a CpG oligonucleotide and a siRNA directed against STAT3. With CAS3/SS3, the growth of tumor cells should be inhibited and immune cells activated. However, the study was withdrawn before starting the patient recruitment [210,211]. Currently, no further siRNA or miRNA therapy approaches targeting specifically myeloid cells in humans were found in preparation for this review.
Table 5. Therapeutic approaches reprogramming myeloid cells in humans.
Table 5. Therapeutic approaches reprogramming myeloid cells in humans.
ModelTherapeuticTargetEffectSource
Ex vivo MDSCs from renal cancer patient’s bloodsiRNACK2 in NOTCH signalingMDSCs reprogramming[208]
Ex vivo PBMCs from solid tumor patients
NCT03087591
siRNA (APN401)Cbl-bIFNγ↑ 1, IL2↑ production of PBMCs[209]
In vivo
NCT04995536
(withdrawn)
CpG + anti-STAT3 siRNA (CAS3/SS3)TLR9, STAT3 -[210,211]
1 ↑ upregulation.

5. Conclusions

Myeloid cells within the TME represent a double-edged sword in anti-tumor immune responses. Since these cells harbor a central role in directing and regulating tumor-specific immune reactions and are present in the TME in high numbers, proper pro-inflammatory functionality of these cells is crucial for a strong anti-tumoral immune response. Re-directing of the immune response can be achieved by reprogramming these myeloid cells in vivo by exploiting siRNA- or miRNA-based therapeutics, as already shown in mice. However, further research regarding driver signaling pathways and complex signaling networks is necessary.

Author Contributions

Conceptualization, A.K.B. and J.D.; writing—original draft preparation, A.K.B.; writing—review and editing, A.K.B., F.B., N.S. and J.D.; visualization, A.K.B. and J.D.; supervision, N.S. and J.D.; funding acquisition, N.S. and J.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from the BMBF in the context of the transnational joint research program ERA-NET TRANSCAN, Project TumorOUT, funding number 01KT2305A, to J.D. This work was also supported by funding from the European Unions’ Horizon Europe research and innovation program under grant agreement number 101057250 (CANCERNA) to N.S. and J.D. on behalf of the CANCERNA Consortium and by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) via the Research Training Group GRK2504/1 (project number 401821119), research project B4 to J.D.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to express our gratitude to the funders of our research.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Neveu, M.; Kim, H.J.; Benner, S.A. The “strong” RNA world hypothesis: Fifty years old. Astrobiology 2013, 13, 391–403. [Google Scholar] [CrossRef] [PubMed]
  2. Baulcombe, D.C. RNA as a target and an initiator of post-transcriptional gene silencing in transgenic plants. Plant Mol. Biol. 1996, 32, 79–88. [Google Scholar] [CrossRef] [PubMed]
  3. Kazimierczyk, M.; Kasprowicz, M.K.; Kasprzyk, M.E.; Wrzesinski, J. Human Long Noncoding RNA Interactome: Detection, Characterization and Function. Int. J. Mol. Sci. 2020, 21, 1027. [Google Scholar] [CrossRef] [PubMed]
  4. Pantaleo, V.; Masuta, C. Diversity of viral RNA silencing suppressors and their involvement in virus-specific symptoms. Adv. Virus Res. 2022, 113, 1–23. [Google Scholar] [CrossRef]
  5. Koonin, E.V. Evolution of RNA- and DNA-guided antivirus defense systems in prokaryotes and eukaryotes: Common ancestry vs convergence. Biol. Direct 2017, 12, 5. [Google Scholar] [CrossRef]
  6. Torri, A.; Jaeger, J.; Pradeu, T.; Saleh, M.C. The origin of RNA interference: Adaptive or neutral evolution? PLoS Biol. 2022, 20, e3001715. [Google Scholar] [CrossRef]
  7. Jacob, F.; Monod, J. Genetic regulatory mechanisms in the synthesis of proteins. J. Mol. Biol. 1961, 3, 318–356. [Google Scholar] [CrossRef]
  8. Wagner, E.G.; Simons, R.W. Antisense RNA control in bacteria, phages, and plasmids. Annu. Rev. Microbiol. 1994, 48, 713–742. [Google Scholar] [CrossRef]
  9. Fire, A.; Albertson, D.; Harrison, S.W.; Moerman, D.G. Production of antisense RNA leads to effective and specific inhibition of gene expression in C. elegans muscle. Development 1991, 113, 503–514. [Google Scholar] [CrossRef]
  10. Fire, A.; Xu, S.; Montgomery, M.K.; Kostas, S.A.; Driver, S.E.; Mello, C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998, 391, 806–811. [Google Scholar] [CrossRef]
  11. Mello, C.C.; Conte, D., Jr. Revealing the world of RNA interference. Nature 2004, 431, 338–342. [Google Scholar] [CrossRef]
  12. Wang, P.; Zhou, Y.; Richards, A.M. Effective tools for RNA-derived therapeutics: siRNA interference or miRNA mimicry. Theranostics 2021, 11, 8771–8796. [Google Scholar] [CrossRef] [PubMed]
  13. Doudna, J.A.; Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 2014, 346, 1258096. [Google Scholar] [CrossRef]
  14. Huang, X.; Chen, Z.; Liu, Y. RNAi-mediated control of CRISPR functions. Theranostics 2020, 10, 6661–6673. [Google Scholar] [CrossRef] [PubMed]
  15. Cech, T.R.; Steitz, J.A. The noncoding RNA revolution-trashing old rules to forge new ones. Cell 2014, 157, 77–94. [Google Scholar] [CrossRef] [PubMed]
  16. Barrangou, R.; Fremaux, C.; Deveau, H.; Richards, M.; Boyaval, P.; Moineau, S.; Romero, D.A.; Horvath, P. CRISPR provides acquired resistance against viruses in prokaryotes. Science 2007, 315, 1709–1712. [Google Scholar] [CrossRef]
  17. Mojica, F.J.; Diez-Villasenor, C.; Garcia-Martinez, J.; Soria, E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J. Mol. Evol. 2005, 60, 174–182. [Google Scholar] [CrossRef]
  18. Bolotin, A.; Quinquis, B.; Sorokin, A.; Ehrlich, S.D. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology 2005, 151, 2551–2561. [Google Scholar] [CrossRef]
  19. Elbashir, S.M.; Harborth, J.; Lendeckel, W.; Yalcin, A.; Weber, K.; Tuschl, T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001, 411, 494–498. [Google Scholar] [CrossRef]
  20. Uszczynska-Ratajczak, B.; Lagarde, J.; Frankish, A.; Guigo, R.; Johnson, R. Towards a complete map of the human long non-coding RNA transcriptome. Nat. Rev. Genet. 2018, 19, 535–548. [Google Scholar] [CrossRef]
  21. Zhang, P.; Wu, W.; Chen, Q.; Chen, M. Non-Coding RNAs and their Integrated Networks. J. Integr. Bioinform. 2019, 16, 20190027. [Google Scholar] [CrossRef]
  22. Passmore, L.A.; Coller, J. Roles of mRNA poly(A) tails in regulation of eukaryotic gene expression. Nat. Rev. Mol. Cell Biol. 2022, 23, 93–106. [Google Scholar] [CrossRef]
  23. Hocine, S.; Singer, R.H.; Grunwald, D. RNA processing and export. Cold Spring Harb. Perspect. Biol. 2010, 2, a000752. [Google Scholar] [CrossRef]
  24. Shin, Y.; Qayyum, M.Z.; Pupov, D.; Esyunina, D.; Kulbachinskiy, A.; Murakami, K.S. Structural basis of ribosomal RNA transcription regulation. Nat. Commun. 2021, 12, 528. [Google Scholar] [CrossRef]
  25. Raina, M.; Ibba, M. tRNAs as regulators of biological processes. Front. Genet. 2014, 5, 171. [Google Scholar] [CrossRef]
  26. Berg, M.D.; Brandl, C.J. Transfer RNAs: Diversity in form and function. RNA Biol. 2021, 18, 316–339. [Google Scholar] [CrossRef]
  27. Kapranov, P.; Cheng, J.; Dike, S.; Nix, D.A.; Duttagupta, R.; Willingham, A.T.; Stadler, P.F.; Hertel, J.; Hackermuller, J.; Hofacker, I.L.; et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 2007, 316, 1484–1488. [Google Scholar] [CrossRef]
  28. St Laurent, G.; Wahlestedt, C.; Kapranov, P. The Landscape of long noncoding RNA classification. Trends Genet. 2015, 31, 239–251. [Google Scholar] [CrossRef]
  29. Mattick, J.S.; Amaral, P.P.; Carninci, P.; Carpenter, S.; Chang, H.Y.; Chen, L.L.; Chen, R.; Dean, C.; Dinger, M.E.; Fitzgerald, K.A.; et al. Long non-coding RNAs: Definitions, functions, challenges and recommendations. Nat. Rev. Mol. Cell Biol. 2023, 24, 430–447. [Google Scholar] [CrossRef]
  30. Yin, Q.F.; Yang, L.; Zhang, Y.; Xiang, J.F.; Wu, Y.W.; Carmichael, G.G.; Chen, L.L. Long noncoding RNAs with snoRNA ends. Mol. Cell 2012, 48, 219–230. [Google Scholar] [CrossRef]
  31. Wilusz, J.E.; Freier, S.M.; Spector, D.L. 3′ end processing of a long nuclear-retained noncoding RNA yields a tRNA-like cytoplasmic RNA. Cell 2008, 135, 919–932. [Google Scholar] [CrossRef]
  32. Kim, T.K.; Hemberg, M.; Gray, J.M.; Costa, A.M.; Bear, D.M.; Wu, J.; Harmin, D.A.; Laptewicz, M.; Barbara-Haley, K.; Kuersten, S.; et al. Widespread transcription at neuronal activity-regulated enhancers. Nature 2010, 465, 182–187. [Google Scholar] [CrossRef]
  33. Orom, U.A.; Derrien, T.; Beringer, M.; Gumireddy, K.; Gardini, A.; Bussotti, G.; Lai, F.; Zytnicki, M.; Notredame, C.; Huang, Q.; et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 2010, 143, 46–58. [Google Scholar] [CrossRef]
  34. Sartorelli, V.; Lauberth, S.M. Enhancer RNAs are an important regulatory layer of the epigenome. Nat. Struct. Mol. Biol. 2020, 27, 521–528. [Google Scholar] [CrossRef]
  35. Arnold, P.R.; Wells, A.D.; Li, X.C. Diversity and Emerging Roles of Enhancer RNA in Regulation of Gene Expression and Cell Fate. Front. Cell Dev. Biol. 2019, 7, 377. [Google Scholar] [CrossRef]
  36. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The biogenesis, biology and characterization of circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef]
  37. Czech, B.; Munafo, M.; Ciabrelli, F.; Eastwood, E.L.; Fabry, M.H.; Kneuss, E.; Hannon, G.J. piRNA-Guided Genome Defense: From Biogenesis to Silencing. Annu. Rev. Genet. 2018, 52, 131–157. [Google Scholar] [CrossRef]
  38. Zhang, T.; Chen, L.; Li, R.; Liu, N.; Huang, X.; Wong, G. PIWI-interacting RNAs in human diseases: Databases and computational models. Brief. Bioinform. 2022, 23, bbac217. [Google Scholar] [CrossRef]
  39. Hadjiolov, A.A.; Venkov, P.V.; Tsanev, R.G. Ribonucleic acids fractionation by density-gradient centrifugation and by agar gel electrophoresis: A comparison. Anal. Biochem. 1966, 17, 263–267. [Google Scholar] [CrossRef]
  40. Fica, S.M.; Tuttle, N.; Novak, T.; Li, N.S.; Lu, J.; Koodathingal, P.; Dai, Q.; Staley, J.P.; Piccirilli, J.A. RNA catalyses nuclear pre-mRNA splicing. Nature 2013, 503, 229–234. [Google Scholar] [CrossRef]
  41. Huang, Z.H.; Du, Y.P.; Wen, J.T.; Lu, B.F.; Zhao, Y. snoRNAs: Functions and mechanisms in biological processes, and roles in tumor pathophysiology. Cell Death Discov. 2022, 8, 259. [Google Scholar] [CrossRef]
  42. Lee, R.C.; Ambros, V. An extensive class of small RNAs in Caenorhabditis elegans. Science 2001, 294, 862–864. [Google Scholar] [CrossRef]
  43. Lagos-Quintana, M.; Rauhut, R.; Lendeckel, W.; Tuschl, T. Identification of novel genes coding for small expressed RNAs. Science 2001, 294, 853–858. [Google Scholar] [CrossRef]
  44. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  45. Carthew, R.W.; Sontheimer, E.J. Origins and Mechanisms of miRNAs and siRNAs. Cell 2009, 136, 642–655. [Google Scholar] [CrossRef]
  46. Ha, M.; Kim, V.N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef]
  47. Jin, W.; Wang, J.; Liu, C.P.; Wang, H.W.; Xu, R.M. Structural Basis for pri-miRNA Recognition by Drosha. Mol. Cell 2020, 78, 423–433.e425. [Google Scholar] [CrossRef]
  48. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Radmark, O.; Kim, S.; et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  49. Yeom, K.H.; Lee, Y.; Han, J.; Suh, M.R.; Kim, V.N. Characterization of DGCR8/Pasha, the essential cofactor for Drosha in primary miRNA processing. Nucleic Acids Res. 2006, 34, 4622–4629. [Google Scholar] [CrossRef]
  50. Lund, E.; Guttinger, S.; Calado, A.; Dahlberg, J.E.; Kutay, U. Nuclear export of microRNA precursors. Science 2004, 303, 95–98. [Google Scholar] [CrossRef]
  51. Bernstein, E.; Caudy, A.A.; Hammond, S.M.; Hannon, G.J. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001, 409, 363–366. [Google Scholar] [CrossRef]
  52. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  53. Eamens, A.L.; Smith, N.A.; Curtin, S.J.; Wang, M.B.; Waterhouse, P.M. The Arabidopsis thaliana double-stranded RNA binding protein DRB1 directs guide strand selection from microRNA duplexes. RNA 2009, 15, 2219–2235. [Google Scholar] [CrossRef]
  54. Piatek, M.J.; Werner, A. Endogenous siRNAs: Regulators of internal affairs. Biochem. Soc. Trans. 2014, 42, 1174–1179. [Google Scholar] [CrossRef]
  55. Yang, N.; Kazazian, H.H., Jr. L1 retrotransposition is suppressed by endogenously encoded small interfering RNAs in human cultured cells. Nat. Struct. Mol. Biol. 2006, 13, 763–771. [Google Scholar] [CrossRef]
  56. Watanabe, T.; Takeda, A.; Tsukiyama, T.; Mise, K.; Okuno, T.; Sasaki, H.; Minami, N.; Imai, H. Identification and characterization of two novel classes of small RNAs in the mouse germline: Retrotransposon-derived siRNAs in oocytes and germline small RNAs in testes. Genes Dev. 2006, 20, 1732–1743. [Google Scholar] [CrossRef]
  57. Castel, S.E.; Martienssen, R.A. RNA interference in the nucleus: Roles for small RNAs in transcription, epigenetics and beyond. Nat. Rev. Genet. 2013, 14, 100–112. [Google Scholar] [CrossRef]
  58. Meister, G.; Tuschl, T. Mechanisms of gene silencing by double-stranded RNA. Nature 2004, 431, 343–349. [Google Scholar] [CrossRef]
  59. Hammond, S.M.; Bernstein, E.; Beach, D.; Hannon, G.J. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature 2000, 404, 293–296. [Google Scholar] [CrossRef]
  60. Martinez, J.; Patkaniowska, A.; Urlaub, H.; Luhrmann, R.; Tuschl, T. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell 2002, 110, 563–574. [Google Scholar] [CrossRef] [PubMed]
  61. Anderson, E.M.; Birmingham, A.; Baskerville, S.; Reynolds, A.; Maksimova, E.; Leake, D.; Fedorov, Y.; Karpilow, J.; Khvorova, A. Experimental validation of the importance of seed complement frequency to siRNA specificity. RNA 2008, 14, 853–861. [Google Scholar] [CrossRef]
  62. Kehl, T.; Backes, C.; Kern, F.; Fehlmann, T.; Ludwig, N.; Meese, E.; Lenhof, H.P.; Keller, A. About miRNAs, miRNA seeds, target genes and target pathways. Oncotarget 2017, 8, 107167–107175. [Google Scholar] [CrossRef]
  63. Lewis, B.P.; Shih, I.H.; Jones-Rhoades, M.W.; Bartel, D.P.; Burge, C.B. Prediction of mammalian microRNA targets. Cell 2003, 115, 787–798. [Google Scholar] [CrossRef] [PubMed]
  64. Hutvagner, G.; Zamore, P.D. A microRNA in a multiple-turnover RNAi enzyme complex. Science 2002, 297, 2056–2060. [Google Scholar] [CrossRef]
  65. Okamura, K.; Ishizuka, A.; Siomi, H.; Siomi, M.C. Distinct roles for Argonaute proteins in small RNA-directed RNA cleavage pathways. Genes Dev. 2004, 18, 1655–1666. [Google Scholar] [CrossRef]
  66. Tang, G.; Reinhart, B.J.; Bartel, D.P.; Zamore, P.D. A biochemical framework for RNA silencing in plants. Genes Dev. 2003, 17, 49–63. [Google Scholar] [CrossRef]
  67. Olsen, P.H.; Ambros, V. The lin-4 regulatory RNA controls developmental timing in Caenorhabditis elegans by blocking LIN-14 protein synthesis after the initiation of translation. Dev. Biol. 1999, 216, 671–680. [Google Scholar] [CrossRef]
  68. Zeng, Y.; Yi, R.; Cullen, B.R. MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc. Natl. Acad. Sci. USA 2003, 100, 9779–9784. [Google Scholar] [CrossRef] [PubMed]
  69. Doench, J.G.; Sharp, P.A. Specificity of microRNA target selection in translational repression. Genes Dev. 2004, 18, 504–511. [Google Scholar] [CrossRef] [PubMed]
  70. Jo, M.H.; Shin, S.; Jung, S.R.; Kim, E.; Song, J.J.; Hohng, S. Human Argonaute 2 Has Diverse Reaction Pathways on Target RNAs. Mol. Cell 2015, 59, 117–124. [Google Scholar] [CrossRef]
  71. Tomari, Y.; Zamore, P.D. Perspective: Machines for RNAi. Genes Dev. 2005, 19, 517–529. [Google Scholar] [CrossRef] [PubMed]
  72. Stenvang, J.; Petri, A.; Lindow, M.; Obad, S.; Kauppinen, S. Inhibition of microRNA function by antimiR oligonucleotides. Silence 2012, 3, 1. [Google Scholar] [CrossRef] [PubMed]
  73. Friedman, R.C.; Farh, K.K.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef] [PubMed]
  74. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef]
  75. Roberts, T.C.; Langer, R.; Wood, M.J.A. Advances in oligonucleotide drug delivery. Nat. Rev. Drug Discov. 2020, 19, 673–694. [Google Scholar] [CrossRef] [PubMed]
  76. Smith, E.S.; Whitty, E.; Yoo, B.; Moore, A.; Sempere, L.F.; Medarova, Z. Clinical Applications of Short Non-Coding RNA-Based Therapies in the Era of Precision Medicine. Cancers 2022, 14, 1588. [Google Scholar] [CrossRef]
  77. Hu, B.; Zhong, L.; Weng, Y.; Peng, L.; Huang, Y.; Zhao, Y.; Liang, X.J. Therapeutic siRNA: State of the art. Signal Transduct. Target. Ther. 2020, 5, 101. [Google Scholar] [CrossRef] [PubMed]
  78. Lauffer, M.C.; van Roon-Mom, W.; Aartsma-Rus, A.; Collaborative, N. Possibilities and limitations of antisense oligonucleotide therapies for the treatment of monogenic disorders. Commun. Med. 2024, 4, 6. [Google Scholar] [CrossRef]
  79. Collotta, D.; Bertocchi, I.; Chiapello, E.; Collino, M. Antisense oligonucleotides: A novel Frontier in pharmacological strategy. Front. Pharmacol. 2023, 14, 1304342. [Google Scholar] [CrossRef]
  80. Mansoor, M.; Melendez, A.J. Advances in antisense oligonucleotide development for target identification, validation, and as novel therapeutics. Gene Regul. Syst. Biol. 2008, 2, 275–295. [Google Scholar] [CrossRef]
  81. Levin, A.A. Treating Disease at the RNA Level with Oligonucleotides. N. Engl. J. Med. 2019, 380, 57–70. [Google Scholar] [CrossRef]
  82. Bennett, C.F.; Swayze, E.E. RNA targeting therapeutics: Molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 259–293. [Google Scholar] [CrossRef] [PubMed]
  83. Singh, N.N.; Lawler, M.N.; Ottesen, E.W.; Upreti, D.; Kaczynski, J.R.; Singh, R.N. An intronic structure enabled by a long-distance interaction serves as a novel target for splicing correction in spinal muscular atrophy. Nucleic Acids Res. 2013, 41, 8144–8165. [Google Scholar] [CrossRef] [PubMed]
  84. Dhuri, K.; Bechtold, C.; Quijano, E.; Pham, H.; Gupta, A.; Vikram, A.; Bahal, R. Antisense Oligonucleotides: An Emerging Area in Drug Discovery and Development. J. Clin. Med. 2020, 9, 2004. [Google Scholar] [CrossRef]
  85. Boiziau, C.; Kurfurst, R.; Cazenave, C.; Roig, V.; Thuong, N.T.; Toulme, J.J. Inhibition of translation initiation by antisense oligonucleotides via an RNase-H independent mechanism. Nucleic Acids Res. 1991, 19, 1113–1119. [Google Scholar] [CrossRef]
  86. Yasuhara, H.; Yoshida, T.; Sasaki, K.; Obika, S.; Inoue, T. Reduction of Off-Target Effects of Gapmer Antisense Oligonucleotides by Oligonucleotide Extension. Mol. Diagn. Ther. 2022, 26, 117–127. [Google Scholar] [CrossRef]
  87. Lam, J.K.; Chow, M.Y.; Zhang, Y.; Leung, S.W. siRNA Versus miRNA as Therapeutics for Gene Silencing. Mol. Ther. Nucleic Acids 2015, 4, e252. [Google Scholar] [CrossRef]
  88. Ebert, M.S.; Sharp, P.A. MicroRNA sponges: Progress and possibilities. RNA 2010, 16, 2043–2050. [Google Scholar] [CrossRef]
  89. Ebert, M.S.; Neilson, J.R.; Sharp, P.A. MicroRNA sponges: Competitive inhibitors of small RNAs in mammalian cells. Nat. Methods 2007, 4, 721–726. [Google Scholar] [CrossRef] [PubMed]
  90. Urits, I.; Swanson, D.; Swett, M.C.; Patel, A.; Berardino, K.; Amgalan, A.; Berger, A.A.; Kassem, H.; Kaye, A.D.; Viswanath, O. A Review of Patisiran (ONPATTRO(R)) for the Treatment of Polyneuropathy in People with Hereditary Transthyretin Amyloidosis. Neurol. Ther. 2020, 9, 301–315. [Google Scholar] [CrossRef]
  91. Fernandez-Ruiz, I. Patisiran is beneficial for the treatment of TTR cardiac amyloidosis. Nat. Rev. Cardiol. 2024, 21, 8. [Google Scholar] [CrossRef] [PubMed]
  92. Mickle, K.; Lasser, K.E.; Hoch, J.S.; Cipriano, L.E.; Dreitlein, W.B.; Pearson, S.D. The Effectiveness and Value of Patisiran and Inotersen for Hereditary Transthyretin Amyloidosis. J. Manag. Care Spec. Pharm. 2019, 25, 10–15. [Google Scholar] [CrossRef] [PubMed]
  93. Traber, G.M.; Yu, A.M. RNAi-Based Therapeutics and Novel RNA Bioengineering Technologies. J. Pharmacol. Exp. Ther. 2023, 384, 133–154. [Google Scholar] [CrossRef]
  94. Ventura, P.; Bonkovsky, H.L.; Gouya, L.; Aguilera-Peiro, P.; Montgomery Bissell, D.; Stein, P.E.; Balwani, M.; Anderson, D.K.E.; Parker, C.; Kuter, D.J.; et al. Efficacy and safety of givosiran for acute hepatic porphyria: 24-month interim analysis of the randomized phase 3 ENVISION study. Liver Int. 2022, 42, 161–172. [Google Scholar] [CrossRef] [PubMed]
  95. Gang, X.; Liu, F.; Mao, J. Lumasiran for primary hyperoxaluria type 1: What we have learned? Front. Pediatr. 2022, 10, 1052625. [Google Scholar] [CrossRef] [PubMed]
  96. Raal, F.J.; Kallend, D.; Ray, K.K.; Turner, T.; Koenig, W.; Wright, R.S.; Wijngaard, P.L.J.; Curcio, D.; Jaros, M.J.; Leiter, L.A.; et al. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N. Engl. J. Med. 2020, 382, 1520–1530. [Google Scholar] [CrossRef]
  97. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 15 March 2024).
  98. Jones, S.; Zhang, X.; Parsons, D.W.; Lin, J.C.; Leary, R.J.; Angenendt, P.; Mankoo, P.; Carter, H.; Kamiyama, H.; Jimeno, A.; et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008, 321, 1801–1806. [Google Scholar] [CrossRef]
  99. Golan, T.; Khvalevsky, E.Z.; Hubert, A.; Gabai, R.M.; Hen, N.; Segal, A.; Domb, A.; Harari, G.; David, E.B.; Raskin, S.; et al. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget 2015, 6, 24560–24570. [Google Scholar] [CrossRef] [PubMed]
  100. Surana, R.; Lebleu, V.; Lee, J.; Smaglo, B.; Zhao, D.; Lee, M.; Wolff, R.; Overman, M.; Mendt, M.; McAndrews, K.; et al. Phase I study of mesenchymal stem cell (MSC)-derived exosomes with KRAS G12D siRNA in patients with metastatic pancreatic cancer harboring a KRAS G12D mutation. J. Clin. Oncol. 2022, 40, TPS633. [Google Scholar] [CrossRef]
  101. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
  102. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263. [Google Scholar] [CrossRef] [PubMed]
  103. Aleku, M.; Schulz, P.; Keil, O.; Santel, A.; Schaeper, U.; Dieckhoff, B.; Janke, O.; Endruschat, J.; Durieux, B.; Roder, N.; et al. Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res. 2008, 68, 9788–9798. [Google Scholar] [CrossRef] [PubMed]
  104. Schultheis, B.; Strumberg, D.; Santel, A.; Vank, C.; Gebhardt, F.; Keil, O.; Lange, C.; Giese, K.; Kaufmann, J.; Khan, M.; et al. First-in-human phase I study of the liposomal RNA interference therapeutic Atu027 in patients with advanced solid tumors. J. Clin. Oncol. 2014, 32, 4141–4148. [Google Scholar] [CrossRef] [PubMed]
  105. El Dika, I.; Lim, H.Y.; Yong, W.P.; Lin, C.C.; Yoon, J.H.; Modiano, M.; Freilich, B.; Choi, H.J.; Chao, T.Y.; Kelley, R.K.; et al. An Open-Label, Multicenter, Phase I, Dose Escalation Study with Phase II Expansion Cohort to Determine the Safety, Pharmacokinetics, and Preliminary Antitumor Activity of Intravenous TKM-080301 in Subjects with Advanced Hepatocellular Carcinoma. Oncologist 2019, 24, 747-e218. [Google Scholar] [CrossRef] [PubMed]
  106. Molyneaux, M.; Xu, J.; Evans, D.M.; Lu, P. Effect on tumor growth by TGF-β1/COX-2 siRNA combination product (STP705) in a human cholangiocarcinoma (HuCCT-1) xenograft tumor model in nude mice. J. Clin. Oncol. 2019, 37, e14652. [Google Scholar] [CrossRef]
  107. Landen, C.N., Jr.; Chavez-Reyes, A.; Bucana, C.; Schmandt, R.; Deavers, M.T.; Lopez-Berestein, G.; Sood, A.K. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 2005, 65, 6910–6918. [Google Scholar] [CrossRef] [PubMed]
  108. Wagner, M.J.; Mitra, R.; McArthur, M.J.; Baze, W.; Barnhart, K.; Wu, S.Y.; Rodriguez-Aguayo, C.; Zhang, X.; Coleman, R.L.; Lopez-Berestein, G.; et al. Preclinical Mammalian Safety Studies of EPHARNA (DOPC Nanoliposomal EphA2-Targeted siRNA). Mol. Cancer Ther. 2017, 16, 1114–1123. [Google Scholar] [CrossRef] [PubMed]
  109. Zheng, Y.S.; Chen, M.L.; Lei, W.D.; Zhu, S.L.; You, X.Q.; Liu, Y. NUDT21 knockdown inhibits proliferation and promotes apoptosis of pancreatic ductal adenocarcinoma through EIF2 signaling. Exp. Cell Res. 2020, 395, 112182. [Google Scholar] [CrossRef] [PubMed]
  110. Hong, D.S.; Kang, Y.K.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.L.; Kim, T.Y.; et al. Phase 1 study of MRX34, a liposomal miR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
  111. Reid, G.; Pel, M.E.; Kirschner, M.B.; Cheng, Y.Y.; Mugridge, N.; Weiss, J.; Williams, M.; Wright, C.; Edelman, J.J.; Vallely, M.P.; et al. Restoring expression of miR-16: A novel approach to therapy for malignant pleural mesothelioma. Ann. Oncol. 2013, 24, 3128–3135. [Google Scholar] [CrossRef]
  112. van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396. [Google Scholar] [CrossRef] [PubMed]
  113. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef] [PubMed]
  114. Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef] [PubMed]
  115. Swann, J.B.; Smyth, M.J. Immune surveillance of tumors. J. Clin. Investig. 2007, 117, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
  116. Kubli, S.P.; Berger, T.; Araujo, D.V.; Siu, L.L.; Mak, T.W. Beyond immune checkpoint blockade: Emerging immunological strategies. Nat. Rev. Drug Discov. 2021, 20, 899–919. [Google Scholar] [CrossRef]
  117. Chen, X.; Song, E. The theory of tumor ecosystem. Cancer Commun. 2022, 42, 587–608. [Google Scholar] [CrossRef] [PubMed]
  118. Bule, P.; Aguiar, S.I.; Aires-Da-Silva, F.; Dias, J.N.R. Chemokine-Directed Tumor Microenvironment Modulation in Cancer Immunotherapy. Int. J. Mol. Sci. 2021, 22, 9804. [Google Scholar] [CrossRef]
  119. Wang, M.; Zhao, J.; Zhang, L.; Wei, F.; Lian, Y.; Wu, Y.; Gong, Z.; Zhang, S.; Zhou, J.; Cao, K.; et al. Role of tumor microenvironment in tumorigenesis. J. Cancer 2017, 8, 761–773. [Google Scholar] [CrossRef]
  120. Whiteside, T.L. Immune suppression in cancer: Effects on immune cells, mechanisms and future therapeutic intervention. Semin. Cancer Biol. 2006, 16, 3–15. [Google Scholar] [CrossRef]
  121. Mihm, M.C., Jr.; Clemente, C.G.; Cascinelli, N. Tumor infiltrating lymphocytes in lymph node melanoma metastases: A histopathologic prognostic indicator and an expression of local immune response. Lab. Investig. 1996, 74, 43–47. [Google Scholar]
  122. Angell, H.; Galon, J. From the immune contexture to the Immunoscore: The role of prognostic and predictive immune markers in cancer. Curr. Opin. Immunol. 2013, 25, 261–267. [Google Scholar] [CrossRef]
  123. Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef] [PubMed]
  124. Ma, J.; Liu, L.; Che, G.; Yu, N.; Dai, F.; You, Z. The M1 form of tumor-associated macrophages in non-small cell lung cancer is positively associated with survival time. BMC Cancer 2010, 10, 112. [Google Scholar] [CrossRef] [PubMed]
  125. Bottcher, J.P.; Reis e Sousa, C. The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity. Trends Cancer 2018, 4, 784–792. [Google Scholar] [CrossRef] [PubMed]
  126. Oshi, M.; Asaoka, M.; Tokumaru, Y.; Yan, L.; Matsuyama, R.; Ishikawa, T.; Endo, I.; Takabe, K. CD8 T Cell Score as a Prognostic Biomarker for Triple Negative Breast Cancer. Int. J. Mol. Sci. 2020, 21, 6968. [Google Scholar] [CrossRef] [PubMed]
  127. Tucci, M.; Passarelli, A.; Mannavola, F.; Felici, C.; Stucci, L.S.; Cives, M.; Silvestris, F. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front. Oncol. 2019, 9, 1148. [Google Scholar] [CrossRef] [PubMed]
  128. Maier, B.; Leader, A.M.; Chen, S.T.; Tung, N.; Chang, C.; LeBerichel, J.; Chudnovskiy, A.; Maskey, S.; Walker, L.; Finnigan, J.P.; et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 2020, 580, 257–262. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, C.; Yang, M.; Zhang, D.; Chen, M.; Zhu, D. Clinical cancer immunotherapy: Current progress and prospects. Front. Immunol. 2022, 13, 961805. [Google Scholar] [CrossRef] [PubMed]
  130. Petitprez, F.; Meylan, M.; de Reynies, A.; Sautes-Fridman, C.; Fridman, W.H. The Tumor Microenvironment in the Response to Immune Checkpoint Blockade Therapies. Front. Immunol. 2020, 11, 784. [Google Scholar] [CrossRef]
  131. Kankeu Fonkoua, L.A.; Sirpilla, O.; Sakemura, R.; Siegler, E.L.; Kenderian, S.S. CAR T cell therapy and the tumor microenvironment: Current challenges and opportunities. Mol. Ther. Oncolyt. 2022, 25, 69–77. [Google Scholar] [CrossRef]
  132. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
  133. Alahdal, M.; Elkord, E. Non-coding RNAs in cancer immunotherapy: Predictive biomarkers and targets. Clin. Transl. Med. 2023, 13, e1425. [Google Scholar] [CrossRef]
  134. Leidinger, P.; Backes, C.; Dahmke, I.N.; Galata, V.; Huwer, H.; Stehle, I.; Bals, R.; Keller, A.; Meese, E. What makes a blood cell based miRNA expression pattern disease specific?—A miRNome analysis of blood cell subsets in lung cancer patients and healthy controls. Oncotarget 2014, 5, 9484–9497. [Google Scholar] [CrossRef]
  135. Afra, F.; Mahboobipour, A.A.; Salehi Farid, A.; Ala, M. Recent progress in the immunotherapy of hepatocellular carcinoma: Non-coding RNA-based immunotherapy may improve the outcome. Biomed. Pharmacother. 2023, 165, 115104. [Google Scholar] [CrossRef]
  136. Wang, J.; Li, S.; Wang, T.; Xu, S.; Wang, X.; Kong, X.; Lu, X.; Zhang, H.; Li, L.; Feng, M.; et al. RNA2Immune: A Database of Experimentally Supported Data Linking Non-coding RNA Regulation to The Immune System. Genom. Proteom. Bioinform. 2023, 21, 283–291. [Google Scholar] [CrossRef]
  137. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef]
  138. Wu, K.; Lin, K.; Li, X.; Yuan, X.; Xu, P.; Ni, P.; Xu, D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front. Immunol. 2020, 11, 1731. [Google Scholar] [CrossRef]
  139. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef]
  140. Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018, 233, 6425–6440. [Google Scholar] [CrossRef]
  141. Kadomoto, S.; Izumi, K.; Mizokami, A. Macrophage Polarity and Disease Control. Int. J. Mol. Sci. 2021, 23, 144. [Google Scholar] [CrossRef]
  142. Cassetta, L.; Pollard, J.W. Targeting macrophages: Therapeutic approaches in cancer. Nat. Rev. Drug Discov. 2018, 17, 887–904. [Google Scholar] [CrossRef]
  143. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090. [Google Scholar] [CrossRef]
  144. Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar] [CrossRef]
  145. Mantovani, A.; Allavena, P.; Marchesi, F.; Garlanda, C. Macrophages as tools and targets in cancer therapy. Nat. Rev. Drug Discov. 2022, 21, 799–820. [Google Scholar] [CrossRef]
  146. Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef]
  147. Li, M.; Wang, M.; Wen, Y.; Zhang, H.; Zhao, G.N.; Gao, Q. Signaling pathways in macrophages: Molecular mechanisms and therapeutic targets. MedComm 2023, 4, e349. [Google Scholar] [CrossRef]
  148. Watford, W.T.; Moriguchi, M.; Morinobu, A.; O’Shea, J.J. The biology of IL-12: Coordinating innate and adaptive immune responses. Cytokine Growth Factor Rev. 2003, 14, 361–368. [Google Scholar] [CrossRef]
  149. Tait Wojno, E.D.; Hunter, C.A.; Stumhofer, J.S. The Immunobiology of the Interleukin-12 Family: Room for Discovery. Immunity 2019, 50, 851–870. [Google Scholar] [CrossRef]
  150. Seok, S.H.; Heo, J.I.; Hwang, J.H.; Na, Y.R.; Yun, J.H.; Lee, E.H.; Park, J.W.; Cho, C.H. Angiopoietin-1 elicits pro-inflammatory responses in monocytes and differentiating macrophages. Mol. Cells 2013, 35, 550–556. [Google Scholar] [CrossRef]
  151. Wang, X.; Zhang, J.; Hu, B.; Qian, F. High Expression of CSF-1R Predicts Poor Prognosis and CSF-1R(high) Tumor-Associated Macrophages Inhibit Anti-Tumor Immunity in Colon Adenocarcinoma. Front. Oncol. 2022, 12, 850767. [Google Scholar] [CrossRef]
  152. Koh, Y.W.; Park, C.; Yoon, D.H.; Suh, C.; Huh, J. CSF-1R expression in tumor-associated macrophages is associated with worse prognosis in classical Hodgkin lymphoma. Am. J. Clin. Pathol. 2014, 141, 573–583. [Google Scholar] [CrossRef]
  153. Coffelt, S.B.; Tal, A.O.; Scholz, A.; De Palma, M.; Patel, S.; Urbich, C.; Biswas, S.K.; Murdoch, C.; Plate, K.H.; Reiss, Y.; et al. Angiopoietin-2 regulates gene expression in TIE2-expressing monocytes and augments their inherent proangiogenic functions. Cancer Res. 2010, 70, 5270–5280. [Google Scholar] [CrossRef]
  154. Shi, L.; Bian, Z.; Kidder, K.; Liang, H.; Liu, Y. Non-Lyn Src Family Kinases Activate SIRPalpha-SHP-1 to Inhibit PI3K-Akt2 and Dampen Proinflammatory Macrophage Polarization. J. Immunol. 2021, 207, 1419–1427. [Google Scholar] [CrossRef]
  155. Kerneur, C.; Cano, C.E.; Olive, D. Major pathways involved in macrophage polarization in cancer. Front. Immunol. 2022, 13, 1026954. [Google Scholar] [CrossRef]
  156. Chen, H.M.; van der Touw, W.; Wang, Y.S.; Kang, K.; Mai, S.; Zhang, J.; Alsina-Beauchamp, D.; Duty, J.A.; Mungamuri, S.K.; Zhang, B.; et al. Blocking immunoinhibitory receptor LILRB2 reprograms tumor-associated myeloid cells and promotes antitumor immunity. J. Clin. Investig. 2018, 128, 5647–5662. [Google Scholar] [CrossRef]
  157. van der Touw, W.; Chen, H.M.; Pan, P.Y.; Chen, S.H. LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol. Immunother. 2017, 66, 1079–1087. [Google Scholar] [CrossRef]
  158. Shi, J.H.; Sun, S.C. Tumor Necrosis Factor Receptor-Associated Factor Regulation of Nuclear Factor kappaB and Mitogen-Activated Protein Kinase Pathways. Front. Immunol. 2018, 9, 1849. [Google Scholar] [CrossRef]
  159. Aggarwal, B.B. Signalling pathways of the TNF superfamily: A double-edged sword. Nat. Rev. Immunol. 2003, 3, 745–756. [Google Scholar] [CrossRef]
  160. Hu, X.; Herrero, C.; Li, W.P.; Antoniv, T.T.; Falck-Pedersen, E.; Koch, A.E.; Woods, J.M.; Haines, G.K.; Ivashkiv, L.B. Sensitization of IFN-gamma Jak-STAT signaling during macrophage activation. Nat. Immunol. 2002, 3, 859–866. [Google Scholar] [CrossRef]
  161. Kawai, T.; Akira, S. Signaling to NF-kappaB by Toll-like receptors. Trends Mol. Med. 2007, 13, 460–469. [Google Scholar] [CrossRef]
  162. Kawai, T.; Akira, S. TLR signaling. Semin. Immunol. 2007, 19, 24–32. [Google Scholar] [CrossRef] [PubMed]
  163. Lehtonen, A.; Matikainen, S.; Miettinen, M.; Julkunen, I. Granulocyte-macrophage colony-stimulating factor (GM-CSF)-induced STAT5 activation and target-gene expression during human monocyte/macrophage differentiation. J. Leukoc. Biol. 2002, 71, 511–519. [Google Scholar] [CrossRef] [PubMed]
  164. Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef]
  165. Scott, T.E.; Lewis, C.V.; Zhu, M.; Wang, C.; Samuel, C.S.; Drummond, G.R.; Kemp-Harper, B.K. IL-4 and IL-13 induce equivalent expression of traditional M2 markers and modulation of reactive oxygen species in human macrophages. Sci. Rep. 2023, 13, 19589. [Google Scholar] [CrossRef] [PubMed]
  166. Wang, S.W.; Sun, Y.M. The IL-6/JAK/STAT3 pathway: Potential therapeutic strategies in treating colorectal cancer (Review). Int. J. Oncol. 2014, 44, 1032–1040. [Google Scholar] [CrossRef] [PubMed]
  167. Saxton, R.A.; Tsutsumi, N.; Su, L.L.; Abhiraman, G.C.; Mohan, K.; Henneberg, L.T.; Aduri, N.G.; Gati, C.; Garcia, K.C. Structure-based decoupling of the pro- and anti-inflammatory functions of interleukin-10. Science 2021, 371. [Google Scholar] [CrossRef]
  168. Batlle, E.; Massague, J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
  169. Segura, E. Human dendritic cell subsets: An updated view of their ontogeny and functional specialization. Eur. J. Immunol. 2022, 52, 1759–1767. [Google Scholar] [CrossRef]
  170. Gerhard, G.M.; Bill, R.; Messemaker, M.; Klein, A.M.; Pittet, M.J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 2021, 218, e20200264. [Google Scholar] [CrossRef]
  171. See, P.; Dutertre, C.A.; Chen, J.; Gunther, P.; McGovern, N.; Irac, S.E.; Gunawan, M.; Beyer, M.; Handler, K.; Duan, K.; et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science 2017, 356, eaag3009. [Google Scholar] [CrossRef]
  172. Cabeza-Cabrerizo, M.; van Blijswijk, J.; Wienert, S.; Heim, D.; Jenkins, R.P.; Chakravarty, P.; Rogers, N.; Frederico, B.; Acton, S.; Beerling, E.; et al. Tissue clonality of dendritic cell subsets and emergency DCpoiesis revealed by multicolor fate mapping of DC progenitors. Sci. Immunol. 2019, 4, eaaw1941. [Google Scholar] [CrossRef]
  173. Dumitriu, I.E.; Dunbar, D.R.; Howie, S.E.; Sethi, T.; Gregory, C.D. Human dendritic cells produce TGF-beta 1 under the influence of lung carcinoma cells and prime the differentiation of CD4+CD25+Foxp3+ regulatory T cells. J. Immunol. 2009, 182, 2795–2807. [Google Scholar] [CrossRef]
  174. Troy, A.J.; Summers, K.L.; Davidson, P.J.; Atkinson, C.H.; Hart, D.N. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin. Cancer Res. 1998, 4, 585–593. [Google Scholar]
  175. Enk, A.H.; Jonuleit, H.; Saloga, J.; Knop, J. Dendritic cells as mediators of tumor-induced tolerance in metastatic melanoma. Int. J. Cancer 1997, 73, 309–316. [Google Scholar] [CrossRef]
  176. Nestle, F.O.; Burg, G.; Fah, J.; Wrone-Smith, T.; Nickoloff, B.J. Human sunlight-induced basal-cell-carcinoma-associated dendritic cells are deficient in T cell co-stimulatory molecules and are impaired as antigen-presenting cells. Am. J. Pathol. 1997, 150, 641–651. [Google Scholar] [PubMed]
  177. Chaux, P.; Favre, N.; Martin, M.; Martin, F. Tumor-infiltrating dendritic cells are defective in their antigen-presenting function and inducible B7 expression in rats. Int. J. Cancer 1997, 72, 619–624. [Google Scholar] [CrossRef]
  178. Bonifaz, L.; Bonnyay, D.; Mahnke, K.; Rivera, M.; Nussenzweig, M.C.; Steinman, R.M. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 2002, 196, 1627–1638. [Google Scholar] [CrossRef]
  179. Gabrilovich, D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat. Rev. Immunol. 2004, 4, 941–952. [Google Scholar] [CrossRef] [PubMed]
  180. DeVito, N.C.; Plebanek, M.P.; Theivanthiran, B.; Hanks, B.A. Role of Tumor-Mediated Dendritic Cell Tolerization in Immune Evasion. Front. Immunol. 2019, 10, 2876. [Google Scholar] [CrossRef]
  181. Scarlett, U.K.; Rutkowski, M.R.; Rauwerdink, A.M.; Fields, J.; Escovar-Fadul, X.; Baird, J.; Cubillos-Ruiz, J.R.; Jacobs, A.C.; Gonzalez, J.L.; Weaver, J.; et al. Ovarian cancer progression is controlled by phenotypic changes in dendritic cells. J. Exp. Med. 2012, 209, 495–506. [Google Scholar] [CrossRef]
  182. Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 1996, 2, 1096–1103. [Google Scholar] [CrossRef]
  183. Lund, A.W.; Duraes, F.V.; Hirosue, S.; Raghavan, V.R.; Nembrini, C.; Thomas, S.N.; Issa, A.; Hugues, S.; Swartz, M.A. VEGF-C promotes immune tolerance in B16 melanomas and cross-presentation of tumor antigen by lymph node lymphatics. Cell Rep. 2012, 1, 191–199. [Google Scholar] [CrossRef]
  184. Su, X.W.; Lu, G.; Leung, C.K.; Liu, Q.; Li, Y.; Tsang, K.S.; Zhao, S.D.; Chan, D.T.M.; Kung, H.F.; Poon, W.S. miR-181d regulates human dendritic cell maturation through NF-kappaB pathway. Cell Prolif. 2017, 50. [Google Scholar] [CrossRef]
  185. Pfeiffer, I.A.; Hoyer, S.; Gerer, K.F.; Voll, R.E.; Knippertz, I.; Guckel, E.; Schuler, G.; Schaft, N.; Dorrie, J. Triggering of NF-kappaB in cytokine-matured human DCs generates superior DCs for T-cell priming in cancer immunotherapy. Eur. J. Immunol. 2014, 44, 3413–3428. [Google Scholar] [CrossRef]
  186. Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
  187. Balan, S.; Radford, K.J.; Bhardwaj, N. Unexplored horizons of cDC1 in immunity and tolerance. Adv. Immunol. 2020, 148, 49–91. [Google Scholar] [CrossRef]
  188. Boks, M.A.; Kager-Groenland, J.R.; Haasjes, M.S.; Zwaginga, J.J.; van Ham, S.M.; ten Brinke, A. IL-10-generated tolerogenic dendritic cells are optimal for functional regulatory T cell induction—A comparative study of human clinical-applicable DC. Clin. Immunol. 2012, 142, 332–342. [Google Scholar] [CrossRef]
  189. Kobie, J.J.; Wu, R.S.; Kurt, R.A.; Lou, S.; Adelman, M.K.; Whitesell, L.J.; Ramanathapuram, L.V.; Arteaga, C.L.; Akporiaye, E.T. Transforming growth factor beta inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines. Cancer Res. 2003, 63, 1860–1864. [Google Scholar]
  190. Steinbrink, K.; Wolfl, M.; Jonuleit, H.; Knop, J.; Enk, A.H. Induction of tolerance by IL-10-treated dendritic cells. J. Immunol. 1997, 159, 4772–4780. [Google Scholar] [CrossRef] [PubMed]
  191. Kudo-Saito, C.; Shirako, H.; Ohike, M.; Tsukamoto, N.; Kawakami, Y. CCL2 is critical for immunosuppression to promote cancer metastasis. Clin. Exp. Metastasis 2013, 30, 393–405. [Google Scholar] [CrossRef] [PubMed]
  192. Liu, Q.; Zhang, C.; Sun, A.; Zheng, Y.; Wang, L.; Cao, X. Tumor-educated CD11bhighIalow regulatory dendritic cells suppress T cell response through arginase I. J. Immunol. 2009, 182, 6207–6216. [Google Scholar] [CrossRef] [PubMed]
  193. Norian, L.A.; Rodriguez, P.C.; O’Mara, L.A.; Zabaleta, J.; Ochoa, A.C.; Cella, M.; Allen, P.M. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res. 2009, 69, 3086–3094. [Google Scholar] [CrossRef] [PubMed]
  194. Gabrilovich, D.I.; Bronte, V.; Chen, S.H.; Colombo, M.P.; Ochoa, A.; Ostrand-Rosenberg, S.; Schreiber, H. The terminology issue for myeloid-derived suppressor cells. Cancer Res. 2007, 67, 425. [Google Scholar] [CrossRef] [PubMed]
  195. Bronte, V.; Brandau, S.; Chen, S.H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [PubMed]
  196. Movahedi, K.; Guilliams, M.; Van den Bossche, J.; Van den Bergh, R.; Gysemans, C.; Beschin, A.; De Baetselier, P.; Van Ginderachter, J.A. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood 2008, 111, 4233–4244. [Google Scholar] [CrossRef] [PubMed]
  197. Lee, C.R.; Lee, W.; Cho, S.K.; Park, S.G. Characterization of Multiple Cytokine Combinations and TGF-beta on Differentiation and Functions of Myeloid-Derived Suppressor Cells. Int. J. Mol. Sci. 2018, 19, 869. [Google Scholar] [CrossRef]
  198. Lechner, M.G.; Liebertz, D.J.; Epstein, A.L. Characterization of cytokine-induced myeloid-derived suppressor cells from normal human peripheral blood mononuclear cells. J. Immunol. 2010, 185, 2273–2284. [Google Scholar] [CrossRef]
  199. Veglia, F.; Sanseviero, E.; Gabrilovich, D.I. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat. Rev. Immunol. 2021, 21, 485–498. [Google Scholar] [CrossRef]
  200. Rossowska, J.; Anger, N.; Szczygiel, A.; Mierzejewska, J.; Pajtasz-Piasecka, E. Reprogramming the murine colon cancer microenvironment using lentivectors encoding shRNA against IL-10 as a component of a potent DC-based chemoimmunotherapy. J. Exp. Clin. Cancer Res. 2018, 37, 126. [Google Scholar] [CrossRef]
  201. Cubillos-Ruiz, J.R.; Engle, X.; Scarlett, U.K.; Martinez, D.; Barber, A.; Elgueta, R.; Wang, L.; Nesbeth, Y.; Durant, Y.; Gewirtz, A.T.; et al. Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J. Clin. Investig. 2009, 119, 2231–2244. [Google Scholar] [CrossRef]
  202. Yuan, R.; Li, S.; Geng, H.; Wang, X.; Guan, Q.; Li, X.; Ren, C.; Yuan, X. Reversing the polarization of tumor-associated macrophages inhibits tumor metastasis. Int. Immunopharmacol. 2017, 49, 30–37. [Google Scholar] [CrossRef] [PubMed]
  203. Zhang, F.; Parayath, N.N.; Ene, C.I.; Stephan, S.B.; Koehne, A.L.; Coon, M.E.; Holland, E.C.; Stephan, M.T. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat. Commun. 2019, 10, 3974. [Google Scholar] [CrossRef] [PubMed]
  204. Sun, Y.; Cronin, M.F.; Mendonca, M.C.P.; Guo, J.; O’Driscoll, C.M. M2pep-Modified Cyclodextrin-siRNA Nanoparticles Modulate the Immunosuppressive Tumor Microenvironment for Prostate Cancer Therapy. Mol. Pharm. 2023, 20, 5921–5936. [Google Scholar] [CrossRef]
  205. Shobaki, N.; Sato, Y.; Suzuki, Y.; Okabe, N.; Harashima, H. Manipulating the function of tumor-associated macrophages by siRNA-loaded lipid nanoparticles for cancer immunotherapy. J. Control. Release 2020, 325, 235–248. [Google Scholar] [CrossRef]
  206. Qian, H.; Zhou, T.; Fu, Y.; Guo, M.; Yang, W.; Zhang, D.; Fang, W.; Yao, M.; Shi, H.; Chai, C.; et al. Self-assembled tetrahedral framework nucleic acid mediates tumor-associated macrophage reprogramming and restores antitumor immunity. Mol. Ther. Nucleic Acids 2022, 27, 763–773. [Google Scholar] [CrossRef]
  207. Troegeler, A.; Lastrucci, C.; Duval, C.; Tanne, A.; Cougoule, C.; Maridonneau-Parini, I.; Neyrolles, O.; Lugo-Villarino, G. An efficient siRNA-mediated gene silencing in primary human monocytes, dendritic cells and macrophages. Immunol. Cell Biol. 2014, 92, 699–708. [Google Scholar] [CrossRef]
  208. Cheng, P.; Kumar, V.; Liu, H.; Youn, J.I.; Fishman, M.; Sherman, S.; Gabrilovich, D. Effects of notch signaling on regulation of myeloid cell differentiation in cancer. Cancer Res. 2014, 74, 141–152. [Google Scholar] [CrossRef] [PubMed]
  209. Triozzi, P.; Kooshki, M.; Alistar, A.; Bitting, R.; Neal, A.; Lametschwandtner, G.; Loibner, H. Phase I clinical trial of adoptive cellular immunotherapy with APN401 in patients with solid tumors. J. Immunother. Cancer 2015, 3, P175. [Google Scholar] [CrossRef]
  210. Kortylewski, M.; Swiderski, P.; Herrmann, A.; Wang, L.; Kowolik, C.; Kujawski, M.; Lee, H.; Scuto, A.; Liu, Y.; Yang, C.; et al. In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses. Nat. Biotechnol. 2009, 27, 925–932. [Google Scholar] [CrossRef]
  211. Zhang, C.; Huang, R.; Ren, L.; Song, J.; Kortylewski, M.; Swiderski, P.; Forman, S.; Yu, H. Local CpG- Stat3 siRNA treatment improves antitumor effects of immune checkpoint inhibitors. bioRxiv 2023. [Google Scholar] [CrossRef]
Figure 2. Overview of mechanisms of actions of anti-sense oligos (ASOs) within the nucleus. (a) DNA ASOs bind to mRNA within the nucleus to form DNA-RNA hybrids, which are recognized and cleaved by the enzyme RNaseH. (b) ASO can mediate exon skipping by jointing two exons that are not adjacent. (c) ASOs can mediate exon retention by binding to adjacent exons and thereby retaining the exon within the mRNA processing.
Figure 2. Overview of mechanisms of actions of anti-sense oligos (ASOs) within the nucleus. (a) DNA ASOs bind to mRNA within the nucleus to form DNA-RNA hybrids, which are recognized and cleaved by the enzyme RNaseH. (b) ASO can mediate exon skipping by jointing two exons that are not adjacent. (c) ASOs can mediate exon retention by binding to adjacent exons and thereby retaining the exon within the mRNA processing.
Ijms 25 07275 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Binder, A.K.; Bremm, F.; Dörrie, J.; Schaft, N. Non-Coding RNA in Tumor Cells and Tumor-Associated Myeloid Cells—Function and Therapeutic Potential. Int. J. Mol. Sci. 2024, 25, 7275. https://doi.org/10.3390/ijms25137275

AMA Style

Binder AK, Bremm F, Dörrie J, Schaft N. Non-Coding RNA in Tumor Cells and Tumor-Associated Myeloid Cells—Function and Therapeutic Potential. International Journal of Molecular Sciences. 2024; 25(13):7275. https://doi.org/10.3390/ijms25137275

Chicago/Turabian Style

Binder, Amanda Katharina, Franziska Bremm, Jan Dörrie, and Niels Schaft. 2024. "Non-Coding RNA in Tumor Cells and Tumor-Associated Myeloid Cells—Function and Therapeutic Potential" International Journal of Molecular Sciences 25, no. 13: 7275. https://doi.org/10.3390/ijms25137275

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop