Next Article in Journal
NS-11021 Modulates Cancer-Associated Processes Independently of BK Channels in Melanoma and Pancreatic Duct Adenocarcinoma Cell Lines
Next Article in Special Issue
Antagonistic Roles of the Tumor Suppressor miR-210-3p and Oncomucin MUC4 Forming a Negative Feedback Loop in Pancreatic Adenocarcinoma
Previous Article in Journal
Serum Sorbitol Dehydrogenase as a Novel Prognostic Factor for Hepatocellular Carcinoma after Surgical Resection
Previous Article in Special Issue
MicroRNAs: Their Role in Metabolism, Tumor Microenvironment, and Therapeutic Implications in Head and Neck Squamous Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

miRNAs in the Regulation of Cancer Immune Response: Effect of miRNAs on Cancer Immunotherapy

by
Faheem Hyder Pottoo
1,
Ashif Iqubal
2,3,
Mohammad Kashif Iqubal
4,5,
Mohammed Salahuddin
6,
Jawad Ur Rahman
7,
Noora AlHajri
8,* and
Mustafa Shehadeh
9,*
1
Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
2
Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
3
Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
4
Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
5
Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd., Gurugram 122001, India
6
Department of Clinical Pharmacy Research, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
7
Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
8
Mayo Clinic, Sheikh Shakhbout Medical City (SSMC), Abu Dhabi 127788, United Arab Emirates
9
College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
*
Authors to whom correspondence should be addressed.
Submission received: 24 October 2021 / Revised: 22 November 2021 / Accepted: 24 November 2021 / Published: 6 December 2021
(This article belongs to the Collection miRNAs: New Insights in Tumor Biology)

Abstract

:

Simple Summary

Cancer is the deadliest disease affecting humans, with more than 14 million cases and 8 million deaths. A successful therapeutic interception of cancer is urgently required. We have reviewed the immunologic involvement of various types of cancers that are well substantiated in the existing literature. Interestingly, multiple signalling pathways of the immune system could be modulated with miRNAs (noncoding RNAs of 22 nucleotides), such as miR-19a-3p, which induces macrophage polarization and the amelioration of cancer progression and metastasis. Developing a specific profile of miRNA within immune cells could also help in diagnosis and treatment. miRNA-based immune therapeutics that help to reduce cancer immune escape hold possibilities for developing cancer chemotherapy.

Abstract

In the last few decades, carcinogenesis has been extensively explored and substantial research has identified immunogenic involvement in various types of cancers. As a result, immune checkpoint blockers and other immune-based therapies were developed as novel immunotherapeutic strategies. However, despite being a promising therapeutic option, immunotherapy has significant constraints such as a high cost of treatment, unpredictable toxicity, and clinical outcomes. miRNAs are non-coding, small RNAs actively involved in modulating the immune system’s multiple signalling pathways by binding to the 3′-UTR of target genes. miRNAs possess a unique advantage in modulating multiple targets of either the same or different signalling pathways. Therefore, miRNA follows a ‘one drug multiple target’ hypothesis. Attempts are made to explore the therapeutic promise of miRNAs in cancer so that it can be transported from bench to bedside for successful immunotherapeutic results. Therefore, in the current manuscript, we discussed, in detail, the mechanism and role of miRNAs in different types of cancers relating to the immune system, its diagnostic and therapeutic aspect, the effect on immune escape, immune-checkpoint molecules, and the tumour microenvironment. We have also discussed the existing limitations, clinical success and the prospective use of miRNAs in cancer.

1. Introduction

According to the World Cancer report 2020, Cancer is the 1st –2nd cause of premature death in 134 out of 183 countries and ranks as the 3rd–4th cause in 45 of the remaining countries. (WHO World cancer report 2020). It has been estimated that by 2030, cancer prevalence might increase to around 26 million and will lead to 17 million deaths [1]. Around 1.9 million people were diagnosed with cancers in the USA alone, which were statistically expected to lead to 0.61M deaths in 2021 [2,3]. The disease presents serious threats and challenges due to its ability to cause tissue damage and difficulties in diagnosis and prospective treatment [4,5,6]. Cancer leads to a devasting effect on patients’ quality of life and leads to a devasting effect on patients’ quality of life and imposes a serious economic burden on nations [1]. Considering the aetiology of tumorigenesis, a successive mutation in response to internal as well as external stimuli in genes or DNA leads to abnormal changes in the cell functions and gradually causes an imbalance between cell division and apoptosis, leading to the initiation and progression of tumorigenesis. Various factors such as smoking (carcinogen), environmentally hazardous chemicals, genetic factors, viruses, bacteria, and radiation influence the cytoplasm and nucleus, ultimately causing genetic disorders and gene mutations, leading to cancer [7,8,9,10].
Currently, cancer treatment relies on various factors such as phenotype, tumour size, tumor volume, localization, stage of spread, mutagenic analysis, coexisting disease condition, the physical status of patients and any possible side of anti-cancer drugs. It also depends on treatment approaches of combining two or more therapies such as chemotherapy, radiotherapy, hormone therapy, etc. However, these approaches are not sufficient to treat cancer in the advanced stage, ultimately leading to metastasis and death [11]. The available therapeutic regimen leads to serious adverse effects with economic burden and poor patients’ quality of life. Studies have shown the involvement of various altered signalling molecules and pathways in the pathogenesis of tumors. In the past, several significant achievements in the field of research and development of anti-cancer drugs and their derivatives have been made, especially in treatment and diagnosis [12,13,14,15]. Apart from the role of drugs in the treatment of cancer, a timely diagnosis also plays a pivotal role in the management of various types of tumours. With the advancement in techniques, various cellular and molecular diagnostic markers have been developed, but cancer’s complexity presents a significant challenge in determining the accurate diagnosis [16,17]. The “omics” revolution has highlighted the complex nature of cancer signalling pathways, types of molecules, and cancer alterations, due to the greater capacity of noncoding MicroRNAs (miRNAs) to alter expression. Various studies have reported miRNAs’ ability to express themselves differentially in multiple types of cancer [18,19,20].
The modulation of miRNA expression is reported to be related to oncogenic miRNA inhibition and the substitution of tumour-suppressive miRNAs and, hence, have been found to be a perfect tool for cancer therapy improvement. The association of miRNA expressions with cancer and other problems has attracted interest in developing a biomarker for diagnosis, detection, and treatment [21,22,23]. miRNAs have been reported to play a protective role as well as a pathogenic role in carcinogenesis. Therefore, it becomes important to selectively identify the exact role of miRNA and, accordingly, miRNA-based therapeutics can be designed and developed. Research is currently underway concerning the development of miRNA-based diagnostic and therapeutic tools for the timely management and treatment of cancer. Thus, the present review mainly focuses on extracellular miRNA’s biogenesis, functions, and role as a biomarker in diagnosing and treating various cancers.

2. miRNAs—Biogenesis, Biochemistry and Functions

It has been reported that DNA acts as an immediate precursor to miRNAs. Initially, miRNAs (pri-miRNAs) are primary are produced and later converted into precursor miRNAs (pre-miRNAs) then mature miRNAs [17,24]. This process begins with the biogenesis of canonical miRNAs in the nucleus due to the activation of polymerase II, leading to primary miRNA transcript (pri-miRNAs) formation outside of the genome non-coding fraction. These primary miRNAs are trims that are processed by a microprocessor with an endonuclease III enzyme and binding protein (DiGeorge syndrome), liberating pre-miRNAs hairpin-like structure [25,26,27]. Within pri-miRNA, DGCR8 binds to pri-miRNA at N6- methyladenylated GGAC and other motifs, whereas Drosha cleaves to the pri-miRNA duplex at the base of the hairpin structure of pri-miRNA to produce precursor miRNA (pre-miRNA). These generated pre-miRNAs are carried to the cytoplasm by Exportin 5 and Ran GTPase complex and then processed by RNase III endonuclease. The direction of the miRNA strand determines the name of the miRNA. Like 5P from the 5′ end of and 3p from the 3′ end of the pre-miRNA hairpin [28,29,30]. In the maturation stage, Enzyme RNase III endonuclease in the cytoplasm, combined with TRBP and PACT proteins, break it to produce a mature miRNA duplex. Then, the mature single-stranded miRNA (generally lower 5′ stability), along with protein (Argonaute family) Ago, are loaded together with RNA, inducing a silencing complex after which it binds to 3′ UTRs (mRNA targets), causing decapping and deadenylation of the mRNA transcript, thus inhibiting various physiological functions. The unloaded miRNA strand is unwound from the guide strand, referred to as the passenger strand [31,32]. After coupling RISC with miRNA, it silences the posttranscriptional modification by tethering to become partially compatible with mRNA within the 3’untranslated region found in the UTR. Binding at seed sites or at the non-canonical seedless site of the miRNA-mRNA linkage network is complex as one miRNA affects multiple genes. One mRNA might be targeted by numerous related or unrelated miRNAs [33,34]. It has been reported that one miRNA complex can target almost two hundred genes of variable functions [35]. About 60% of human genes can be targeted by these miRNAs, once their corresponding mRNAs targets are regulated by miRNA, influencing various pathways, including cancer [36,37].
Since the identification of tumour-suppressive miRNA gene miR-15a/miR-16-1, the functional significance of miRNAs has been realized, and nearly 2500 miRNAs are recorded in the database of miRBase [38]. These miRNAs are small, non-coding RNA molecules of 22nt (average) in size and are responsible for gene regulation [39], for various biological processes and carcinogenesis [11,40]. miRNAs act as central regulators of gene expression by acting on mRNA or its translation. They are crucial in remedying various human ailments and cancer [41].
Several researchers have reported the location of most miRNAs’ genes at cancer-related genomic regions known as fragile sites. The expression data of cancer cells shows that these miRNAs are expressed by rule rather than the exception [42,43]. miRNAs play an important role and have been reported to be involved in lung, colon, breast, and other cancers. Most researchers have reported that miRNAs exhibit biochemical and molecular effects by targeting multiple mRNAs, and a few of these mRNAs are present in the cellular pathway. A few studies have reported that these mRNA were redundant and repressed the same mRNA target. In the animal model, the link between miRNA and cancer is detected by overexpressing miRNA features [43,44].
mRNAs functions have been categorized as homeostatic regulation of gene expression and cellular responses robustness. These two factors play important functions by fine-tuning translation and cell-fate decisions through complex reciprocal negative-feedback loops. Stress requires a robust response from miRNA, which may act as switches and help adapt to microenvironment changes [11,45]. This function can be best observed in glioblastoma, where glucose level reduction causes a reduction of miR-451 levels. When sufficient energy is available, the elevated level of miR-451 suppresses the cell signalling pathway and stimulates the activation of the mammalian target of rapamycin and cell proliferation [46,47]. Every tumour has its own miRNA sign, which can be helpful in differentiating these from other healthy tissues and in identifying the different types of cancer. Based on these miRNA signs, most cancers can be divided into prognostic groups. The functions of amplification or deletion, methylation, and transcription factors are responsible for altering the MicroRNA expression [47,48].

2.1. Oncogenic miRNAs

Many miRNAs have been reported to be involved in various biological functions, including gene regulation. Variations in the expression of these miRNAs were found to be responsible for causing different diseases, including cancer. Specifically, changes in miRNA levels with oncogenic or cancer suppression properties are related to carcinogenesis, metastasis, and the anticancer responses to drugs and therapies. Intergenic regions between genes possess several miRNA sequences. In some cases, miRNAs that are located close to each other are controlled by the same promoter and play an important role in more than one cancer by acting through various pathways. Their expression results in the formation of a multiple–looped polycistronic sequence from which multiple mature miRNA are synthesized [49,50,51,52]. These miRNAs are termed as “oncomirs” or “oncomiR” and regulate the progression of the tumour (Table 1). For example, miR-21 acts as an oncogene and suppresses TPM1 and PDCD4 (tumor genes) by downregulation in high proliferated breast cancer [49,53,54]. Cancer is caused due to imbalances in the cell cycle. Different mechanisms such as cell cycle regulation, detection, and damage repair control these imbalances in regulatory pathways. Cyclins and cyclin-dependent kinases (CDKs) are regulatory proteins and are responsible for cell progression in the cell cycle [24,53].
Several studies support the function of miRNA as tumour suppressors [55], e.g., miR-34 represses tumour progression through epithelial-mesenchymal transition (EMT) via EMT-transcription factors when dysregulated via the synergistic effect of the p53 tumor suppressor gene and some important signal pathways. Several invitro studies on various cancer cell lines found that through different signalling pathways downregulation of the family of miR-34 can lead to colon, prostate, lung, osteosarcoma, and other types of cancers [54,55,56,57,58,59]. Similarly, Pichiorri et al. found the overexpression of miR-106b∼25 cluster, miR-181a and b and miR-21 in MM cell lines leading to the tumour compared to normal cell controls thus acting as an oncogene when two miRNA cluster. In contrast, miRNA-106b, miRNA-181a and b were found to be deregulated, thus acting as tumour suppressors [58].

2.2. miRNAs as Biomarkers

miRNAs can function as biomarkers to identify various diseases, be found in plasma, serum, semen, and other biological fluids, and even be stable in degrading conditions [81,82,83]. There are two clusters or populations of circulating miRNAs, one cluster is present in vesicles and the other is linked with proteins. Their association with these two locations has been contradicted by researchers who hypothesize that most circulating miRNAs are exosome miRNAs [41,84,85].
The ceramide-dependent pathway is involved in the release of exosomal miRNAs in response to injury or even the cell’s death. It thereby produces regulatory effects in target cells [86]. Interleukin-4 and docosahexaenoic acid regulate the secretion of miRNAs via exosomes. In breast cancer, interleukin-4 activation leads to the secretion of oncogenic miRNAs. It stimulates cancer cell invasiveness whereas, docosahexaenoic acid exhibits anticancer effects by stimulating the exosomal miRNA secretion, thereby producing tumour angiogenesis [83,87,88]. Recent studies have reported exosome miRNAs’, such as miR-105, role in the metastatic breast cancer cell, miR-21-3p from umbilical cord blood, in destructing the barrier function, metastasis and the migration of fibroblasts, leading to the angiogenic action by endothelial cells. miRNAs such as miR-342–3p and miR-1246 from oral cancer are reported to be responsible for inducing metastasis in metastatic cancer cell line [83,89,90,91].
The identification of serum levels of cancer patients was performed using seven miRNAs and found a significant reduction in serum levels of advanced stage astrocytoma’s patients and helps in precisely distinguishing between normal and cancer patients [92,93]. Thus, circulating miRNAs possess a differential expression ability and can act as a potential tool for screening cancer patients without harm. During the early stage, progression and even after the metastasis stage, there abnormal changes in miRNAs levels occur. Therefore, these miRNAs may act as potential biomarkers to differentiate tumours, formulating treatment strategy and prove helpful in the outcomes of the treatment [94,95,96]. miRNAs produce pleiotropic effects and are helpful in the diagnosis and prognosis of patient evaluations (Table 2). Therefore, these miRNAs are recognized as chemical messenger and regulate cellular communications [97,98,99].
In various published reports, a correlation between types of breast cancer was identified relating to the overexpression of miR-21, miR-155 and miR-106a and under-expression of miR-335, miR-126 & miR-199a with the non-tumour samples [83,100,101]. Studies have shown that miR-21 expression is not only able to distinguish between a normal and breast cancer but can also help to differentiate a metastatic patient from a patient with locoregional reoccurrence [102]. A similar study reported the overexpression of miR-21, miR-155, miR-10b in plasma samples in breast cancer patients as compared to normal control. Additionally, the level of the miRNA mentioned above complex was reduced after different treatment approaches [103,104].
Multiple myeloma is a cancer of the bone marrow, affecting various parts of the body. Several studies reported that the association of the disease progression or stages involves circulating miRNA [58], e.g., the combination of miRNA-720 and -1308 can be helpful in distinguishing a Healthy Donor from Multiple myeloma patients. Moreover, the association of miRNA-1246 and MiRNA-1308 can be helpful in determining monoclonal gammopathy of undetermined significance (MGUS) from the MM condition [105]. Xu et al. performed a meta-analysis of the 15 publications and found 7 miRNAs related to MM patients’ survival and prognosis. The results of the study revealed that reduced miR-744, miR-16, miR-15a, miR-25 and let-7e expression causes minimal overall survival in MM patients while the downregulation of miR-15a, miR-25 and miR-16, and upregulation of miR-92a were associated with shorter progression-free survival [106].

3. Innate and Adaptive Immunity

The immune system refers to various biochemical processes that protect multiple organs from pathogenic infections, toxins, and cancerous cells [108]. Apart from the presence of different immune organs such as the thymus and spleen, the immune system is broadly studied under the heading: innate and adaptive immunity, where innate immunity offers the first line of defence [109]. Innate immunity is considered antigen-independent, affording protection within a short period but devoid of immunogenic memory [110]. However, adaptive immunity is antigen-dependent and, therefore, takes time to exert a protective effect [111]. Additionally, adaptive immunity involves immunogenic memory and therefore, upon the repeated exposure of the antigen, a more efficient and rapid immunogenic response is observed [112]. Hence, innate and adaptive immunity are complementary immunogenic mechanisms whereby a defect in one system affects the other.
Innate immunity primarily functions to recruit various immune cells (phagocytes such as neutrophils and macrophages, natural killer cells (NK cells), dendritic cells, basophils, eosinophils, and mast cells) at the site of infection via the production of various cytokines and chemokines [111]. Tumour Necrosis Factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 (IL-1), etc., are some of the commonly observed cytokines that help in clearing the pathogens, but their dysregulated production results in inflammation [113]. Furthermore, the immune system also activates the adaptive immunogenic response via a regulation of antigen-presenting cells (APCs). Phagocytes (neutrophils and macrophages) exhibit bactericidal properties whereby neutrophils are short-lived, whereas macrophages are long-lived and, along with dendritic cells (DCs), additionally assist in the presentation of antigen to the T-cells [112]. Natural killer cells are an important component of innate immunity and are actively involved in the anti-cancer effect by killing tumour cells [114]. NK cells release perforin granzymes and release interferon-gamma (INF-y) that induces apoptosis, leads to the mobilization of APCs, and exhibit the anti-tumour effect [115].
Adaptive immunity works alongside innate immunity, and its function becomes important when innate immunity is ineffective or dysfunctional [111]. Antigen-specific T-cells are the primary component of adaptive immunity that become activated and proliferated in response to APCs and B cells. T cells are the derivative of hematopoietic stem cells (HPSc) and become matured in the thymus. T-cells possess T-cell receptors (TCRs) that can bind with the antigens in response to the signals from APCs, DCs, macrophages or B cells and eliminate them. APCs are immune cells that have a portentous component known as major histocompatibility complex I or II (MHC-I or II) on its surface [112]. APCs digest the antigens and display appropriate protein fragments bounded to the MHC, identified by the T-cells and activated. Activated T cells further regulate the production of cytotoxic T cells, i.e., CD8+ cells or T-helper cells (Th cells), i.e., CD4+ cells.
On the one hand, CD8+ cells destroy the infected cells or tumour cells. On the other hand, CD4+ cells are devoid of a direct cytotoxic effect but release certain cytokines that regulate the immunogenic response [112]. Additionally, Th1, Th2 and Th3 are some of the common Th cells, and Th1 regulates the production of INF-y and modulates the differentiation of B cells to produce antibodies; Th2 cells regulate the release of IL-4, IL-5, IL-13 and regulate immunoglobulin E development, whereas Th-17 regulates the formation of the IL-17 family [116]. In addition to the subtypes of T helper cells, T regulatory (T reg) cells (a subset of T-helper cells) are also an important component of adaptive immunity and ideally act as immunosuppressors and thus, control the hyperimmune responses and play an important role in killing cancerous cells [116].

4. Cancer Immune Escape

As is well known, the immune response in the anticancer effect is complex and consists of multiple immunogenic events whereby the production and release of cancer-associated antigens (CAAs), the processing of APCs, priming, the activation of T-cells and the cytotoxic effect of effectors are important steps [117]. Apart from this, CD8+ cytotoxic T cells (CTL) and CD4+ helper T (Th)1 cells have been reported to curb tumour progression via the release of cytotoxins and production of INF-γ [118]. These findings unequivocally demonstrated that immune editing could be used to manage and treat various types of cancers [117]. Therefore, attempts have been made to stimulate immune editing whereby the innate or adaptive immune system recognizes the tumour or presence of tumour antigens cells and destroys them. Thus, an appropriate balance between immune-inhibitory and immune-stimulatory factors is pivotal for different immune-pharmacotherapeutics’ significant anti-cancer effect. However, while using immunoediting, cancer immune evasion was found to be a major challenge for oncologists as it restricts the favourable clinical outcome. The role of regulatory immune cells, alteration in APCs, immune inhibitory cytokines and immune checkpoints are considered major contributors to the cancer-immune escape [119]. Treg are important immunosuppressive CD+ T cells, and the hyperactivation of Treg has been reported to mitigate the anti-tumour effect of effector cells via the production of TGF-β and IL-10 [120]. Much of the published evidence has shown that Tregs, derived from tumour cells, exhibit an increased tumour evasion effect as compared to naturally existing Tregs [121]. Additionally, Treg downregulates the level of IL-2 and upregulates the level of programmed death-1 (PD-L1), T-cell immunoglobulin mucin-3 (TIM-3), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) etc. and stimulates cancer immune escape [122]. As it is well documented, the cytotoxic potential of effector cells depends on antigens’ expression on cancer cells. However, cancer cells cause alterations in the antigen processing machinery, resulting in the loss of these tumour-associated antigens (TAAs) and cause cancer immune escape [123]. Further, alterations in the major histocompatibility complex-1 and transporters of antigens processing mutation restrict the identification of cancer cells and, thus, promote cancer immune escape [124]. Apart from the factors mentioned above that are responsible for cancer immune escape, the role of inhibitory cytokines and immune checkpoints cannot be ignored. Numerous published studies have confirmed the role of Transforming growth factor-beta (TGF-β) in altering the TME. TGF-β, IL-8, TNF-α and IL-6, Colony-stimulating factor (CSF)-1, IFNs cause an alteration in Treg cell diffraction and stimulate a Treg-mediated increased expression of PD-L-1, inhibiting the anti-tumour effect of cancer-effector cells and promoting cancer immune escape [125]. Nevertheless, the overexpression of immune checkpoints and their ligand, such as PD-L1, plays a critical role in tumor immune evasion by stimulating the production of defective tumour-infiltrating lymphocytes that eventually become devoid of tumour-killing properties [126] (Table 3).

5. Immuno-miRNAs: Central Regulators of Immunity

In recent years, various miRNAs have been explored for their pivotal role in regulating innate and adaptive immunity’s immunogenic key processes related to cancer. These miRNAs and miRNA clusters were crystallized, and attempts were made to understand the signalling pathway and the modulation of various genes related to the immune system [84,128]. Among the various signalling pathways, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) was found to be a major player in the immune system (innate and adaptive immunity) during tumorigenesis (Figure 1). Under normal physiological conditions, NF-kB remains in the cytoplasm in association with its negative regulator, ikB, which in turn is regulated by the IκB kinase (IKK) complex [129]. In response to the oncogenic stimulus, the level of IKK reduces, leading to the phosphorylation of iKB and the activation of NF-kB, thus triggering its nuclear translocation and macrophage activation, which ultimately results in an inflammatory state [130]. Furthermore, MiR-223 is reported to increase the level of IKK, which in turn suppresses the ikB and keeps the NF-kB sequestered in the cytoplasm, leading to the mitigation of inflammation [131]. Apart from miR-223, various other miRNAs play a critical role in regulating NF-kB mediated immunogenic status (Figure 1). Moreover, miR-23-27-24 clusters have been reported to regulate the immunogenic status. The positive transcription of this cluster is induced by the myeloid transcription factor (MTF) known as PU.1. In contrast, it is negatively regulated by the lymphoid transcription factor (LTF) such as paired Box 5 (PAX5), Early B-cell Factor-1 (EBF1) and E2A [132]. This miR cluster has been reported to alter lymphoid cells’ differentiation and promote the lineage commitment of myeloid lineage [133]. Additionally, these clusters negatively impact T cells and repress the Th2 immunity via interaction with the IL-4 signalling pathway [134]. Apart from the negative regulation of Th2, miR-24 has been reported to promote the differentiation of Th1, T reg cells and Th17 whereas miR-23 and 27 restrained the functions of Th1 and Treg/Th17 differentiation [134]. Studies have also shown the involvement of these clusters in the suppression of CD8+ and the stimulation of forkhead box P3 (FoxP3) via the inhibition of NF-kB under the influence of TGF-β as well as via TGF-β mediated cMyc expression [135]. Furthermore, miR17-92 clusters are also important miRs, present on chromosome 13 and accountable for the transcription of six polycistronic miRNAs, i.e., miR-17, miR-92, miR-19b, miR-81a, miR-19a, miR-20a and [136]. This cluster of miRs is pro-inflammatory and plays a pivotal role in T-cell mediated inflammation. The activation of NF-kB is one such cause, which in turn regulates the Janus kinase signal transducer and activator of transcription (JAK-STAT) signalling pathway and stimulates the Th1-mediated INFϒ response [127]. Furthermore, miR17-92 clusters amplify the production of IL-4, IL-5, IL-13, and the development of B cells and modulation of the Inducible T-cell costimulator-phosphatidylinositol-3-kinase (ICOS-PI3K) pathway [39]. Recently emerged evidence has shown that miR17-92 clusters cause macrophage differentiation suppression, by targeting hypoxia-inducible factor-alpha (HIF-α) [137]. Other important miRs include MiR-146a and 155, which are found in abundance in innate and adaptive immunogenic cells [138]. Interestingly, miR-146a exerts an anti-inflammatory effect, whereas miR-155 exhibits a pro-inflammatory effect. The pro-inflammatory effect of miR-146a is exhibited by targeting interleukin-1 receptor-associated kinase 1 or 2 (IRAKI 1 or 2), myeloid differentiation primary response 88 (Myd88), tumour necrosis factor receptor (TNFR)-associated factor6 (TRAF6), Toll-like receptors (TLRs) and NOTCH1. The anti-inflammatory effect in the innate immune cells is exhibited by targeting TNF-α and IL-6, whereas in the adaptive immunogenic cells, it is caused via Th1 differentiation [139]. Furthermore, the anti-inflammatory effect of miR-146a is strengthened by findings where miR-146a deficient mice showed hyper inflammation and the activation of CD8+ and CD4+ cells [140]. MiR-155, on the other hand, showed a pro-inflammatory effect via the modulation of INF-ϒ and production as well as activation of CD8+ and CD4+ cells [141]. The pro-inflammatory effect of miR-155 is further strengthened by the findings where the deficiency of miR-155 causes impairment of Th1 and Th2 activities [142]. Thus, based on these facts, it can be concluded that miR-146a and 155 form ‘ying-yang’ and regulate both innate as well as adaptive immunogenic cells. Both MiR-181 and miR-223 are important anti-inflammatory miRs, and miR-223 is considered as one of the classical innate miRNAs. Earlier, this miR was thought to be myeloid-specific (expressed only in granulocytes cells), but studies have now demonstrated its expression in monocytes and as well as in the macrophages [138]. During monocyte and macrophage differentiation, the level of miR-223 decreases, whereas its level increases that upregulate the activity of IKK-α. The upregulation of IKK-α prevents the phosphorylation of ikB and its activation and the nuclear translocation of NF-kB, leading to the amelioration of TNF-α, IL-6, IL-1β production [143]. However, the activation of the macrophage, which eventually activates the NF-kB signalling pathway, has been reported to downregulate the expression level of miR-223. Thus, miR-223 can be considered as multifactorial in the function whereby it regulates NF-kB, TLRs, NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) and mitogen-activated protein kinase (MAPKs) signalling pathway and exhibits a potent anti-inflammatory effect [143]. In addition to miR-223, miR-181 is another potent anti-inflammatory miR-NA that exhibits an anti-inflammatory effect in innate and adaptive immunogenic cells. Studies have shown that miR-181 mediates the suppression of tyrosine as well as extracellular signal-regulated kinase (ERK) phosphatases, Th1 differentiation and Treg differentiation by targeting the Smad7/TGF-β signalling pathway [144]. Additionally, direct targeting of miR-181 has been reported to modulate CCAAT/enhancer-binding protein alpha (C/EBP-α) and IL-1α in the macrophage and the dendritic cells, respectively [145]. Thus, miR-181 promotes M2 polarization in the macrophage, whereas in the dendritic cell, it acts on cFOS (pro-inflammatory transcription factors) [146].

6. Immune Cell Pathways Regulated by miRNAs in Cancer

In recent years, numerous published studies have highlighted the relationship between the immune system and tumorigenesis. In lieu of this, miRNAs have been reported to play a central role in regulating various immunogenic cells that directly affect the tumorigenesis event.

6.1. Regulation of Monocytes and Macrophages by miRNA

The direct impact of various monocytes has been reported on the maturation, proliferation, and differentiation of monocytes. Furthermore, MiR17-5p, miR-106p, and miR-20a have been reported to involve monocytes’ maturation via the modulation of the expression level of acute myeloid leukaemia-1 (AML-1), a well-known transcription factor [147]. Overexpression of this transcription factor in response to the aforementioned miRs stimulates the M-CSF transcription receptor that eventually causes monocytic differentiation and maturation [148]. In addition, miR-17-92 and miR-21, miR-424 also modulate monocytes’ differentiation via acting on MAPKs, JAK-STAT and TGF-β signalling pathways [149]. Macrophages (M1 and M2 exhibit the opposite effect on tumorigenesis, where M1 macrophages are activated via Toll-like receptors (TLRs) and INF-y leads to the production of pro-inflammatory cytokines such as TNF-α, IL-6, 1L-12, and IL-23 [150]. M1 macrophage also regulates T helper cells’ activity and thus is involved in the anti-tumor effect. The M2 macrophage, on the other hand, is activated in response to IL-4 and IL-13, releases IL-10 and TGF-β and is involved in tumorigenesis pathways [150]. Both miR-21 and miR-146a have been reported to attenuate the activation of M1 macrophage via TLR/NF-kB pathways, whereas miR-155 causes M2 macrophage activation via the regulation of cytokine signalling-1 supression1 [151].

6.2. Regulation of Natural Killer Cells (NK Cells) by miRNA

NK cells are an important component of innate immunity and are directly involved in the tumour-killing activity and the release of INF-y. Although miR-150 regulates its maturation, miR-181 regulates its development via CD3+ cells and Nemo-like kinase signalling molecules, miR-155 stimulates the production of INF-y and miR-29 mitigates its production [152]. As we have already discussed, NK cells exhibit cytotoxic activity against tumour cells via perforin and granzyme B and miR-30e, miR-378 have been reported to stimulate these proteins’ release as mentioned earlier [153].

6.3. Regulation of T Helper Cells and Cytotoxic T Cells by miRNA

T cells are the main component in adaptive immunity and miRs have been reported to regulate these cells’ activity. Furthermore, miR-155 stimulates the CD+ T cells differentiation towards Th2, whereas miR-17 and miR-19b regulate the activity of Th1 cells and increases the production of INF-y [136]. Additionally, Th17 is another important subtype of T helper cells that is responsible for the production of IL-17 [154]. Th17 has been reported to exhibit an antitumor effect via stimulating the activity of INF-y, CD8+ and NK cells in the TME [154]. Both miR-326 and miR-181c have been reported to regulate the development of Th17 by acting on Ets-1 and Smad7, respectively [155]. Furthermore, the increased level and activity of cytotoxic T cells (CD8+ cells) is responsible for the tumour-killing activity. It has been reported that miR-17-92 clusters that are present in the CD8+ cells stimulate the release of INF-y and enhance cytotoxicity towards tumour cells [156]. While miR-155, miR-21, and miR-30b promote the proliferation and cytotoxic activity of CD8+ cells via targeting cytokine signalling 1 (SOCS-1) suppression and Dual Specificity Phosphatase-10 (DUSP-10), miR-29 negatively regulates the activity of CD8+ cells and reduce the expression level of mRNA responsible for producing INF-y [157].

6.4. Regulation of Immune-Checkpoint Molecules by miRNA

Programmed Cell Death Protein 1 (PD-1) and its ligand, which is PD-L1, have been reported to play a pivotal role in tumorigenesis via T cell activation. PD-1 is extensively expressed on NK cells, T cells, macrophages, and on the monocytes and thus, inhibits an innate and adaptive immunogenic response [158]. PD-L-1 on the one hand is extensively expressed on tumour cell surfaces. When it binds with the PD-1, it activates tumour survival pathways, contributes to tumour immune escape, and stimulates tumour proliferation [159]. Various miRNAs have been reported to interact and modulate the activity of PD-1 and PD-L-1. It has been found that miR-155 increases PD-L-1 expression, whereas miR-34a has been reported to inhibit the expression of PD-L-1 [160]. Similarly, miR 33a, miR-21 and miR-873 have been reported to negatively regulate the expression of PD-L-1, whereas miR-146a stimulates the expression of PD-L-1 [161] (Table 4).

7. Effect of miRNAs on Cancer Immunotherapy

Initially, miRNAs were used as biomarkers in cancer diagnosis, prognosis, and response of pharmacotherapeutics [185]. However, it was later found that these miRNAs possess intense modulatory properties against tumorigenesis and hence, can be used as a therapeutic tool as well as a target against various types of cancers [186]. We have also discussed the way that miRNA modulates immunogenic cells in the microenvironment of tumours. Therefore, miRNA emerges as a potential future therapeutic tool against cancer treatment [187]. Currently, two broad categories of microRNA, i.e., miRNA mimics and miRNA inhibitory therapy, are being explored to determine their role as an immunotherapeutic agent in managing and treating cancer. More specifically, miRNA mimics functions to restore or promote the miRNAs that exhibit tumour-suppressing or antitumor effects whereas miRNA antagonists act against those miRNAs that stimulate tumorigenesis [187].
Additionally, miR-43a acts as an inhibitor of PD-L-1, reduces the expression of PD-L-1 mRNA, exhibits an anti-tumor effect, and mimics MRX34 enters a clinical trial to explore its antitumor effect [188]. Aside from reducing the expression of PD-L-1MRX34 also stimulate CD8+ cells tumour infiltration in non-small-cell-lungs carcinoma [188]. Further studies have shown an enhanced antitumor effect of radiotherapy when combined with MRX34 [188]. The administration of mimic MiR-124 also possesses an anti-tumour effect, and hence, miR-124 increases the level of INF-y, IL-2, and TNF-α [180].
Apart from miRNA mimics, miRNA inhibitors have also been explored as potential immunotherapeutic agents. As we know, miRNAs with pro-oncogenic properties on the tumour cells and lead to their inhibition present a potential immunotherapeutic approach. miRNA inhibitors include locked nucleic acid (LNA), anti-sense anti-miRNA oligonucleotides (AMOs), miRNA sponges, anti-miRNAs, miRNAs masks and small molecule inhibitors of miRNAs as shown in Figure 2. AMOs are single-stranded nucleotides (17-22) and act as a competitive inhibitor of pro-oncogenic miRNAs [187]. LNA is also a type of AMOs, but in AMOs, ribose sugar is modified by forming a methyl bridge and this specific modification results in thermal stability, higher water solubility and stabile in the metabolic environment characterizing it as an ideal form for delivery [189]. Until now, LNA-anti-miR-221 has been used in a preclinical xenograft model and showed a significant antitumor effect [190]. miRNA sponges are enclosed in the vector and, when delivered, act as a competitor for the binding site of mRNA of pro-oncogenic miRNA [191]. miRNA sponges have been used to inhibit the progression of metastasis in breasts where the pro-oncogenic effect of miR-9 is masked via the amelioration of miR-9 binding site’s interaction with the mRNA. Apart from the aforementioned immuno-therapeutics of miRNA in cancer, miRNA antagonists are also extensively used, as this class of miRNA antagonizes the pro-oncogenic effect of miRNA by interacting with the various immunogenic cells.
Furthermore, miRNA-155 is known to increase the expression of PD-L-1 and its antagonist (MRG105) has been reported to ameliorate tumour growth [192]. In addition to miR-155, miR-23a, which is released under the influence of TGF-β (secreted from tumors), has been reported to diminish the cytotoxic potential of CD8+ cells antagonist, and exhibited a potent antitumor effect against melanoma [178]. Additionally, the use of miR-17-92 clusters in combination with chimeric antigen cells and in combination with temozolomide has increased the therapeutic efficacy and survival rate among patients of glioblastoma [192].

8. Conclusions

Based on the discussion, it seems persuasive that miRNA acts as potent modulators of carcinogenesis. Numerous published studies present concrete evidence for their immunotherapeutic implication against various types of cancers [24]. Furthermore, miRNA is not only implicated in cancer immunotherapy, but it also plays a pivotal role in the diagnosis and prognosis of cancer [193]. This is achieved by either determining the specific profile of miRNA within the immune cells or by analyzing the miRNA derived from the micro-vesicles of tumour patients [193]. However, further intensive research is needed to develop a diagnostic approach and to transport such a concept from bench to bedside. Moreover, some of the shortcomings of miRNA-based therapeutic interventions must be considered. A high level of inconsistencies in the formulation of miRNA and its delivery system is observed [85]. We herein speculate that the standard pharmacological preparations and precise pharmacodynamic–pharmacokinetics will control the toxic effect of these miRNA-based therapeutics [85]. Furthermore, based on ample published evidence, the development of miRNA-based immune therapeutics and to reduce immune escape has been attempted.
Additionally, we also observed that most of the preclinical study was conducted using an acute dose regimen, and thus, the effects observed via long-term (chronic) study need to be explored in detail. It is also important to discuss that despite tremendous ongoing preclinical and clinical work to develop miRNA-based immunotherapy satisfactory clinical outcome in cancer has not yet been achieved. For example, MRX34 showed a potent anticancer effect in the preclinical study, but immunogenic severe toxicity was observed when tested in cancer patients. However, this unexpected outcome confirmed that the miRNA could cause unwanted non-specific immune-related responses [194,195]. RNA interference (RNAi) causes TLR signalling pathway activation and interferes with the speculated therapeutic outcome [196]. To cater to these issues, a strategy such as combinatorial use of miRNA mimic and siRNA or miRNA along with chemotherapy can be used to mitigate the immune complication and other toxicities [192,197]. Apart from the combinatorial approach, various nanoparticle-based miRNA, also known as ‘miRNome’ and novel drug delivery systems (nanotubes and liposome), have been developed to offer additional advantages over conventional miRNA of safety, efficacy, and stability [198]. This approach also possesses benefits and constraints. The benefits include the fact that nanoparticles can be produced rapidly, drug loading capacity can be modulated, a controlled amount of miRNA can be delivered, and targeted delivery can be achieved [198]. However, some of the constraints for using nanoparticles as a carrier of miRNA include a low efficacy in transfusion, the requirement of multiple administrations, and an increased risk of immunological toxicity [199]. Therefore, we conclude that miRNA-based immunotherapy is a novel and emerging therapeutic method for the treatment and management of cancer, but more extensive studies are required for the safe and effective targeted delivery of miRNA in cancer patients.

Author Contributions

Conceptualization, Methodology, Investigation, Writing—Original Draft: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R. Conceptualization: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R. Conceptualization: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R. Formal Analysis: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R., N.A., M.S. (Mustafa Shehadeh). Formal Analysis: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R., N.A., M.S. (Mustafa Shehadeh). Supervision, Writing—Review & Editing: F.H.P., A.I., M.K.I., M.S. (Mohammed Salahuddin), J.U.R., N.A., M.S. (Mustafa Shehadeh). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this review. Data sharing is not applicable to this article.

Acknowledgments

The authors acknowledge the support of “Khalifa University, Abudhabi, UAE”, “Sheikh Shakhbout Medical City (SSMC), Mayoclinic, Abu Dhabi, United Arab Emirates” and ”Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thun, M.J.; DeLancey, J.O.; Center, M.M.; Jemal, A.; Ward, E.M. The Global Burden of Cancer: Priorities for Prevention. Carcinogenesis 2010, 31, 100–110. [Google Scholar] [CrossRef] [Green Version]
  2. Hassanpour, S.H.; Dehghani, M. Review of Cancer from Perspective of Molecular. J. Cancer Res. Pract. 2017, 4, 127–129. [Google Scholar] [CrossRef]
  3. American Cancer Society. Global Cancer Facts & Figures. Available online: https://www.cancer.org/research/cancer-facts-statistics/global.html (accessed on 4 February 2021).
  4. Fisher, R.; Pusztai, L.; Swanton, C. Cancer Heterogeneity: Implications for Targeted Therapeutics. Br. J. Cancer 2013, 108, 479–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Chaffer, C.L.; Weinberg, R.A. A Perspective on Cancer Cell Metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef]
  6. Alhmied, F.I.; Alammar, A.H.; Alsultan, B.M.; Alshehri, M.; Pottoo, F.H. Molecular Mechanisms of Thymoquinone as Anticancer Agent. Comb. Chem. High. Throughput Screen 2020. [Google Scholar] [CrossRef]
  7. Poon, S.L.; McPherson, J.R.; Tan, P.; Teh, B.T.; Rozen, S.G. Mutation Signatures of Carcinogen Exposure: Genome-Wide Detection and New Opportunities for Cancer Prevention. Genome Med. 2014, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  8. Trafialek, J.; Kolanowski, W. Dietary Exposure to Meat-Related Carcinogenic Substances: Is There a Way to Estimate the Risk? Int. J. Food Sci. Nutr. 2014, 65, 774–780. [Google Scholar] [CrossRef]
  9. Cumberbatch, M.G.K.; Cox, A.; Teare, D.; Catto, J.W.F. Contemporary Occupational Carcinogen Exposure and Bladder Cancer: A Systematic Review and Meta-Analysis. JAMA Oncol. 2015, 1, 1282–1290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Antwi, S.O.; Eckert, E.C.; Sabaque, C.V.; Leof, E.R.; Hawthorne, K.M.; Bamlet, W.R.; Chaffee, K.G.; Oberg, A.L.; Petersen, G.M. Exposure to Environmental Chemicals and Heavy Metals, and Risk of Pancreatic Cancer. Cancer Causes Control. 2015, 26, 1583–1591. [Google Scholar] [CrossRef] [Green Version]
  11. Lan, H.; Lu, H.; Wang, X.; Jin, H. MicroRNAs as Potential Biomarkers in Cancer: Opportunities and Challenges. Available online: https://www.hindawi.com/journals/bmri/2015/125094/ (accessed on 4 February 2021).
  12. Aboelmagd, A.; Rayes, S.M.E.; Gomaa, M.S.; Ali, I.A.I.; Fathalla, W.; Pottoo, F.H.; Khan, F.A.; Khalifa, M.E. The Synthesis and Antiproliferative Activity of New N-Allyl Quinoxalinecarboxamides and Their O-Regioisomers. New J. Chem. 2021, 45, 831–849. [Google Scholar] [CrossRef]
  13. Aboelmagd, A.; Alotaibi, S.H.; Rayes, S.M.E.; Elsayed, G.M.; Ali, I.A.I.; Fathalla, W.; Pottoo, F.H.; Khan, F.A. Synthesis and Anti Proliferative Activity of New N-Pentylquinoxaline Carboxamides and Their O-Regioisomer. ChemistrySelect 2020, 5, 13439–13453. [Google Scholar] [CrossRef]
  14. Nawaz, F.; Alam, O.; Perwez, A.; Rizvi, M.A.; Naim, M.J.; Siddiqui, N.; Pottoo, F.H.; Jha, M. 3′-(4-(Benzyloxy)Phenyl)-1′-Phenyl-5-(Heteroaryl/Aryl)-3,4-Dihydro-1′H,2H-[3,4′-Bipyrazole]-2-Carboxamides as EGFR Kinase Inhibitors: Synthesis, Anticancer Evaluation, and Molecular Docking Studies. Arch. Der Pharm. 2020, 353, 1900262. [Google Scholar] [CrossRef] [PubMed]
  15. Rayes, S.M.E.; Aboelmagd, A.; Gomaa, M.S.; Fathalla, W.; Ali, I.A.; Pottoo, F.H.; Khan, F.A. Newly Synthesized 3-(4-Chloro-Phenyl)-3-Hydroxy-2,2-Dimethyl-Propionic Acid Methyl Ester Derivatives Selectively Inhibit the Proliferation of Colon Cancer Cells. RSC Adv. 2020, 10, 8825–8841. [Google Scholar] [CrossRef]
  16. Barger, J.F.; Rahman, M.A.; Jackson, D.; Acunzo, M.; Nana-Sinkam, S.P. Extracellular MiRNAs as Biomarkers in Cancer. Food Chem. Toxicol. 2016, 98, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Sohel, M.M.H. Circulating MicroRNAs as Biomarkers in Cancer Diagnosis. Life Sci. 2020, 248, 117473. [Google Scholar] [CrossRef] [PubMed]
  18. Hunter, M.P.; Ismail, N.; Zhang, X.; Aguda, B.D.; Lee, E.J.; Yu, L.; Xiao, T.; Schafer, J.; Lee, M.-L.T.; Schmittgen, T.D.; et al. Detection of MicroRNA Expression in Human Peripheral Blood Microvesicles. PLoS ONE 2008, 3, e3694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Pottoo, F.H.; Javed, M.N.; Rahman, J.U.; Abu-Izneid, T.; Khan, F.A. Targeted Delivery of MiRNA Based Therapeuticals in the Clinical Management of Glioblastoma Multiforme. Semin. Cancer Biol. 2020. [Google Scholar] [CrossRef]
  20. Pottoo, F.H.; Barkat, M.A.; Ansari, M.A.; Javed, M.N.; Jamal, Q.M.S.; Kamal, M.A. Nanotechnological Based MiRNA Intervention in the Therapeutic Management of Neuroblastoma. In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar] [CrossRef]
  21. Sohel, M.H. Extracellular/Circulating MicroRNAs: Release Mechanisms, Functions and Challenges. Achiev. Life Sci. 2016, 10, 175–186. [Google Scholar] [CrossRef] [Green Version]
  22. Kong, Y.W.; Ferland-McCollough, D.; Jackson, T.J.; Bushell, M. MicroRNAs in Cancer Management. Lancet Oncol. 2012, 13, e249–e258. [Google Scholar] [CrossRef]
  23. Shafi, S.; Khan, S.; Hoda, F.; Fayaz, F.; Singh, A.; Khan, M.A.; Ali, R.; Pottoo, F.H.; Tariq, S.; Najmi, A.K. Decoding Novel Mechanisms and Emerging Therapeutic Strategies in Breast Cancer Resistance. Curr. Drug Metab. 2020, 21, 199–210. [Google Scholar] [CrossRef]
  24. Peng, Y.; Croce, C.M. The Role of MicroRNAs in Human Cancer. Signal Transduct. Target. Ther. 2016, 1, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Borchert, G.M.; Lanier, W.; Davidson, B.L. RNA Polymerase III Transcribes Human MicroRNAs. Nat. Struct. Mol. Biol. 2006, 13, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, Y.; Kim, M.; Han, J.; Yeom, K.-H.; Lee, S.; Baek, S.H.; Kim, V.N. MicroRNA Genes Are Transcribed by RNA Polymerase II. EMBO J. 2004, 23, 4051–4060. [Google Scholar] [CrossRef] [PubMed]
  27. Denli, A.M.; Tops, B.B.J.; Plasterk, R.H.A.; Ketting, R.F.; Hannon, G.J. Processing of Primary MicroRNAs by the Microprocessor Complex. Nature 2004, 432, 231–235. [Google Scholar] [CrossRef] [PubMed]
  28. Alarcón, C.R.; Lee, H.; Goodarzi, H.; Halberg, N.; Tavazoie, S.F. N6-Methyladenosine Marks Primary MicroRNAs for Processing. Nature 2015, 519, 482–485. [Google Scholar] [CrossRef] [PubMed]
  29. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 Complexes Carry a Population of Circulating MicroRNAs Independent of Vesicles in Human Plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
  30. Xie, M.; Li, M.; Vilborg, A.; Lee, N.; Shu, M.-D.; Yartseva, V.; Šestan, N.; Steitz, J.A. Mammalian 5’-Capped MicroRNA Precursors That Generate a Single MicroRNA. Cell 2013, 155, 1568–1580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Prud’homme, G.J.; Glinka, Y.; Lichner, Z.; Yousef, G.M. Neuropilin-1 Is a Receptor for Extracellular MiRNA and AGO2/MiRNA Complexes and Mediates the Internalization of MiRNAs That Modulate Cell Function. Oncotarget 2016, 7, 68057–68071. [Google Scholar] [CrossRef] [Green Version]
  32. Yang, J.-S.; Maurin, T.; Robine, N.; Rasmussen, K.D.; Jeffrey, K.L.; Chandwani, R.; Papapetrou, E.P.; Sadelain, M.; O’Carroll, D.; Lai, E.C. Conserved Vertebrate Mir-451 Provides a Platform for Dicer-Independent, Ago2-Mediated MicroRNA Biogenesis. Proc. Natl. Acad. Sci. USA 2010. [Google Scholar] [CrossRef] [Green Version]
  33. Ruby, J.G.; Jan, C.H.; Bartel, D.P. Intronic MicroRNA Precursors That Bypass Drosha Processing. Nature 2007, 448, 83–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gandellini, P.; Giovannetti, E.; Nicassio, F. MicroRNAs in Cancer Management: Big Challenges for Small Molecules. Biomed. Res. Int. 2015, 2015, 982156. [Google Scholar] [CrossRef] [PubMed]
  35. Otmani, K.; Lewalle, P. Tumor Suppressor MiRNA in Cancer Cells and the Tumor Microenvironment: Mechanism of Deregulation and Clinical Implications. Front. Oncol. 2021, 11, 708765. [Google Scholar] [CrossRef] [PubMed]
  36. Coronnello, C.; Benos, P.V. ComiR: Combinatorial MicroRNA Target Prediction Tool. Nucleic Acids Res. 2013, 41, W159–W164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Friedman, Y.; Karsenty, S.; Linial, M. MiRror-Suite: Decoding Coordinated Regulation by MicroRNAs. Database 2014, 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Griffiths-Jones, S. The MicroRNA Registry. Nucleic Acids Res. 2004, 32, D109–D111. [Google Scholar] [CrossRef] [PubMed]
  39. Skinner, J.P.J.; Keown, A.A.; Chong, M.M.W. The MiR-17∼92a Cluster of MicroRNAs Is Required for the Fitness of Foxp3+ Regulatory T Cells. PLoS ONE 2014, 9, e88997. [Google Scholar] [CrossRef]
  40. Kohlhapp, F.; Mitra, A.K.; Lengyel, E.; Peter, M.E. MicroRNAs as mediators and communicators between cancer cells and the tumor microenvironment. Oncogene 2015, 34, 5857–5868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. 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] [PubMed] [Green Version]
  42. Calin, G.A.; Sevignani, C.; Dumitru, C.D.; Hyslop, T.; Noch, E.; Yendamuri, S.; Shimizu, M.; Rattan, S.; Bullrich, F.; Negrini, M.; et al. Human MicroRNA Genes Are Frequently Located at Fragile Sites and Genomic Regions Involved in Cancers. Proc. Natl. Acad. Sci. USA 2004, 101, 2999–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Reddy, K.B. MicroRNA (MiRNA) in Cancer. Cancer Cell Int. 2015, 15, 38. [Google Scholar] [CrossRef] [Green Version]
  44. Izumchenko, E.; Chang, X.; Michailidi, C.; Kagohara, L.; Ravi, R.; Paz, K.; Brait, M.; Hoque, M.O.; Ling, S.; Bedi, A.; et al. The TGFβ-MiR200-MIG6 Pathway Orchestrates the EMT-Associated Kinase Switch That Induces Resistance to EGFR Inhibitors. Cancer Res. 2014, 74, 3995–4005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Chiocca, E.A.; Lawler, S.E. The Many Functions of MicroRNAs in Glioblastoma. World Neurosurg. 2010, 73, 598–601. [Google Scholar] [CrossRef] [PubMed]
  46. Godlewski, J.; Bronisz, A.; Nowicki, M.O.; Chiocca, E.A.; Lawler, S. MicroRNA-451: A Conditional Switch Controlling Glioma Cell Proliferation and Migration. Cell Cycle 2010, 9, 2742–2748. [Google Scholar] [CrossRef] [Green Version]
  47. Hayes, J.; Peruzzi, P.P.; Lawler, S. MicroRNAs in Cancer: Biomarkers, Functions and Therapy. Trends Mol. Med. 2014, 20, 460–469. [Google Scholar] [CrossRef] [PubMed]
  48. Wiemer, E.A.C. The Role of MicroRNAs in Cancer: No Small Matter. Eur. J. Cancer 2007, 43, 1529–1544. [Google Scholar] [CrossRef] [PubMed]
  49. Mehrgou, A.; Akouchekian, M. Therapeutic Impacts of MicroRNAs in Breast Cancer by Their Roles in Regulating Processes Involved in This Disease. J. Res. Med. Sci. 2017, 22, 130. [Google Scholar] [CrossRef] [PubMed]
  50. Han, C.; Yu, Z.; Duan, Z.; Kan, Q. Role of MicroRNA-1 in Human Cancer and Its Therapeutic Potentials. Biomed. Res. Int. 2014, 2014, 428371. [Google Scholar] [CrossRef] [Green Version]
  51. Lee, Y.; Jeon, K.; Lee, J.-T.; Kim, S.; Kim, V.N. MicroRNA Maturation: Stepwise Processing and Subcellular Localization. EMBO J. 2002, 21, 4663–4670. [Google Scholar] [CrossRef] [Green Version]
  52. Ha, M.; Kim, V.N. Regulation of MicroRNA Biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  53. Frixa, T.; Donzelli, S.; Blandino, G. Oncogenic MicroRNAs: Key Players in Malignant Transformation. Cancers 2015, 7, 2466–2485. [Google Scholar] [CrossRef] [PubMed]
  54. Zhang, B.; Pan, X.; Cobb, G.P.; Anderson, T.A. MicroRNAs as Oncogenes and Tumor Suppressors. Dev. Biol. 2007, 302, 1–12. [Google Scholar] [CrossRef] [Green Version]
  55. Zhang, L.; Liao, Y.; Tang, L. MicroRNA-34 Family: A Potential Tumor Suppressor and Therapeutic Candidate in Cancer. J. Exp. Clin. Cancer Res. 2019, 38, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chen, J.; Wu, L.; Sun, Y.; Yin, Q.; Chen, X.; Liang, S.; Meng, Q.; Long, H.; Li, F.; Luo, C.; et al. Mir-421 in Plasma as a Potential Diagnostic Biomarker for Precancerous Gastric Lesions and Early Gastric Cancer. PeerJ 2019, 7, e7002. [Google Scholar] [CrossRef] [Green Version]
  57. Chen, T.-H.; Lee, C.; Chiu, C.-T.; Chu, Y.-Y.; Cheng, H.-T.; Hsu, J.-T.; Tsou, Y.-K.; Wu, R.-C.; Chen, T.-C.; Chang, N.-C.; et al. Circulating MicroRNA-196a Is an Early Gastric Cancer Biomarker. Oncotarget 2018, 9, 10317–10323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Desantis, V.; Solimando, A.G.; Saltarella, I.; Sacco, A.; Giustini, V.; Bento, M.; Lamanuzzi, A.; Melaccio, A.; Frassanito, M.A.; Paradiso, A.; et al. MicroRNAs as a Potential New Preventive Approach in the Transition from Asymptomatic to Symptomatic Multiple Myeloma Disease. Cancers 2021, 13, 3650. [Google Scholar] [CrossRef]
  59. Marcucci, G.; Mrózek, K.; Radmacher, M.D.; Garzon, R.; Bloomfield, C.D. The Prognostic and Functional Role of MicroRNAs in Acute Myeloid Leukemia. Blood 2011, 117, 1121–1129. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, L.; Wang, L.; Dong, D.; Wang, Z.; Ji, W.; Yu, M.; Zhang, F.; Niu, R.; Zhou, Y. MiR-34b/c-5p and the Neurokinin-1 Receptor Regulate Breast Cancer Cell Proliferation and Apoptosis. Cell Prolif. 2019, 52, e12527. [Google Scholar] [CrossRef]
  61. Yang, S.; Li, Y.; Gao, J.; Zhang, T.; Li, S.; Luo, A.; Chen, H.; Ding, F.; Wang, X.; Liu, Z. MicroRNA-34 Suppresses Breast Cancer Invasion and Metastasis by Directly Targeting Fra-1. Oncogene 2013, 32, 4294–4303. [Google Scholar] [CrossRef]
  62. Acunzo, M.; Romano, G.; Palmieri, D.; Laganá, A.; Garofalo, M.; Balatti, V.; Drusco, A.; Chiariello, M.; Nana-Sinkam, P.; Croce, C.M. Cross-Talk between MET and EGFR in Non-Small Cell Lung Cancer Involves MiR-27a and Sprouty2. Proc. Natl. Acad. Sci. USA 2013, 110, 8573–8578. [Google Scholar] [CrossRef] [Green Version]
  63. Outeiro-Pinho, G.; Barros-Silva, D.; Aznar, E.; Sousa, A.-I.; Vieira-Coimbra, M.; Oliveira, J.; Gonçalves, C.S.; Costa, B.M.; Junker, K.; Henrique, R.; et al. MicroRNA-30a-5pme: A Novel Diagnostic and Prognostic Biomarker for Clear Cell Renal Cell Carcinoma in Tissue and Urine Samples. J. Exp. Clin. Cancer Res. 2020, 39, 98. [Google Scholar] [CrossRef]
  64. Song, S.; Long, M.; Yu, G.; Cheng, Y.; Yang, Q.; Liu, J.; Wang, Y.; Sheng, J.; Wang, L.; Wang, Z.; et al. Urinary Exosome MiR-30c-5p as a Biomarker of Clear Cell Renal Cell Carcinoma That Inhibits Progression by Targeting HSPA5. J. Cell. Mol. Med. 2019, 23, 6755–6765. [Google Scholar] [CrossRef]
  65. Li, Y.-J.; Du, L.; Aldana-Masangkay, G.; Wang, X.; Urak, R.; Forman, S.J.; Rosen, S.T.; Chen, Y. Regulation of MiR-34b/c-Targeted Gene Expression Program by SUMOylation. Nucleic Acids Res. 2018, 46, 7108–7123. [Google Scholar] [CrossRef] [PubMed]
  66. Roy, S.; Levi, E.; Majumdar, A.P.; Sarkar, F.H. Expression of MiR-34 Is Lost in Colon Cancer Which Can Be Re-Expressed by a Novel Agent CDF. J. Hematol. Oncol. 2012, 5, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jiang, L.; Hermeking, H. MiR-34a and MiR-34b/c Suppress Intestinal Tumorigenesis. Cancer Res. 2017, 77, 2746–2758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Liang, J.; Li, Y.; Daniels, G.; Sfanos, K.; De Marzo, A.; Wei, J.; Li, X.; Chen, W.; Wang, J.; Zhong, X.; et al. LEF1 Targeting EMT in Prostate Cancer Invasion Is Regulated by MiR-34a. Mol. Cancer Res. 2015, 13, 681–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Fang, L.-L.; Sun, B.-F.; Huang, L.-R.; Yuan, H.-B.; Zhang, S.; Chen, J.; Yu, Z.-J.; Luo, H. Potent Inhibition of MiR-34b on Migration and Invasion in Metastatic Prostate Cancer Cells by Regulating the TGF-β Pathway. Int. J. Mol. Sci. 2017, 18, E2762. [Google Scholar] [CrossRef] [Green Version]
  70. Stahlhut, C.; Slack, F.J. Combinatorial Action of MicroRNAs Let-7 and MiR-34 Effectively Synergizes with Erlotinib to Suppress Non-Small Cell Lung Cancer Cell Proliferation. Cell Cycle 2015, 14, 2171–2180. [Google Scholar] [CrossRef] [Green Version]
  71. Zhao, K.; Cheng, J.; Chen, B.; Liu, Q.; Xu, D.; Zhang, Y. Circulating MicroRNA-34 Family Low Expression Correlates with Poor Prognosis in Patients with Non-Small Cell Lung Cancer. J. Thorac. Dis. 2017, 9, 3735–3746. [Google Scholar] [CrossRef] [Green Version]
  72. Kasinski, A.L.; Slack, F.J. MiRNA-34 Prevents Cancer Initiation and Progression in a Therapeutically Resistant K-Ras and P53-Induced Mouse Model of Lung Adenocarcinoma. Cancer Res. 2012, 72, 5576–5587. [Google Scholar] [CrossRef] [Green Version]
  73. Xie, K.; Liu, J.; Chen, J.; Dong, J.; Ma, H.; Liu, Y.; Hu, Z. Methylation-Associated Silencing of MicroRNA-34b in Hepatocellular Carcinoma Cancer. Gene 2014, 543, 101–107. [Google Scholar] [CrossRef]
  74. Wang, Y.; Jia, L.-S.; Yuan, W.; Wu, Z.; Wang, H.-B.; Xu, T.; Sun, J.-C.; Cheng, K.-F.; Shi, J.-G. Low MiR-34a and MiR-192 Are Associated with Unfavorable Prognosis in Patients Suffering from Osteosarcoma. Am. J. Transl. Res. 2015, 7, 111–119. [Google Scholar]
  75. Gang, L.; Qun, L.; Liu, W.-D.; Li, Y.-S.; Xu, Y.-Z.; Yuan, D.-T. MicroRNA-34a Promotes Cell Cycle Arrest and Apoptosis and Suppresses Cell Adhesion by Targeting DUSP1 in Osteosarcoma. Am. J. Transl. Res. 2017, 9, 5388–5399. [Google Scholar]
  76. Xi, L.; Zhang, Y.; Kong, S.; Liang, W. MiR-34 Inhibits Growth and Promotes Apoptosis of Osteosarcoma in Nude Mice through Targetly Regulating TGIF2 Expression. Biosci. Rep. 2018, 38, BSR20180078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Liu, L.; Ren, W.; Chen, K. MiR-34a Promotes Apoptosis and Inhibits Autophagy by Targeting HMGB1 in Acute Myeloid Leukemia Cells. Cell. Physiol. Biochem. 2017, 41, 1981–1992. [Google Scholar] [CrossRef] [PubMed]
  78. Peng, D.; Wang, H.; Li, L.; Ma, X.; Chen, Y.; Zhou, H.; Luo, Y.; Xiao, Y.; Liu, L. MiR-34c-5p Promotes Eradication of Acute Myeloid Leukemia Stem Cells by Inducing Senescence through Selective RAB27B Targeting to Inhibit Exosome Shedding. Leukemia 2018, 32, 1180–1188. [Google Scholar] [CrossRef]
  79. Wong, K.Y.; Yim, R.L.H.; So, C.C.; Jin, D.-Y.; Liang, R.; Chim, C.S. Epigenetic Inactivation of the MIR34B/C in Multiple Myeloma. Blood 2011, 118, 5901–5904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent Deletions and Down-Regulation of Micro-RNA Genes MiR15 and MiR16 at 13q14 in Chronic Lymphocytic Leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Roderburg, C.; Luedde, T. Circulating MicroRNAs as Markers of Liver Inflammation, Fibrosis and Cancer. J. Hepatol. 2014, 61, 1434–1437. [Google Scholar] [CrossRef] [Green Version]
  82. Sohn, W.; Kim, J.; Kang, S.H.; Yang, S.R.; Cho, J.-Y.; Cho, H.C.; Shim, S.G.; Paik, Y.-H. Serum Exosomal MicroRNAs as Novel Biomarkers for Hepatocellular Carcinoma. Exp. Mol. Med. 2015, 47, e184. [Google Scholar] [CrossRef]
  83. De Silva, S.; Tennekoon, K.H.; Karunanayake, E.H. Interaction of Gut Microbiome and Host MicroRNAs with the Occurrence of Colorectal and Breast Cancer and Their Impact on Patient Immunity. Oncotarget 2021, 14, 5115–5129. [Google Scholar] [CrossRef]
  84. Iftikhar, H.; Carney, G.E. Evidence and Potential in Vivo Functions for Biofluid MiRNAs: From Expression Profiling to Functional Testing. BioEssays 2016, 38, 367–378. [Google Scholar] [CrossRef] [PubMed]
  85. Cui, M.; Wang, H.; Yao, X.; Zhang, D.; Xie, Y.; Cui, R.; Zhang, X. Circulating MicroRNAs in Cancer: Potential and Challenge. Front. Genet. 2019, 10, 626. [Google Scholar] [CrossRef] [Green Version]
  86. Kosaka, N.; Iguchi, H.; Yoshioka, Y.; Takeshita, F.; Matsuki, Y.; Ochiya, T. Secretory Mechanisms and Intercellular Transfer of MicroRNAs in Living Cells. J. Biol. Chem. 2010, 285, 17442–17452. [Google Scholar] [CrossRef] [Green Version]
  87. Hannafon, B.N.; Carpenter, K.J.; Berry, W.L.; Janknecht, R.; Dooley, W.C.; Ding, W.-Q. Exosome-Mediated MicroRNA Signaling from Breast Cancer Cells Is Altered by the Anti-Angiogenesis Agent Docosahexaenoic Acid (DHA). Mol. Cancer 2015, 14, 133. [Google Scholar] [CrossRef] [Green Version]
  88. Yang, M.; Chen, J.; Su, F.; Yu, B.; Su, F.; Lin, L.; Liu, Y.; Huang, J.-D.; Song, E. Microvesicles Secreted by Macrophages Shuttle Invasion-Potentiating MicroRNAs into Breast Cancer Cells. Mol. Cancer 2011, 10, 117. [Google Scholar] [CrossRef] [Green Version]
  89. Sakha, S.; Muramatsu, T.; Ueda, K.; Inazawa, J. Exosomal MicroRNA MiR-1246 Induces Cell Motility and Invasion through the Regulation of DENND2D in Oral Squamous Cell Carcinoma. Sci. Rep. 2016, 6, 38750. [Google Scholar] [CrossRef] [Green Version]
  90. Hu, Y.; Rao, S.-S.; Wang, Z.-X.; Cao, J.; Tan, Y.-J.; Luo, J.; Li, H.-M.; Zhang, W.-S.; Chen, C.-Y.; Xie, H. Exosomes from Human Umbilical Cord Blood Accelerate Cutaneous Wound Healing through MiR-21-3p-Mediated Promotion of Angiogenesis and Fibroblast Function. Theranostics 2018, 8, 169–184. [Google Scholar] [CrossRef]
  91. Zhou, W.; Fong, M.Y.; Min, Y.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-Secreted MiR-105 Destroys Vascular Endothelial Barriers to Promote Metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Price, C.; Chen, J. MicroRNAs in Cancer Biology and Therapy: Current Status and Perspectives. Genes Dis. 2014, 1, 53–63. [Google Scholar] [CrossRef] [Green Version]
  93. Yang, C.; Wang, C.; Chen, X.; Chen, S.; Zhang, Y.; Zhi, F.; Wang, J.; Li, L.; Zhou, X.; Li, N.; et al. Identification of Seven Serum MicroRNAs from a Genome-Wide Serum MicroRNA Expression Profile as Potential Noninvasive Biomarkers for Malignant Astrocytomas. Int. J. Cancer 2013, 132, 116–127. [Google Scholar] [CrossRef] [PubMed]
  94. Arab, A.; Karimipoor, M.; Irani, S.; Kiani, A.; Zeinali, S.; Tafsiri, E.; Sheikhy, K. Potential Circulating MiRNA Signature for Early Detection of NSCLC. Cancer Genet. 2017, 216–217, 150–158. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, H.; Peng, R.; Wang, J.; Qin, Z.; Xue, L. Circulating MicroRNAs as Potential Cancer Biomarkers: The Advantage and Disadvantage. Clin. Epigenetics 2018, 10, 59. [Google Scholar] [CrossRef] [Green Version]
  96. Zhang, H.; Mao, F.; Shen, T.; Luo, Q.; Ding, Z.; Qian, L.; Huang, J. Plasma MiR-145, MiR-20a, MiR-21 and MiR-223 as Novel Biomarkers for Screening Early-Stage Non-Small Cell Lung Cancer. Oncol. Lett. 2017, 13, 669–676. [Google Scholar] [CrossRef] [Green Version]
  97. Lehmann, S.M.; Krüger, C.; Park, B.; Derkow, K.; Rosenberger, K.; Baumgart, J.; Trimbuch, T.; Eom, G.; Hinz, M.; Kaul, D.; et al. An Unconventional Role for MiRNA: Let-7 Activates Toll-like Receptor 7 and Causes Neurodegeneration. Nat. Neurosci. 2012, 15, 827–835. [Google Scholar] [CrossRef]
  98. Fabbri, M. MicroRNAs and MiRceptors: A New Mechanism of Action for Intercellular Communication. Philos. Trans. R Soc. Lond. B Biol. Sci. 2018, 373. [Google Scholar] [CrossRef]
  99. Khan, K.; Javed, Z.; Sadia, H.; Sharifi-Rad, J.; Cho, W.C.; Luparello, C. Quercetin and MicroRNA Interplay in Apoptosis Regulation in Ovarian Cancer. Curr. Pharm. Des. 2021, 27, 2328–2336. [Google Scholar] [CrossRef]
  100. Wang, F.; Zheng, Z.; Guo, J.; Ding, X. Correlation and Quantitation of MicroRNA Aberrant Expression in Tissues and Sera from Patients with Breast Tumor. Gynecol. Oncol. 2010, 119, 586–593. [Google Scholar] [CrossRef]
  101. Bhat, S.A.; Majid, S.; Hassan, T. MicroRNAs and Its Emerging Role as Breast Cancer Diagnostic Marker—A Review. Adv. Biomark. Sci. Technol. 2019, 1, 1–8. [Google Scholar] [CrossRef]
  102. Asaga, S.; Kuo, C.; Nguyen, T.; Terpenning, M.; Giuliano, A.E.; Hoon, D.S.B. Direct Serum Assay for MicroRNA-21 Concentrations in Early and Advanced Breast Cancer. Clin. Chem. 2011, 57, 84–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Zelli, V.; Compagnoni, C.; Capelli, R.; Cannita, K.; Sidoni, T.; Ficorella, C.; Capalbo, C.; Zazzeroni, F.; Tessitore, A.; Alesse, E. Circulating MicroRNAs as Prognostic and Therapeutic Biomarkers in Breast Cancer Molecular Subtypes. J. Pers. Med. 2020, 10, E98. [Google Scholar] [CrossRef] [PubMed]
  104. Khalighfard, S.; Alizadeh, A.M.; Irani, S.; Omranipour, R. Plasma MiR-21, MiR-155, MiR-10b, and Let-7a as the Potential Biomarkers for the Monitoring of Breast Cancer Patients. Sci. Rep. 2018, 8, 17981. [Google Scholar] [CrossRef]
  105. Jones, C.I.; Zabolotskaya, M.V.; King, A.J.; Stewart, H.J.S.; Horne, G.A.; Chevassut, T.J.; Newbury, S.F. Identification of Circulating MicroRNAs as Diagnostic Biomarkers for Use in Multiple Myeloma. Br. J. Cancer 2012, 107, 1987–1996. [Google Scholar] [CrossRef]
  106. Xu, P.; Xia, T.; Ling, Y.; Chen, B. MiRNAs with Prognostic Significance in Multiple Myeloma: A Systemic Review and Meta-Analysis. Medicine 2019, 98, e16711. [Google Scholar] [CrossRef]
  107. Condrat, C.E.; Thompson, D.C.; Barbu, M.G.; Bugnar, O.L.; Boboc, A.; Cretoiu, D.; Suciu, N.; Cretoiu, S.M.; Voinea, S.C. MiRNAs as Biomarkers in Disease: Latest Findings Regarding Their Role in Diagnosis and Prognosis. Cells 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Chaplin, D.D. Overview of the Immune Response. J. Allergy Clin. Immunol. 2010, 125, S3–S23. [Google Scholar] [CrossRef] [PubMed]
  109. Grizzi, F.; Borroni, E.M.; Yiu, D.; Farina, F.M.; Cananzi, F.C.M.; Laghi, L. Prognostic Value of Innate and Adaptive Immunity in Cancers. In Cancer Immunology: A Translational Medicine Context; Rezaei, N., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 403–415. ISBN 978-3-030-30845-2. [Google Scholar]
  110. Thaiss, C.A.; Zmora, N.; Levy, M.; Elinav, E. The Microbiome and Innate Immunity. Nature 2016, 535, 65–74. [Google Scholar] [CrossRef] [PubMed]
  111. Chang, R.B.; Beatty, G.L. The Interplay between Innate and Adaptive Immunity in Cancer Shapes the Productivity of Cancer Immunosurveillance. J. Leukoc. Biol. 2020, 108, 363–376. [Google Scholar] [CrossRef]
  112. Bonilla, F.A.; Oettgen, H.C. Adaptive Immunity. J. Allergy Clin. Immunol. 2010, 125, S33–S40. [Google Scholar] [CrossRef]
  113. Mantovani, A.; Dinarello, C.A.; Molgora, M.; Garlanda, C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019, 50, 778–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Vivier, E.; Raulet, D.H.; Moretta, A.; Caligiuri, M.A.; Zitvogel, L.; Lanier, L.L.; Yokoyama, W.M.; Ugolini, S. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 2011, 331, 44–49. [Google Scholar] [CrossRef] [Green Version]
  115. Stone, K.D.; Prussin, C.; Metcalfe, D.D. IgE, Mast Cells, Basophils, and Eosinophils. J. Allergy Clin. Immunol. 2010, 125, S73–S80. [Google Scholar] [CrossRef]
  116. Zhu, J.; Paul, W.E. Heterogeneity and Plasticity of T Helper Cells. Cell Res. 2010, 20, 4–12. [Google Scholar] [CrossRef] [PubMed]
  117. O’Sullivan, D.; Sanin, D.E.; Pearce, E.J.; Pearce, E.L. Metabolic Interventions in the Immune Response to Cancer. Nat. Rev. Immunol. 2019, 19, 324–335. [Google Scholar] [CrossRef] [PubMed]
  118. Zamarron, B.F.; Chen, W. Dual Roles of Immune Cells and Their Factors in Cancer Development and Progression. Int. J. Biol. Sci. 2011, 7, 651–658. [Google Scholar] [CrossRef] [PubMed]
  119. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The Immunobiology of Cancer Immunosurveillance and Immunoediting. Immunity 2004, 21, 137–148. [Google Scholar] [CrossRef] [Green Version]
  120. Taylor, A.; Verhagen, J.; Blaser, K.; Akdis, M.; Akdis, C.A. Mechanisms of Immune Suppression by Interleukin-10 and Transforming Growth Factor-Beta: The Role of T Regulatory Cells. Immunology 2006, 117, 433–442. [Google Scholar] [CrossRef]
  121. Gasparoto, T.H.; de Souza Malaspina, T.S.; Benevides, L.; de Melo, E.J.F.; Costa, M.R.S.N.; Damante, J.H.; Ikoma, M.R.V.; Garlet, G.P.; Cavassani, K.A.; da Silva, J.S.; et al. Patients with Oral Squamous Cell Carcinoma Are Characterized by Increased Frequency of Suppressive Regulatory T Cells in the Blood and Tumor Microenvironment. Cancer Immunol. Immunother. 2010, 59, 819–828. [Google Scholar] [CrossRef]
  122. Marin-Acevedo, J.A.; Dholaria, B.; Soyano, A.E.; Knutson, K.L.; Chumsri, S.; Lou, Y. Next Generation of Immune Checkpoint Therapy in Cancer: New Developments and Challenges. J. Hematol. Oncol. 2018, 11, 39. [Google Scholar] [CrossRef]
  123. Yi, M.; Xu, L.; Jiao, Y.; Luo, S.; Li, A.; Wu, K. The Role of Cancer-Derived MicroRNAs in Cancer Immune Escape. J. Hematol. Oncol. 2020, 13, 25. [Google Scholar] [CrossRef] [Green Version]
  124. Restifo, N.P.; Esquivel, F.; Kawakami, Y.; Yewdell, J.W.; Mulé, J.J.; Rosenberg, S.A.; Bennink, J.R. Identification of Human Cancers Deficient in Antigen Processing. J. Exp. Med. 1993, 177, 265–272. [Google Scholar] [CrossRef]
  125. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel Iii, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ Attenuates Tumour Response to PD-L1 Blockade by Contributing to Exclusion of T Cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  126. Lin, A.; Zhang, H.; Hu, X.; Chen, X.; Wu, G.; Luo, P.; Zhang, J. Age, Sex, and Specific Gene Mutations Affect the Effects of Immune Checkpoint Inhibitors in Colorectal Cancer. Pharmacol. Res. 2020, 159, 105028. [Google Scholar] [CrossRef]
  127. Hirschberger, S.; Hinske, L.C.; Kreth, S. MiRNAs: Dynamic Regulators of Immune Cell Functions in Inflammation and Cancer. Cancer Lett. 2018, 431, 11–21. [Google Scholar] [CrossRef]
  128. Bourquin, C.; Pommier, A.; Hotz, C. Harnessing the Immune System to Fight Cancer with Toll-like Receptor and RIG-I-like Receptor Agonists. Pharmacol. Res. 2020, 154, 104192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Iqubal, A.; Sharma, S.; Najmi, A.K.; Syed, M.A.; Ali, J.; Alam, M.M.; Haque, S.E. Nerolidol Ameliorates Cyclophosphamide-Induced Oxidative Stress, Neuroinflammation and Cognitive Dysfunction: Plausible Role of Nrf2 and NF- ΚB. Life Sci. 2019, 236, 116867. [Google Scholar] [CrossRef] [PubMed]
  130. Iqubal, A.; Sharma, S.; Ansari, M.A.; Najmi, A.K.; Syed, M.A.; Ali, J.; Alam, M.M.; Ahmad, S.; Haque, S.E. Nerolidol Attenuates Cyclophosphamide-Induced Cardiac Inflammation, Apoptosis and Fibrosis in Swiss Albino Mice. Eur. J. Pharmacol. 2019, 863, 172666. [Google Scholar] [CrossRef] [PubMed]
  131. Fukao, T.; Fukuda, Y.; Kiga, K.; Sharif, J.; Hino, K.; Enomoto, Y.; Kawamura, A.; Nakamura, K.; Takeuchi, T.; Tanabe, M. An Evolutionarily Conserved Mechanism for MicroRNA-223 Expression Revealed by MicroRNA Gene Profiling. Cell 2007, 129, 617–631. [Google Scholar] [CrossRef] [Green Version]
  132. Kurkewich, J.L.; Hansen, J.; Klopfenstein, N.; Zhang, H.; Wood, C.; Boucher, A.; Hickman, J.; Muench, D.E.; Grimes, H.L.; Dahl, R. The MiR-23a~27a~24-2 MicroRNA Cluster Buffers Transcription and Signaling Pathways during Hematopoiesis. PLOS Genet. 2017, 13, e1006887. [Google Scholar] [CrossRef] [Green Version]
  133. Wang, G.; Li, B.; Fu, Y.; He, M.; Wang, J.; Shen, P.; Bai, L. MiR-23a Suppresses Proliferation of Osteosarcoma Cells by Targeting SATB1. Tumour Biol. 2015, 36, 4715–4721. [Google Scholar] [CrossRef]
  134. Cho, S.; Wu, C.-J.; Yasuda, T.; Cruz, L.O.; Khan, A.A.; Lin, L.-L.; Nguyen, D.T.; Miller, M.; Lee, H.-M.; Kuo, M.-L.; et al. MiR-23∼27∼24 Clusters Control Effector T Cell Differentiation and Function. J. Exp. Med. 2016, 213, 235–249. [Google Scholar] [CrossRef] [Green Version]
  135. Zheng, J.; Jiang, H.-Y.; Li, J.; Tang, H.-C.; Zhang, X.-M.; Wang, X.-R.; Du, J.-T.; Li, H.-B.; Xu, G. MicroRNA-23b Promotes Tolerogenic Properties of Dendritic Cells in Vitro through Inhibiting Notch1/NF-ΚB Signalling Pathways. Allergy 2012, 67, 362–370. [Google Scholar] [CrossRef] [PubMed]
  136. Jiang, S.; Li, C.; Olive, V.; Lykken, E.; Feng, F.; Sevilla, J.; Wan, Y.; He, L.; Li, Q.-J. Molecular Dissection of the MiR-17-92 Cluster’s Critical Dual Roles in Promoting Th1 Responses and Preventing Inducible Treg Differentiation. Blood 2011, 118, 5487–5497. [Google Scholar] [CrossRef] [PubMed]
  137. Baumjohann, D.; Kageyama, R.; Clingan, J.M.; Morar, M.M.; Patel, S.; de Kouchkovsky, D.; Bannard, O.; Bluestone, J.A.; Matloubian, M.; Ansel, K.M.; et al. The MicroRNA Cluster MiR-17∼92 Promotes TFH Cell Differentiation and Represses Subset-Inappropriate Gene Expression. Nat. Immunol. 2013, 14, 840–848. [Google Scholar] [CrossRef] [PubMed]
  138. O’Connell, R.M.; Zhao, J.L.; Rao, D.S. MicroRNA Function in Myeloid Biology. Blood 2011, 118, 2960–2969. [Google Scholar] [CrossRef] [Green Version]
  139. Möhnle, P.; Schütz, S.V.; van der Heide, V.; Hübner, M.; Luchting, B.; Sedlbauer, J.; Limbeck, E.; Hinske, L.C.; Briegel, J.; Kreth, S. MicroRNA-146a Controls Th1-Cell Differentiation of Human CD4+ T Lymphocytes by Targeting PRKCε. Eur. J. Immunol. 2015, 45, 260–272. [Google Scholar] [CrossRef]
  140. Lu, L.-F.; Boldin, M.P.; Chaudhry, A.; Lin, L.-L.; Taganov, K.D.; Hanada, T.; Yoshimura, A.; Baltimore, D.; Rudensky, A.Y. Function of MiR-146a in Controlling Treg Cell-Mediated Regulation of Th1 Responses. Cell 2010, 142, 914–929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Lu, L.-F.; Thai, T.-H.; Calado, D.P.; Chaudhry, A.; Kubo, M.; Tanaka, K.; Loeb, G.B.; Lee, H.; Yoshimura, A.; Rajewsky, K.; et al. Foxp3-Dependent MicroRNA155 Confers Competitive Fitness to Regulatory T Cells by Targeting SOCS1 Protein. Immunity 2009, 30, 80–91. [Google Scholar] [CrossRef] [Green Version]
  142. Rodriguez, A.; Vigorito, E.; Clare, S.; Warren, M.V.; Couttet, P.; Soond, D.R.; van Dongen, S.; Grocock, R.J.; Das, P.P.; Miska, E.A.; et al. Requirement of Bic/MicroRNA-155 for Normal Immune Function. Science 2007, 316, 608–611. [Google Scholar] [CrossRef] [Green Version]
  143. Neudecker, V.; Haneklaus, M.; Jensen, O.; Khailova, L.; Masterson, J.C.; Tye, H.; Biette, K.; Jedlicka, P.; Brodsky, K.S.; Gerich, M.E.; et al. Myeloid-Derived MiR-223 Regulates Intestinal Inflammation via Repression of the NLRP3 Inflammasome. J. Exp. Med. 2017, 214, 1737–1752. [Google Scholar] [CrossRef]
  144. Ghorbani, S.; Talebi, F.; Chan, W.F.; Masoumi, F.; Vojgani, M.; Power, C.; Noorbakhsh, F. MicroRNA-181 Variants Regulate T Cell Phenotype in the Context of Autoimmune Neuroinflammation. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  145. Zhu, J.; Wang, F.-L.; Wang, H.-B.; Dong, N.; Zhu, X.-M.; Wu, Y.; Wang, Y.-T.; Yao, Y.-M. TNF-α MRNA Is Negatively Regulated by MicroRNA-181a-5p in Maturation of Dendritic Cells Induced by High Mobility Group Box-1 Protein. Sci. Rep. 2017, 7, 12239. [Google Scholar] [CrossRef] [Green Version]
  146. Bi, J.; Zeng, X.; Zhao, L.; Wei, Q.; Yu, L.; Wang, X.; Yu, Z.; Cao, Y.; Shan, F.; Wei, M. MiR-181a Induces Macrophage Polarized to M2 Phenotype and Promotes M2 Macrophage-Mediated Tumor Cell Metastasis by Targeting KLF6 and C/EBPα. Mol. Ther. Nucleic Acids 2016, 5, e368. [Google Scholar] [CrossRef] [Green Version]
  147. Cortez, M.A.; Anfossi, S.; Ramapriyan, R.; Menon, H.; Atalar, S.C.; Aliru, M.; Welsh, J.; Calin, G.A. Role of MiRNAs in Immune Responses and Immunotherapy in Cancer. Genes Chromosomes Cancer 2019, 58, 244–253. [Google Scholar] [CrossRef]
  148. Fontana, L.; Pelosi, E.; Greco, P.; Racanicchi, S.; Testa, U.; Liuzzi, F.; Croce, C.M.; Brunetti, E.; Grignani, F.; Peschle, C. MicroRNAs 17-5p-20a-106a Control Monocytopoiesis through AML1 Targeting and M-CSF Receptor Upregulation. Nat. Cell Biol. 2007, 9, 775–787. [Google Scholar] [CrossRef] [PubMed]
  149. Schmeier, S.; MacPherson, C.R.; Essack, M.; Kaur, M.; Schaefer, U.; Suzuki, H.; Hayashizaki, Y.; Bajic, V.B. Deciphering the Transcriptional Circuitry of MicroRNA Genes Expressed during Human Monocytic Differentiation. BMC Genom. 2009, 10, 595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Aras, S.; Zaidi, M.R. TAMeless Traitors: Macrophages in Cancer Progression and Metastasis. Br. J. Cancer 2017, 117, 1583–1591. [Google Scholar] [CrossRef] [Green Version]
  151. Self-Fordham, J.B.; Naqvi, A.R.; Uttamani, J.R.; Kulkarni, V.; Nares, S. MicroRNA: Dynamic Regulators of Macrophage Polarization and Plasticity. Front. Immunol. 2017, 8, 1062. [Google Scholar] [CrossRef] [Green Version]
  152. Trotta, R.; Chen, L.; Ciarlariello, D.; Josyula, S.; Mao, C.; Costinean, S.; Yu, L.; Butchar, J.P.; Tridandapani, S.; Croce, C.M.; et al. MiR-155 Regulates IFN-γ Production in Natural Killer Cells. Blood 2012, 119, 3478–3485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Wang, P.; Gu, Y.; Zhang, Q.; Han, Y.; Hou, J.; Lin, L.; Wu, C.; Bao, Y.; Su, X.; Jiang, M.; et al. Identification of Resting and Type I IFN-Activated Human NK Cell MiRNomes Reveals MicroRNA-378 and MicroRNA-30e as Negative Regulators of NK Cell Cytotoxicity. J. Immunol. 2012, 189, 211–221. [Google Scholar] [CrossRef] [Green Version]
  154. Perez, L.G.; Kempski, J.; McGee, H.M.; Pelzcar, P.; Agalioti, T.; Giannou, A.; Konczalla, L.; Brockmann, L.; Wahib, R.; Xu, H.; et al. TGF-β Signaling in Th17 Cells Promotes IL-22 Production and Colitis-Associated Colon Cancer. Nat. Commun. 2020, 11, 2608. [Google Scholar] [CrossRef]
  155. Kosaka, A.; Ohkuri, T.; Ikeura, M.; Kohanbash, G.; Okada, H. Transgene-Derived Overexpression of MiR-17-92 in CD8+ T-Cells Confers Enhanced Cytotoxic Activity. Biochem. Biophys. Res. Commun. 2015, 458, 549–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zhang, Z.; Xue, Z.; Liu, Y.; Liu, H.; Guo, X.; Li, Y.; Yang, H.; Zhang, L.; Da, Y.; Yao, Z.; et al. MicroRNA-181c Promotes Th17 Cell Differentiation and Mediates Experimental Autoimmune Encephalomyelitis. Brain Behav. Immun. 2018, 70, 305–314. [Google Scholar] [CrossRef] [PubMed]
  157. Gracias, D.T.; Stelekati, E.; Hope, J.L.; Boesteanu, A.C.; Doering, T.A.; Norton, J.; Mueller, Y.M.; Fraietta, J.A.; Wherry, E.J.; Turner, M.; et al. The MicroRNA MiR-155 Controls CD8(+) T Cell Responses by Regulating Interferon Signaling. Nat. Immunol. 2013, 14, 593–602. [Google Scholar] [CrossRef] [Green Version]
  158. Bodor, J.N.; Boumber, Y.; Borghaei, H. Biomarkers for Immune Checkpoint Inhibition in Non–Small Cell Lung Cancer (NSCLC). Cancer 2020, 126, 260–270. [Google Scholar] [CrossRef]
  159. Rompré-Brodeur, A.; Shinde-Jadhav, S.; Ayoub, M.; Piccirillo, C.A.; Seuntjens, J.; Brimo, F.; Mansure, J.J.; Kassouf, W. PD-1/PD-L1 Immune Checkpoint Inhibition with Radiation in Bladder Cancer: In Situ and Abscopal Effects. Mol. Cancer Ther. 2020, 19, 211–220. [Google Scholar] [CrossRef] [Green Version]
  160. Wu, F.; Zhang, Y.; Sun, B.; McMahon, A.P.; Wang, Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem. Biol. 2017, 24, 252–280. [Google Scholar] [CrossRef] [Green Version]
  161. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 Pathway: Current Researches in Cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
  162. Yang, J.; Zhang, Z.; Chen, C.; Liu, Y.; Si, Q.; Chuang, T.-H.; Li, N.; Gomez-Cabrero, A.; Reisfeld, R.A.; Xiang, R.; et al. MicroRNA-19a-3p Inhibits Breast Cancer Progression and Metastasis by Inducing Macrophage Polarization through Downregulated Expression of Fra-1 Proto-Oncogene. Oncogene 2014, 33, 3014–3023. [Google Scholar] [CrossRef] [Green Version]
  163. Wang, Z.; Han, J.; Cui, Y.; Zhou, X.; Fan, K. MiRNA-21 Inhibition Enhances RANTES and IP-10 Release in MCF-7 via PIAS3 and STAT3 Signalling and Causes Increased Lymphocyte Migration. Biochem. Biophys. Res. Commun. 2013, 439, 384–389. [Google Scholar] [CrossRef]
  164. Ma, S.; Liu, M.; Xu, Z.; Li, Y.; Guo, H.; Ge, Y.; Liu, Y.; Zheng, D.; Shi, J. A Double Feedback Loop Mediated by MicroRNA-23a/27a/24-2 Regulates M1 versus M2 Macrophage Polarization and Thus Regulates Cancer Progression. Oncotarget 2016, 7, 13502–13519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Zhang, Y.; Yang, P.; Sun, T.; Li, D.; Xu, X.; Rui, Y.; Li, C.; Chong, M.; Ibrahim, T.; Mercatali, L.; et al. MiR-126 and MiR-126* Repress Recruitment of Mesenchymal Stem Cells and Inflammatory Monocytes to Inhibit Breast Cancer Metastasis. Nat. Cell Biol. 2013, 15, 284–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Jiang, S.; Zhang, H.-W.; Lu, M.-H.; He, X.-H.; Li, Y.; Gu, H.; Liu, M.-F.; Wang, E.-D. MicroRNA-155 Functions as an OncomiR in Breast Cancer by Targeting the Suppressor of Cytokine Signaling 1 Gene. Cancer Res. 2010, 70, 3119–3127. [Google Scholar] [CrossRef] [Green Version]
  167. Liu, Y.; Lai, L.; Chen, Q.; Song, Y.; Xu, S.; Ma, F.; Wang, X.; Wang, J.; Yu, H.; Cao, X.; et al. MicroRNA-494 Is Required for the Accumulation and Functions of Tumor-Expanded Myeloid-Derived Suppressor Cells via Targeting of PTEN. J. Immunol. 2012, 188, 5500–5510. [Google Scholar] [CrossRef] [PubMed]
  168. Xie, J.; Liu, M.; Li, Y.; Nie, Y.; Mi, Q.; Zhao, S. Ovarian Tumor-Associated MicroRNA-20a Decreases Natural Killer Cell Cytotoxicity by Downregulating MICA/B Expression. Cell. Mol. Immunol. 2014, 11, 495–502. [Google Scholar] [CrossRef] [PubMed]
  169. Xu, S.; Tao, Z.; Hai, B.; Liang, H.; Shi, Y.; Wang, T.; Song, W.; Chen, Y.; OuYang, J.; Chen, J.; et al. MiR-424(322) Reverses Chemoresistance via T-Cell Immune Response Activation by Blocking the PD-L1 Immune Checkpoint. Nat. Commun. 2016, 7, 11406. [Google Scholar] [CrossRef]
  170. Chen, R.; Alvero, A.B.; Silasi, D.A.; Kelly, M.G.; Fest, S.; Visintin, I.; Leiser, A.; Schwartz, P.E.; Rutherford, T.; Mor, G. Regulation of IKKbeta by MiR-199a Affects NF-KappaB Activity in Ovarian Cancer Cells. Oncogene 2008, 27, 4712–4723. [Google Scholar] [CrossRef] [Green Version]
  171. Yang, P.; Li, Q.-J.; Feng, Y.; Zhang, Y.; Markowitz, G.J.; Ning, S.; Deng, Y.; Zhao, J.; Jiang, S.; Yuan, Y.; et al. TGF-β-MiR-34a-CCL22 Signaling-Induced Treg Cell Recruitment Promotes Venous Metastases of HBV-Positive Hepatocellular Carcinoma. Cancer Cell 2012, 22, 291–303. [Google Scholar] [CrossRef] [Green Version]
  172. Li, H.; Gupta, S.; Du, W.W.; Yang, B.B. MicroRNA-17 Inhibits Tumor Growth by Stimulating T-Cell Mediated Host Immune Response. Oncoscience 2014, 1, 531–539. [Google Scholar] [CrossRef] [Green Version]
  173. Wang, J.; Yu, F.; Jia, X.; Iwanowycz, S.; Wang, Y.; Huang, S.; Ai, W.; Fan, D. MicroRNA-155 Deficiency Enhances the Recruitment and Functions of Myeloid-Derived Suppressor Cells in Tumor Microenvironment and Promotes Solid Tumor Growth. Int. J. Cancer 2015, 136, E602–E613. [Google Scholar] [CrossRef] [Green Version]
  174. Wang, Y.; Zhang, X.; Li, H.; Yu, J.; Ren, X. The Role of MiRNA-29 Family in Cancer. Eur. J. Cell Biol. 2013, 92, 123–128. [Google Scholar] [CrossRef]
  175. Yin, Y.; Cai, X.; Chen, X.; Liang, H.; Zhang, Y.; Li, J.; Wang, Z.; Chen, X.; Zhang, W.; Yokoyama, S.; et al. Tumor-Secreted MiR-214 Induces Regulatory T Cells: A Major Link between Immune Evasion and Tumor Growth. Cell Res. 2014, 24, 1164–1180. [Google Scholar] [CrossRef] [PubMed]
  176. Chen, L.; Gibbons, D.L.; Goswami, S.; Cortez, M.A.; Ahn, Y.-H.; Byers, L.A.; Zhang, X.; Yi, X.; Dwyer, D.; Lin, W.; et al. Metastasis Is Regulated via MicroRNA-200/ZEB1 Axis Control of Tumour Cell PD-L1 Expression and Intratumoral Immunosuppression. Nat. Commun. 2014, 5, 5241. [Google Scholar] [CrossRef]
  177. Yang, N.; Zhu, S.; Lv, X.; Qiao, Y.; Liu, Y.-J.; Chen, J. MicroRNAs: Pleiotropic Regulators in the Tumor Microenvironment. Front. Immunol. 2018, 9, 2491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Lin, R.; Chen, L.; Chen, G.; Hu, C.; Jiang, S.; Sevilla, J.; Wan, Y.; Sampson, J.H.; Zhu, B.; Li, Q.-J. Targeting MiR-23a in CD8+ Cytotoxic T Lymphocytes Prevents Tumor-Dependent Immunosuppression. J. Clin. Invest. 2014, 124, 5352–5367. [Google Scholar] [CrossRef]
  179. Pyfferoen, L.; Mestdagh, P.; Vergote, K.; De Cabooter, N.; Vandesompele, J.; Lambrecht, B.N.; Vermaelen, K.Y. Lung Tumours Reprogram Pulmonary Dendritic Cell Immunogenicity at the MicroRNA Level. Int. J. Cancer 2014, 135, 2868–2877. [Google Scholar] [CrossRef]
  180. Wei, J.; Wang, F.; Kong, L.-Y.; Xu, S.; Doucette, T.; Ferguson, S.D.; Yang, Y.; McEnery, K.; Jethwa, K.; Gjyshi, O.; et al. MiR-124 Inhibits STAT3 Signaling to Enhance T Cell-Mediated Immune Clearance of Glioma. Cancer Res. 2013, 73, 3913–3926. [Google Scholar] [CrossRef] [Green Version]
  181. Yang, J.; Liu, R.; Deng, Y.; Qian, J.; Lu, Z.; Wang, Y.; Zhang, D.; Luo, F.; Chu, Y. MiR-15a/16 Deficiency Enhances Anti-Tumor Immunity of Glioma-Infiltrating CD8+ T Cells through Targeting MTOR. Int. J. Cancer 2017, 141, 2082–2092. [Google Scholar] [CrossRef] [Green Version]
  182. Li, Q.; Johnston, N.; Zheng, X.; Wang, H.; Zhang, X.; Gao, D.; Min, W. MiR-28 Modulates Exhaustive Differentiation of T Cells through Silencing Programmed Cell Death-1 and Regulating Cytokine Secretion. Oncotarget 2016, 7, 53735–53750. [Google Scholar] [CrossRef] [Green Version]
  183. Wei, J.; Nduom, E.K.; Kong, L.-Y.; Hashimoto, Y.; Xu, S.; Gabrusiewicz, K.; Ling, X.; Huang, N.; Qiao, W.; Zhou, S.; et al. MiR-138 Exerts Anti-Glioma Efficacy by Targeting Immune Checkpoints. Neuro Oncol. 2016, 18, 639–648. [Google Scholar] [CrossRef] [Green Version]
  184. Abdelrahman, M.M.; Fawzy, I.O.; Bassiouni, A.A.; Gomaa, A.I.; Esmat, G.; Waked, I.; Abdelaziz, A.I. Enhancing NK Cell Cytotoxicity by MiR-182 in Hepatocellular Carcinoma. Hum. Immunol. 2016, 77, 667–673. [Google Scholar] [CrossRef] [PubMed]
  185. Chen, B.; Xia, Z.; Deng, Y.-N.; Yang, Y.; Zhang, P.; Zhu, H.; Xu, N.; Liang, S. Emerging MicroRNA Biomarkers for Colorectal Cancer Diagnosis and Prognosis. Open Biol. 2019, 9, 180212. [Google Scholar] [CrossRef] [Green Version]
  186. Kim, D.H.; Khan, H.; Ullah, H.; Hassan, S.T.S.; Šmejkal, K.; Efferth, T.; Mahomoodally, M.F.; Xu, S.; Habtemariam, S.; Filosa, R.; et al. MicroRNA Targeting by Quercetin in Cancer Treatment and Chemoprotection. Pharmacol. Res. 2019, 147, 104346. [Google Scholar] [CrossRef]
  187. Gomarasca, M.; Maroni, P.; Banfi, G.; Lombardi, G. MicroRNAs in the Antitumor Immune Response and in Bone Metastasis of Breast Cancer: From Biological Mechanisms to Therapeutics. Int. J. Mol. Sci. 2020, 21, 2805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Bouchie, A. First MicroRNA Mimic Enters Clinic. Nat. Biotechnol. 2013, 31, 577. [Google Scholar] [CrossRef]
  189. Shah, M.Y.; Ferrajoli, A.; Sood, A.K.; Lopez-Berestein, G.; Calin, G.A. MicroRNA Therapeutics in Cancer—An Emerging Concept. EBioMedicine 2016, 12, 34–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Cantafio, M.E.G.; Nielsen, B.S.; Mignogna, C.; Arbitrio, M.; Botta, C.; Frandsen, N.M.; Rolfo, C.; Tagliaferri, P.; Tassone, P.; Martino, M.T.D. Pharmacokinetics and Pharmacodynamics of a 13-Mer LNA-Inhibitor-MiR-221 in Mice and Non-Human Primates. Mol. Ther. Nucleic Acids 2016, 5. [Google Scholar] [CrossRef] [Green Version]
  191. 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]
  192. Van Roosbroeck, K.; Fanini, F.; Setoyama, T.; Ivan, C.; Rodriguez-Aguayo, C.; Fuentes-Mattei, E.; Xiao, L.; Vannini, I.; Redis, R.S.; D’Abundo, L.; et al. Combining Anti-Mir-155 with Chemotherapy for the Treatment of Lung Cancers. Clin. Cancer Res. 2017, 23, 2891–2904. [Google Scholar] [CrossRef] [Green Version]
  193. Paranjape, T.; Slack, F.J.; Weidhaas, J.B. MicroRNAs: Tools for Cancer Diagnostics. Gut 2009, 58, 1546–1554. [Google Scholar] [CrossRef] [PubMed]
  194. Cortez, M.A.; Ivan, C.; Valdecanas, D.; Wang, X.; Peltier, H.J.; Ye, Y.; Araujo, L.; Carbone, D.P.; Shilo, K.; Giri, D.K.; et al. PDL1 Regulation by P53 via MiR-34. J. Natl. Cancer Inst. 2016, 108. [Google Scholar] [CrossRef] [Green Version]
  195. Wiggins, J.F.; Ruffino, L.; Kelnar, K.; Omotola, M.; Patrawala, L.; Brown, D.; Bader, A.G. Development of a Lung Cancer Therapeutic Based on the Tumor Suppressor MicroRNA-34. Cancer Res. 2010, 70, 5923–5930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs Bind to Toll-like Receptors to Induce Prometastatic Inflammatory Response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef] [Green Version]
  197. Yin, Z.; Deng, Z.; Zhao, W.; Cao, Z. Searching Synergistic Dose Combinations for Anticancer Drugs. Front. Pharmacol. 2018, 9, 535. [Google Scholar] [CrossRef] [PubMed]
  198. Lakhal, S.; Wood, M.J.A. Exosome Nanotechnology: An Emerging Paradigm Shift in Drug Delivery: Exploitation of Exosome Nanovesicles for Systemic in Vivo Delivery of RNAi Heralds New Horizons for Drug Delivery across Biological Barriers. Bioessays 2011, 33, 737–741. [Google Scholar] [CrossRef] [PubMed]
  199. Fischer, H.C.; Chan, W.C.W. Nanotoxicity: The Growing Need for in Vivo Study. Curr. Opin. Biotechnol. 2007, 18, 565–571. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Showing the involvement of miRNAs as the central regulator of immunity via NF-kB pathway. TIRAP, TRAM, TLR4, TNF-R, RIP1, FADD, TRAF-2, TRADD, IRAK, TAB-2, TGF-β-activated kinase 1/MAP3K7-binding protein 2; TAK, IKK, and CYLD.
Figure 1. Showing the involvement of miRNAs as the central regulator of immunity via NF-kB pathway. TIRAP, TRAM, TLR4, TNF-R, RIP1, FADD, TRAF-2, TRADD, IRAK, TAB-2, TGF-β-activated kinase 1/MAP3K7-binding protein 2; TAK, IKK, and CYLD.
Cancers 13 06145 g001
Figure 2. Showing the immunotherapeutic approach of miRNA in cancer.
Figure 2. Showing the immunotherapeutic approach of miRNA in cancer.
Cancers 13 06145 g002
Table 1. Examples of oncogenic miRNAs.
Table 1. Examples of oncogenic miRNAs.
S. No.Type of CancermiRNAs Involved References
1Acute Myeloid LeukemiamiR-196b; miR-126;
miR-9; miR-17-92
Oncogenic[59]
2GastricmiR-421; miR-196aOncogenic[56,57]
3BreastmiR-181a/b, miR-21
miR-34, miR-34 b/c
Oncogenic
Tumor Suppressor
[60,61]
4Non-Small Cell Lung CancermiR-27aOncogenic[62]
5Renal Cell CarcinomasmiR-30a/cOncogenic[63,64]
6ColorectalmiR-34a, miR-34b, miR34b/cTumor suppressor[65,66,67]
7ProstatemiR-34a, miR-34b, miR34b/cTumor suppressor[68,69]
8LungmiR-34a, miR-34b, miR34b/cTumor Suppressor[70,71,72]
9LivermiR-34bTumor Suppressor[73]
10OstosarcomamiR-34a, miR34b, miR-192Tumor Suppressor[74,75,76]
11haematological neoplasmsmiR-34a, miR-34b-5p, miR-34c, miR-34b/cTumor Suppressor[77,78,79]
12Lymphophytic leukemiamiR-15/16Tumor Suppressor[80]
Table 2. miRNAs biomarkers and their role in various types of cancer [11,95,99,107].
Table 2. miRNAs biomarkers and their role in various types of cancer [11,95,99,107].
Sl. No.Type of CancermiRNAs Biomarkers
PredictivePrognosticDiagnostic
1Prostate cancermiR-21miR-20a; miR -21; miR-141miR-141, -375; let-7c, -7e
miR-141
2Lung cancermiR-128bmiR-221; let-7a, -137; miR-372; miR -182; miR-15b, -16miR-16, miR-17, -19b, miR-200 family; miR-29c, -30c
3Breast cancermiR-125bmiR-210; miR-10bmiR-21, -30a; miR-141, -145; miR- 801
4Ovarian cancermiR-181a,b, -213; miR-23a, -27a; let-7g, 3pmiR-200 family; miR-410, -645miR-126, -127; miR-200 & let-7 family; miR-21, -29a
5Liver cancermiR-21, -200bmiR-200 family; miR-21, -22, -26miR-200c, -203, -224; miR-222, -223
Table 3. Role of miRNAs in Cancer immune escape [127].
Table 3. Role of miRNAs in Cancer immune escape [127].
miRNAEffect/MechanismCancer Types
miR-24-3pIncreased T reg cells activity and reduced Th17 proliferation via targeting fibroblast growth factor 11Nasopharyngeal cancer
miR-183Reduced the activity of NK cellsLung cancer
miR-23aReduced activity of CD8+ cells via acting on B lymphocyte-induced maturation protein-1
miR-155Reduced activity of MDSCs via acing on hypoxia inducing factor-αpancreatic cancer
miR-212-3pIncreased immune response of DC via acting on Regulatory Factor X Associated Protein
miR-92a-3pIncreased tumor-associated macrophages and IL-6 levelLiposarcoma
miR-25-3pIncreased tumor-associated macrophages and IL-6 level
miR-155Reduced activity of MDSCs via acing on hypoxia inducing factor-αSkin cancer
miR-210Reduced activity of MDSCs via acing on C-X-C motif chemokine 12 and IL-16
miR-34aReduced recruitment of T reg cells via acting on C-X-C motif chemokine 12Hepatic cancer
miR-20a and miR-17-5pReduced activity of MDSCs via acing on Reduced activity of MDSCs via acing on C-X-C motif chemokine 12 and IL-16Colon cancer
miR-494Reduced activity of MDSCs via acing on Phosphatase and tensin homologBreast cancer
miR-222-3pIncreased polarization of M2 macrophage via acting on suppressor of cytokine signaling 3Ovarian cancer
Table 4. Showing the modulatory effect of miRNAs via immune cells on tumorigenesis.
Table 4. Showing the modulatory effect of miRNAs via immune cells on tumorigenesis.
miRNAImmune Cells/Cancer CellsEffectCancer TypesReferences
miR-19a-3pMacrophages (M2)Polarization of macrophage and amelioration of cancer progression and metastasisBreast cancer[162]
miR-21T reg cellsReduce the proliferation of T reg cells leading to reduced survival and proliferation[163]
miR-23a/27a/24-2MacrophagePolarization of M2 macrophage and promote tumour growth[164]
miR-126Cancerous cellsReduced monocyte recruitment and stimulated metastasis[165]
miR-146aMacrophagesActivation of NF-kB and promote tumour cell invasion[140]
miR-155Cancerous cellsActivation of JAK-STAT pathway and proliferation of tumor cells.[166]
miR-223Macrophages (M2)Differentiation of macrophage and stimulate tumour cells invasion[88]
miR-494MDSCsMDSCs accumulation stimulate tumor cells invasion and metastasis[167]
miR-20aCancerous cellsSuppress NK-mediated antitumor effect and promote tumour invasion as well as proliferationOvarian cancer[168]
miR-424Cancerous cellsActivate T cells and stimulate sensitivity of tucells towards chemotherapy[169]
miR-199aCancerous cellsProduction of cytokines and progression of tumorigenesis[170]
miR-34a/cCancerous cellsSuppress NK-mediated antitumor effect and promote tumour invasion as well as proliferationSkin cancer[171]
miR-17T cellsAlters the function of T cells and promote tumour growth[172]
miR-155MDSCsIncreased HIF-α and promote tumor growth[173]
miR-29Cancer cellsMitigate the NK cell’s function, T cells and promote the growth of tumorsSolid tumors[174]
miR-214CD4+ and CD25+ T cellsStimulate T reg cells and promote tumour growth[175]
miR-146aCancer cellsReduced IL-8, TRAF-6 and exhibit the antitumor effectGastric cancer[176]
miR-5, 18 and 22Cancer cellsReduced PD-1 expression 6 and exhibit antitumor effect[177]
miR-23aT cellsInhibit CD8+ function and promote tumour growth and TGF-β induced tumour invasionLung cancer[178]
miR-155Dendritic cellStimulate dendritic cell maturation and activation of T cells[179]
miR-124T cellsInhibit STAT-3 and promote T cell-induced killing of tumour cellsGlioblastoma[180]
miR-15a and 16aT cellsReduced PD-1 and increased CD8+ mediated antitumor effect[181]
miR-28T cellsEnervation of T cells and reduced PD-1[182]
miR-138T cellsDiminished expression of PD-1 and exhibit the anti-tumor effect[183]
miR-182NK cellsStimulate the tumour-killing potential of NK cells and increase the release of perforin-1Hepatic cancer[184]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pottoo, F.H.; Iqubal, A.; Iqubal, M.K.; Salahuddin, M.; Rahman, J.U.; AlHajri, N.; Shehadeh, M. miRNAs in the Regulation of Cancer Immune Response: Effect of miRNAs on Cancer Immunotherapy. Cancers 2021, 13, 6145. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13236145

AMA Style

Pottoo FH, Iqubal A, Iqubal MK, Salahuddin M, Rahman JU, AlHajri N, Shehadeh M. miRNAs in the Regulation of Cancer Immune Response: Effect of miRNAs on Cancer Immunotherapy. Cancers. 2021; 13(23):6145. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13236145

Chicago/Turabian Style

Pottoo, Faheem Hyder, Ashif Iqubal, Mohammad Kashif Iqubal, Mohammed Salahuddin, Jawad Ur Rahman, Noora AlHajri, and Mustafa Shehadeh. 2021. "miRNAs in the Regulation of Cancer Immune Response: Effect of miRNAs on Cancer Immunotherapy" Cancers 13, no. 23: 6145. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13236145

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

Article Metrics

Back to TopTop