Skip to main content
British Journal of Clinical Pharmacology logoLink to British Journal of Clinical Pharmacology
. 2016 Jun 3;82(3):659–672. doi: 10.1111/bcp.12987

Therapeutic nucleic acids: current clinical status

Kannan Sridharan 1, Nithya Jaideep Gogtay 2,
PMCID: PMC5338117  PMID: 27111518

Abstract

Deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) are simple linear polymers that have been the subject of considerable research in the last two decades and have now moved into the realm of being stand‐alone therapeutic agents. Much of this has stemmed from the appreciation that they carry out myriad functions that go beyond mere storage of genetic information and protein synthesis. Therapy with nucleic acids either uses unmodified DNA or RNA or closely related compounds. From both a development and regulatory perspective, they fall somewhere between small molecules and biologics. Several of these compounds are in clinical development and many have received regulatory approval for human use. This review addresses therapeutic uses of DNA based on antisense oligonucleotides, DNA aptamers and gene therapy; and therapeutic uses of RNA including micro RNAs, short interfering RNAs, ribozymes, RNA decoys and circular RNAs. With their specificity, functional diversity and limited toxicity, therapeutic nucleic acids hold enormous promise. However, challenges that need to be addressed include targeted delivery, mass production at low cost, sustaining efficacy and minimizing off‐target toxicity. Technological developments will hold the key to this and help accelerate drug approvals in the years to come.

Keywords: antisense oligonucleotides, aptamers, gene therapy, ribozymes, RNA decoys

Introduction

Deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) are simple linear polymers that consist of only four major subunits, yet these molecules carry out myriad functions both within the cell and in the laboratory 1. Early assessment of nucleic acid function was rather narrow and restricted, but research in the past few decades has seen remarkable progress in developing nucleic acid‐based therapeutics. The progress has covered diverse fields of research and a significant number of scientists and engineers are now involved in this area 2. A series of pivotal discoveries in the last three decades has made this possible. First, a large body of work has obviously followed the decoding of the human genome that unlocked several molecular pathways that are important in disease. Secondly, several types of RNA with complex biological functions have been discovered in addition to messenger RNA (mRNA) and transfer RNA (tRNA) 1. For example, two non‐coding RNA types that were not considered essential but are now extremely relevant to therapeutics are the microRNA (miRNA) and the short interfering RNA (siRNA). Thirdly, the appreciation that RNAs can act as enzymes has led to the development of RNA analogues with useful or unusual properties. Of the analogues, the locked nucleic acids or LNAs have found therapeutic applications. In view of their polar nature, the cellular delivery of nucleic acids is poor relative to conventional low molecular weight drugs. The fourth major advance has been enhancing the bioavailability of nucleic acid‐based drugs. None of these would have been possible without technological advances in DNA synthesis, including the de novo synthesis of increasingly longer DNA constructs and use of DNA shuffling, bioprospecting, combinatorial chemistry, high throughput screening and genetic engineering of viruses. Advances in synthetic biology, systems biology, computational biology, bioinformatics and nanotechnology among others have greatly aided development in this area and set a new paradigm for the nucleic acids in therapy 3, 4, 5.

The fundamental basis of using nucleic acids except for gene therapy in therapeutics is either inhibition of DNA or RNA expression, thereby halting production of abnormal protein related to a disease while leaving all other proteins unaffected 6. Therapeutic nucleic acids (TNAs) are nucleic acids themselves or closely related compounds used to treat disease. Although various types of TNAs exist, they share a common mechanism of action that is mediated by sequence‐specific recognition of endogenous nucleic acids through Watson–Crick base pairing 7. Their drug development has specific requirements that are unique as they fall somewhere between small molecules and biologics. TNA are charged, high molecular weight compounds with physicochemical properties different from small molecule drugs, and are unstable in a biological environment. In addition, TNA have to be delivered to the correct intracellular compartment. Because they are chemically synthesized, regulators considered that they are new chemical entities (NCEs). However, the above‐mentioned characteristics make them closer to new biological entities (NBEs) 8. Clinical trial regulations related to TNAs and subsequent approval for human use are more complex than for NCEs 9.

The present review divides nucleic acid therapeutics broadly into DNA therapeutics (antisense oligonucleotides, DNA aptamers and gene therapy) and RNA therapeutics (micro RNAs, short interfering RNAs, ribozymes, RNA decoys and circular RNAs). This review will restrict its focus to nucleic acids that are either in clinical development or already available for clinical use.

DNA Therapeutics

Anti‐sense oligonucleotides (ASOs) and DNA aptamers

ASOs are single, short‐stranded sequences, 8–50 base pairs in length, binding to the target mRNA by means of standard Watson–Crick base pairing. After an ASO binds with the mRNA, either the target complex will be degraded by endogenous cellular RNase H or a functional blockade of mRNA occurs due to steric hindrance 10, 11.

Aptamers (from the Latin aptus, to fit), also called ‘chemical antibodies’, are single‐stranded synthetic DNA or RNA molecules, 56–120 nucleotides long, that can bind the nucleotide coding for proteins with high affinity and thus serve as decoys. DNA aptamers are short single‐stranded oligonucleotide sequences similar to ASO with very high affinity for the target nucleic acids through structural recognition 12. DNA aptamers that target coding nucleotides for lysozyme, thrombin, human immunodeficiency virus trans‐acting responsive element, hemin, interferon γ, vascular endothelial growth factor, prostate specific antigen, dopamine and heat shock factor are still under development 13. Aptamers are isolated from a large pool of nucleic acids by a process called Systematic Evolution of Ligands by EXponential Enrichment, or SELEX 14. AptaBid is another selection process for generating appropriate aptamers 15. They represent attractive alternatives to monoclonal antibodies because they are non‐toxic, non‐immunogenic, have good tissue penetration, are straightforward to make, can be modified easily in vitro and can be given either intravenously or subcutaneously 16, 17, 18, 19.

The ASOs that were initially developed were unmodified and highly susceptible to nuclease enzyme 20, 21. Chemical modifications to the backbone and ribose component of the nucleotides have improved their stability, binding strength and specificity to the target nucleic acid 22. Second‐generation ASO may have a phosphodiester or phosphoramidate or phosphorothioate modification 23. Of these, phospohorothioate modification, wherein one of the non‐bridging oxygen atoms in the backbone is replaced by a sulphur atom, was the most common type in second‐generation ASO 24. Phosphorothioate ASOs showed enhanced uptake by the target cells, more stability and so longer action, and high target specificity 25. Modification of the ribose sugar at the 2′ position (most commonly either 2′‐O‐methyl or 2′‐O‐methoxy ethyl) further increases the resistance to exonucleases of ASOs modified by phosphorothioate replacement in the backbone 26. ASO with a chemically modified non‐sugar furanose ring show additional improvements in nuclease stability, target affinity and pharmacokinetic profiles. Locked nucleic acid (LNA), peptide nucleic acid (PNA) and morpholino phosphoroamidates (MF) are the three most commonly used third‐generation ASOs 27. The efficacy and safety of various ASOs is being explored in various currently incurable neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and other neurodegenerative diseases 28. However, till now, only two ASOs have been approved by the United States Food and Drug Administration (US FDA) to be used on human beings, namely, fomivirsen, a first‐generation ASO, and mipomersen, a second‐generation ASO 29, 30.

Fomivirsen

The first ASO approved for treating humans is fomivirsen, developed by Isis pharmaceuticals for the management of cytomegalovirus (CMV) retinitis in 1998 31. Fomivirsen is a phosphorothioate ASO (first‐generation) and binds to the IE2 fragment of mRNA of CMV that is involved in viral replication. Fomivirsen is administered as an intravitreal injection and has been found to have long duration of action; plasma concentrations are negligible 32. Metabolism of fomivirsen is locally mediated by exonuclease enzyme in the eye. No significant drug interactions have been observed with fomivirsen except that it should not be administered within 4 weeks following administration of the antiretroviral agent cidofovir 33. The drug is administered intravitreally once a week and the median time for progression of retinitis in patients was found to be 71–90 days 34. No serious adverse drug reactions were observed during clinical trials for intravitreal fomivirsen 35. Reports of occurrence of reversible bull's‐eye maculopathy with fomivirsen administration have been documented without any effect on vision 36. Following the advent of highly active antiretroviral drugs for treating infections with human immunodeficiency virus, a decline in the rate of CMV retinitis in patients was observed. Hence, fomivirsen was withdrawn by the marketing authorization holder for commercial reasons.

Mipomersen

Mipomersen is an ASO that acts as an inhibitor of apolipoprotein B‐100 synthesis, which is an essential component of both very‐low‐density lipoprotein (VLDL) and low‐density lipoprotein (LDL) 37. It has been approved for patients diagnosed with homozygous familial hypercholesterolemia (HoFH). Familial hypercholesterolemia is an autosomal dominant genetic disorder resulting from mutations of the low‐density lipoprotein‐cholesterol (LDL‐C) receptor, apolipoprotein B (apo B), or pro‐protein convertase subtilisin/kexin 9 (PCSK9) 38. The standard of care before approval of mipomersen for patients with HoFH was weekly or bi‐weekly LDL apheresis. Mipomersen binds to the mRNA of the abnormal apo B in patients with HoFH and inhibits the synthesis of mutated apo B protein so that LDL‐C is phagocytosed in the hepatocytes and cleared from the blood stream. Mipomersen is a long‐acting ASO with an elimination half‐life ranging between 21 and 33 days depending on the administered dose, and attributed largely to the fact that almost 90% of the compound is bound to plasma proteins 30. Hence, mipomersen is administered once a week as subcutaneous injection. At 26 weeks a mean reduction of LDL was observed between 25% and 36% compared with placebo 39, 40. The most commonly observed adverse drug reactions include local injection site reactions such as pruritis, erythema and pain and systemic reactions such as fatigue, pyrexia, chills, malaise, myalgia and arthralgia 39, 40, 41. As liver enzymes were found to be elevated with the drug administration, mipomersen is contra‐indicated in patients with moderate or severe hepatic impairment.

ASO in evaluation in clinical trials

ASOs are being evaluated for various cancer states and neurodegenerative diseases. Table 1 lists the clinical studies identified from clinicaltrials.gov using the key word ‘Antisense oligonucleotides’. Of these agents a promising one is Factor XI ASO (FXI ASO) that has been proven to have potential in reducing the incidence of venous thromboembolism. Recently, Büller et al. 42 conducted a randomized, open‐label study amongst 300 patients undergoing total knee arthroplasty receiving either FXI ASO or enoxaparin. The incidence of thromboembolism was observed to be 30% with enoxaparin while only 27% and 4% with a lower and higher dose of FXI ASO respectively. Additionally, the rate of bleeding was also significantly lower in the FXI ASO (3%) group as compared to enoxaparin (8%). The study results suggest that strategies to lower factor XI levels can be more effective in reducing postoperative thrombosis than conventional anticoagulants, without increasing the risk of bleeding.

Table 1.

Summary of registered clinical trials with ASO in clinical trials.gov

Study id; year of registration; country of conduct of study Sponsors Disease condition Phase of clinical trial; target mRNA for ASO
NCT02627820; 2015; United States GlaxoSmithKline Isis Pharmaceuticals Senile cardiac amyloidosis Transthyretin
NCT02406833; 2015; Germany Isarna Therapeutics GmbH Glaucoma patients undergoing trabeculectomy Transforming growth factor‐β.
NCT00780052; 2008; United States of America University of Pennsylvania Advanced haematological malignancies (acute myeloid or lymphoid leukaemia; or myeloproliferative disorder (MPD) including chronic myelogenous leukaemia (CML); or myelodysplastic syndrome (MDS); or non‐Hodgkin's lymphoma (including CLL); or multiple myeloma) Phase 1; c‐myb ASO
NCT00159250; 2005; United Kingdom Imperial College, London Duchenne muscular dystrophy Phase 1/2 studies against AVI‐4658 PMO
NCT02417753; 2015; United States of America National Cancer Institute Malignant ascites Phase 2; STAT3
NCT02507583; 2015; United States of America Thomas Jefferson University Malignant glioma Phase 1; Insulin like growth factor‐1
NCT00466583; 2007; United States of America Enzon Pharmaceuticals, Inc. Advanced solid tumours or lymphoma Phase 1; Hypoxia‐inducible factor‐1α
NCT01550523; 2012; United States of America Thomas Jefferson University Recurrent malignant glioma Phase 1; Insulin like growth factor receptor
NCT00100672; 2005; United States of America INSYS Therapeutics Inc Advanced cancer Phase 1; c‐raf
NCT01120288; 2010; United States of America National Cancer Institute Advanced solid tumours with metastasis Phase 1; Hypoxia inducible factor‐1α
NCT01159028; 2010; United States of America Bio‐Path Holdings, Inc. Recurrent adult acute myeloid leukaemia or acute lymphoblastic leukaemia or myelodysplastic syndrome or Ph1 Positive chronic myeloid leukaemia Phase 1; Growth factor receptor bound protein‐2
NCT00959868; 2009; Canada Vancouver Coastal Health Superficial bladder tumour Phase 1; Heat shock protein 27
NCT00048321; 2002; United States of America Isis Pharmaceuticals Rheumatoid arthritis Phase 2; Tumor necrosis factor
NCT02423590; 2014; United Kingdom Queen Mary University of London Advanced squamous cell lung cancer Phase 2; Heat shock protein 27
NCT00445913; 2007; United States of America University of Pittsburgh Type 1 diabetes mellitus Phase 1; CD40, CD80 and CD86
NCT01470911; 2011; Germany NCT01554319; 2012; Germany
NCT01577953; 2012; Germany
Sterna Biologicals GmbH & Co. KG Bronchial asthma Phase 1; GATA‐3
NCT01743768; 2012; Germany Sterna Biologicals GmbH & Co. KG Bronchial asthma Phase 2; GATA‐3
NCT02564354; 2015; United States of America ProQR Therapeutics Cystic fibrosis Phase 1; ΔF508
NCT02532764; 2015; United States of America ProQR Therapeutics Cystic fibrosis Phase 1/2; ΔF508
NCT00021749; United States of America Genta Incorporated Chronic lymphocytic leukaemia Phase 1/2; Bcl‐2
NCT00002592; 1999; United States of America Abramson Cancer Center of the University of Pennsylvania Chronic myelogenous leukaemia Phase 2; c‐myb
NCT00487786; 2007; United States of America OncoGenex Technologies Prostate cancer, ovarian cancer, non‐small cell lung cancer, breast cancer, bladder cancer Phase 1; Heat shock protein 27
NCT00005032; 2000; United States of America University of Chicago Recurrent small cell lung cancer Phase 1/2; Bcl‐2
NCT00059813; 2003; United States of America National Cancer Institute (NCI) Metastatic renal cell cancer Phase 2; Bcl‐2
NCT00078234; 2004; United States of America Genta Incorporated Chronic lymphocytic leukaemia Phase 1/2; Bcl‐2
NCT00017251; 2001; United States of America National Cancer Institute (NCI) Extensive‐Stage Small Cell Lung Cancer Phase 1; Bcl‐2
NCT00471432: 2007; United States of America NCIC Clinical Trials Group Bladder cancer, breast cancer, kidney cancer, lung cancer, ovarian cancer, prostate cancer Phase 1; Clusterin
NCT00063934; 2003; United States of America National Cancer Institute (NCI) Breast cancer Phase 1/2; Bcl‐2
NCT01563302; 2012; United States of America Isis Pharmaceuticals Lymphoma Phase 1/2; STAT 3
NCT00016263; 2001; United States of America Genta Incorporated Malignant melanoma Phase 3; Bcl‐2
NCT00258375; 2005; United States of America NCIC Clinical Trials Group Metastatic breast cancer Phase 2; Clusterin
NCT00070083; 2003; United Kingdom British Columbia Cancer Agency Diffuse large B cell lymphoma Phase 1; Bcl‐2
NCT00017602; 2001; United States of America Genta Incorporated Multiple myeloma Phase 3; Bcl‐2
NCT00365781; 2006; United States of America Isis Pharmaceuticals Type 2 diabetes mellitus Phase 1; Tyrosine phosphatase 1B
NCT00030641; 2002; United States of America Genta Incorporated Non small cell lung cancer Phase 2/3; Bcl‐2
NCT00070343; 2003; United States of America Jonsson Comprehensive Cancer Center Malignant melanoma Bcl‐2
NCT00024440; 2001; United States of America Genta Incorporated Chronic lymphocytic leukaemia Phase 3; Bcl‐2
NCT00085228; 2004; Belgium European Organisation for Research and Treatment of Cancer Adenocarcinoma prostate Phase 2; Bcl‐2
NCT00543205; 2007; United States of America Genta Incorporated Melanoma Phase 2/3; Bcl‐2
NCT00054106; 2003; United States of America NCIC Clinical Trials Group Prostate cancer Phase 1; Bcl‐2
NCT01710852; 2012; United States of America Isis Pharmaceuticals Paroxysmal atrial fibrillation Phase 2; CRP
NCT00004870; 2000; United States of America The University of Texas Health Science Center at San Antonio Colorectal cancer Phase 1/2; Bcl‐2
NCT00003236; 1999; United States of America Eastern cancer cooperative agency Metastatic breast cancer Phase 2; Protein kinase‐alpha
NCT01120470; 2010; United Kingdom British Columbia Cancer Agency Castration resistant prostate cancer Phase 2; Heat shock protein‐27
NCT00543231; 2007; United States of America Genta Incorporated Solid tumours Phase 1; Bcl‐2
NCT00636545 ; 2007; United States of America Genta Incorporated Solid tumours Phase 1; Bcl‐2
NCT00896857; 2009; United States of America Comprehensive Cancer Center of Wake Forest University Breast cancer Bcl‐2
NCT00024648; 2001; United States of America INSYS Therapeutics Inc Advanced malignancies Phase 1; Raf‐1
NCT00024661; 2001; United States of America INSYS Therapeutics Inc Advanced solid tumours Phase 1; Raf‐1
NCT02079688; 2014; United States of America Sterna Biologicals GmbH & Co. KG Atopic eczema Phase 2; GATA‐3
NCT01839604; 2013; Hong Kong AstraZeneca Metastatic hepatocellular carcinoma Phase 1; STAT‐3
NCT00264966; 2005; Canada Pharmaxis Asthma Phase ½; CCR‐3
NCT00056173; 2003; United States of America Aptose Biosciences Inc. Renal cell carcinoma Phase 1/2; ribonucleotide reductase
NCT00048113; 2002; United States of America Isis Pharmaceuticals Crohn's disease Phase 3; intercellular adhesion molecule
NCT00048295; 2002; United States of America Isis Pharmaceuticals Crohn's disease Phase 3; intercellular adhesion molecule
NCT01780545; 2013; United States of America OncoGenex Technologies Metastatic bladder cancer Phase 2; Heat shock protein 27
NCT01598948; 2012; Germany Ludwig‐Maximilians ‐ University of Munich Atherosclerosis Phase 3; apolipoprotein B
NCT01083615; 2010; United States of America OncoGenex Technologies Prostate cancer Phase 3; Clusterin
NCT02549651; 2015; United States of America MedImmune LLC Diffuse large B‐cell lymphoma Phase 1; STAT 3
NCT02243124; 2014; United States of America Eleos, Inc. Myelodysplastic syndrome Phase 1; P 53
NCT02144051; 2014; United States of America AstraZeneca Advanced solid tumours with androgen receptor pathway as a potential factor Phase 1; Androgen receptor
NCT00363974; 2006; United States of America Aegera Therapeutics Acute myelomonocytic leukaemia Phase 1; X‐linked inhibitor of apoptosis protein
NCT00054548; 2003; United States of America National Cancer Institute Advanced solid tumours Phase 1; Bcl‐2
NCT00557596; 2007; United States of America Aegera Therapeutics Advance pancreatic cancer Phase 1/2; X‐linked inhibitor of apoptosis protein
NCT00558922; 2007; United States of America Aegera Therapeutics Non‐small cell lung cancer Phase 1/2; X‐linked inhibitor of apoptosis protein
NCT00080847; 2004; United States of America National Cancer Institute Advanced diffuse large B‐cell non‐Hodgkin's lymphoma Phase 2; Bcl‐2
NCT00385775; 2006; United States of America Aegera Therapeutics Advanced tumours Phase 1; X‐linked inhibitor of apoptosis protein
NCT00967512; 2009; United States of America Eleos, Inc. Acute myelogenous leukaemia Phase 2; P 53
NCT00558545; 2007; United States of America Aegera Therapeutics Advanced breast cancer Phase 1/2; X‐linked inhibitor of apoptosis protein

Some of the registered studies did not have any mention of the phase of clinical trials and so the details are not captured in this table.

Despite a large number of molecules being evaluated in the clinical trials, the clinical progress of ASO has been slow because of the challenges involved in intracellular delivery of the large, highly charged molecules within acceptable limits of toxicity. Progress is also limited by the need to synthesize the TNA at an affordable cost. Various strategies to deliver the nucleic acids successfully are being evaluated. They include the cationic polymer, poly(ethylene imine); a biodegradable polymer, poly(2‐dimethylamino ethylamino) phosphazene; and dendritic α,ɛ‐poly(l‐lysine)s 43, 44. Direct delivery of ASO to lung to avoid systemic exposure has also been promising in preclinical animal studies and is likely to be tested in humans 45. Conjugated ASO is another arena for technological development. For instance, α‐tocopherol‐conjugated chimeric ASO was shown to have more tropism to hepatocytes and high efficacy in silencing the target gene in vivo 46.

Gene therapy

Gene therapy replaces the function of an abnormal or non‐functional gene with a functioning variant. Although the concept was proposed three decades ago, advances in the methods of delivery were made only over the past decade. Treatment now covers genetic disorders such as adenosine deaminase deficiency, α‐1‐antitrypsin deficiency, cystic fibrosis, familial hypercholesterolemia, Gaucher's disease and haemophilia B 47. The desired gene is delivered with the aid of a vector, which is usually a virus – most commonly a retrovirus or rarely adenovirus – as a therapeutic gene expression cassette. A therapeutic gene expression cassette is typically composed of a promoter that drives gene transcription, the transgene of interest, and a termination signal to end gene transcription 48. Also, there exists two approaches for gene delivery, namely germ line and somatic gene therapy. The former involves the introduction of the desirable gene into germ cells which are transmissible to the next generations while the latter involves the transfer of the desired gene to the somatic cells that are not heriTable 49, 50. Although several case reports for gene therapy were published in the 1970s, a major setback was seen in the year 1999 when an 18‐year‐old patient with ornithine transcarboxylase deficiency died during a clinical trial of gene therapy 51. A review of clinical trials with gene therapy from 1989 to 2015 revealed a total of 2210 clinical trials with the maximum number of trials (n = 130) conducted in the year 2014 and only 4% entering phase 3 clinical trial 52. Also the authors found that 64% of the trials are being carried out in the field of oncology. Despite so many ongoing clinical trials, only two gene therapies are approved for human use: gendicine and alipogene tiparvovec.

Gendicine

Gendicine is a recombinant adenovirus engineered to express the tumour suppressor gene p53, for patients with head and neck squamous cell carcinoma and was approved by the Chinese Food and Drug Administration in 2003 53. When combined with chemotherapy and radiotherapy, gendicine can improve treatment efficacy by 3.4‐fold (complete regression of tumour in 64% when gendicine is combined with radiotherapy as against 19% with radiotherapy alone) in addition to amelioration of the associated toxicity 54. Furthermore, gendicine has been shown to improve the therapeutic effect of radiation in pancreatic cancer cells 55. However, clinical trials in pancreatic cancer are yet to follow.

Alipgene

Alipogene tiparvovec, an adeno‐associated virus with lipoprotein lipase gene, is a gene therapy approved by the European Union for patients with lipoprotein lipase deficiency in 2012 56. As the condition is very rare (one in a million individuals), the decision was based on the results obtained from a study of just 27 patients.

The major concern related to gene therapy is the uncertain long‐term risk. The Office of Cellular, Tissue, and Gene Therapies (OCTGT) in the Centre for Biologics Evaluation and Research regulates the clinical trials that are evaluating gene therapy. The long‐term risk of gene therapy is uncertain but the US FDA has drafted a framework that is based on the integration of the vector with the target human genome and the potential for latency and reactivation. The US FDA recommends the sponsors to have a 15‐year follow‐up for study subjects with gene therapy, with a minimum of five years of annual examination and ten years of annual queries either in person or by questionnaire 57. A major challenge resides in the successful delivery of target genes. Modified human immunodeficiency virus, lentivirus, adenovirus, adeno‐associated virus and herpes simplex virus are the most widely used viral vectors. Non‐viral vectors for gene therapy are promising especially for the non‐invasive administration through skin, eyes and lungs 58. Novel layer‐by‐layer based nanoparticles 59 and liposomes 60 have been engineered that might play a useful role in successful delivery of genes into the human cells. Also, cell‐penetrating peptides have been shown in mouse models to deliver hepatitis B DNA that has potential applications in the management of chronic hepatitis B infections 61.

RNA Therapeutics

RNA interference [RNAi] and short interfering RNAs [siRNAs]

The understanding of the role of RNA in cell biology underwent a sea change in the early 1980s, when RNA moved from being perceived as a passive player in the transfer of information between DNA and protein synthesis to playing significant structural, enzymatic and information‐decoding roles during protein synthesis. Walter Gilbert first proposed the concept of ‘The RNA World’ in 1986 62, 63 and the pathway of RNA interference [RNAi] was discovered by Fire et al. and colleagues in the nematode Caenorhabditis elegans in 1998 64. The natural function of RNAi appears to be protection of the genome against invasion by mobile genetic material such as from viruses that produce aberrant RNA or dsRNA when they become active 65 and thus RNA interference serves as a powerful defence mechanism. Briefly, RNAi is a process by which RNA molecules with sequences complementary to a gene's coding sequence induce degradation of the corresponding messenger RNAs [mRNAs] thus blocking the translation of mRNA into protein. Computational models that can quantify the important parameters of the RNAi process in living mammalian cells have recently been developed 66. Therapy with siRNA thus has great potential application for diseases caused by abnormal expression or mutation such as cancers, viral infections and genetic disorders 67 as RNA interference can be experimentally triggered. The fact that the process of interference occurs in the cytoplasm and does not require nuclear penetration makes it even more attractive. In 2001, Elbashir and colleagues used synthetic siRNA to knock down a gene in several mammalian cell lines 65. Overexpression of Fas protein has been associated with death of hepatocytes and is implicated in a broad variety of liver disorders 68. Song et al. 69 injected Fas siRNAs that protected mice from autoimmune hepatitis which marked the more rapid development of RNA therapeutics.

siRNAs are ‘short’ double‐stranded molecules in that they are only 21–23 nucleotides long and generally can be chemically synthesized. siRNAs have the advantage over DNA oligonucleotides in that they are always delivered as duplexes, which are more stabile 70. Two major issues with siRNAs relate to their off‐target effects and delivery into the cell. Off‐target effects occur by suppression of genes other than the target gene, which can have deleterious consequences. This results primarily from partial sequence homology that exists with other mRNAs that are not the target mRNAs. siRNA is also recognized by immunoreceptors, like the Toll‐like receptors, that can lead to cytokine release and alteration in gene expression.

The first report of the application of the RNAi technology was to demonstrate the safety and efficacy of an siRNA ALN‐RSV01 in 101 healthy participants with experimentally induced respiratory syncytial virus (RSV) infection 71. ALN‐RSV01 targets the N protein of the RSV and was subsequently evaluated in a cohort of. Encouraging results were observed with the ALN‐RSV01 in 16 lung transplant recipients 72. The latter study also raised the possibility that siRNA can silence genes involved in allograft rejection 73. According to the database of clinical trials [clinicaltrials.gov], a total of 40 siRNAs are in various stages of development and one or more of them could receive approval in the next few years (Table 2).

Table 2.

Registered clinical studies with siRNA

Study id; year of registration; country of conduct of study Sponsors Disease condition Phase of clinical trial; target mRNA for ASO
NCT00716014; 2008; United States of America Pachyonychia congenita project Pachyonychia congenital (PC) Phase 1; PC keratins ‐ K6a
NCT00363714; 2006; United States of America Allergan Age‐related macular degeneration Phase 1/2; Vascular endothelial growth factor
NCT00672542; 2008; United States of America Scott Pruitt Metastatic melanoma Phase 1; immunoproteasome beta subunits LMP2, LMP7, and MECL1
NCT02166255; 2014; United States of America Comprehensive Cancer Center of Wake Forest University Melanoma, renal or pancreatic cancer Phase 1; E3 ubiquitin ligase Cbl‐b
NCT00395057; 2006; United States of America Allergan Age‐related macular degeneration Phase 2; Vascular endothelial growth factor
NCT00938574; 2009; Germany Silence Therapeutics GmbH Advanced solid tumours Phase 1; Protein kinase 3
NCT00689065; 2008; United States of America Calando Pharmaceuticals Solid tumours Phase 1; ribonucleotide reductase
NCT01188785; 2010; United States of America Silenseed Ltd Pancreatic adenocarcinoma Phase 1; KRAS (G12D) oncogene
NCT01591356; 2012; United States of America M.D. Anderson Cancer Center Advanced cancers Phase 1; EphA2
NCT01064505; 2010; United States of America Quark Pharmaceuticals Optic atrophy Phase 1; pro‐apoptotic protein, Caspase 2
NCT00927459; 2009; United States of America Arbutus Biopharma Corporation Hypercholesterolemia Phase 1; polo‐like kinase I, kinesin spindle protein
NCT01437007; 2011; United States of America National Cancer Institute Hepatocellular carcinoma Phase 1; polo‐like kinase 1
NCT01858935; 2013; United States of America Nitto Denko Corporation Healthy individuals Phase 1; Heat shock protein 47
NCT02314052; 2014; United States of America Dicerna Pharmaceuticals, Inc. Hepatocellular carcinoma Phase 1/2; MYC oncoprotein
NCT02227459; 2014; United States of America Nitto Denko Corporation Hepatic fibrosis Phase 1; Heat shock proteins
NCT02110563; 2014; United States of America Dicerna Pharmaceuticals, Inc. Multiple Myeloma Non‐Hodgkins Lymphoma Phase 1; MYC oncoprotein
NCT00306904; 2006; United States of America OPKO Health, Inc. Diabetic macular oedema Phase 2; Vascular endothelial growth factor
NCT02250612; 2014; United States of America Sylentis, S.A. Open angle glaucoma Phase 1; Beta‐2 adrenergic receptor
NCT01776658; 2012; United States of America Sylentis, S.A. Dry eye syndrome Phase 1/2; Vascular endothelial growth factor
NCT01739244; 2012; United States of America Sylentis, S.A. Open angle glaucoma Phase 2; Vascular endothelial growth factor
NCT01676259; 2012; Israel Silenseed Ltd Advanced pancreatic cancer Phase 2; KRAS (G12D) oncogene
NCT01438281; 2011; Spain Sylentis, S.A. Dry eye syndrome Phase 1; transient receptor potential cation channel subfamily V member 1 (TRPV1)
NCT00557791; 2007; United States of America OPKO Health, Inc. Age related macular degeneration Phase 3; Vascular endothelial growth factor
NCT00802347; 2008; Canada Quark Pharmaceuticals Renal transplantation Phase 1/2; P 53
NCT01808638; 2013; Germany Silence Therapeutics GmbH Pancreatic ductal carcinoma Phase 1/2; Protein kinase 3
NCT02596347; 2015; United States of America National Jewish Health Chronic beryllium disease JAM2
NCT00154934; 2005; Taiwan National Taiwan University Hospital Pre‐eclampsia IL‐10
NCT01075360; 2010; Taiwan National Taiwan University Hospital Neuroblastoma E2F1
NCT01834105; 2013; China Fujian Institute Of Traditional Chinese Medicine Post menopausal osteoporosis Phase 2; cardiotrophin‐like cytokine factor 1
NCT01227291; 2010; Spain Sylentis, S.A. Glaucoma Phase 1/2; Beta‐2 adrenergic receptor
NCT01382511; 2011; Israel Tel‐Aviv Sourasky Medical Center Pachyonychia congenita STAT 1
NCT02528682; 2015; Netherlands Radboud University Hematological malignancies Phase 1/2; PD‐1/PD‐L1 co‐inhibitory pathway
NCT02055846; 2012; France Institut Paoli‐Calmettes Prostate cancer Heat shock protein 27
NCT00554359; 2007; United States of America Quark Pharmaceuticals Acute renal failure Phase 1; P53
NCT01058798; 2010; Taiwan National Taiwan University Hospital Neuroblastoma β1,4‐N‐acetylgalactosaminyltransferase III
NCT01445899; 2011; Israel Quark Pharmaceuticals Diabetic macular oedema Phase 2; hypoxia‐inductible gene
NCT00259753; 2005; United States of America OPKO Health, Inc. Age related macular degeneration Phase 2; Vascular endothelial growth factor

Some of the registered studies did not have any mention of the phase of clinical trials and so the details are not captured in this table

MicroRNAs – their targeting and the use of antagonists and mimics of microRNA as therapeutic agents

Components of the genome that had hitherto been considered non‐functional were discovered to have gene regulatory capacity approximately 20 years ago. MicroRNAs (miRNAs) were first discovered in 1993 74 and have today made the transition from worm genetics to human disease. Victor Ambros and Gary Ruvkun discovered the first miRNA gene in C. elegans when a small RNA encoded by the lin‐4 locus was found to be associated with the developmental timing of the nematode. Since their discovery, miRNAs have been shown to be an essential part of the non‐coding genome. miRNAs are short single‐stranded non‐coding RNA molecules of 15–22 nucloetides, which function as key negative regulators of post‐transcriptional modulation in almost all biological processes. Today, miRNA dysregulation is frequently shown to be associated with pathogenesis of human disease thus making them attractive therapeutic targets 75, 76. Their advantage as therapeutic agents lies in their ability to target multiple molecules unlike the traditional approach of targeting a single gene thus making them extremely efficient in regulating distinct biological cell processes relevant to both normal and cancer cell biology 77.

Early evidence of the involvement of miRNAs in human cancer came from studies on chronic lymphocytic leukaemia (CLL). The human miRNA genes mir‐15a and mir‐16‐1 were shown to be deleted or downregulated in over two‐thirds of the cases of CLL 78. The first oncogenic miRNAs described were the miR‐17‐92 cluster, which is amplified in B cell lymphomas among other cancers, and miR‐155, which is overexpressed in haematological cancers 79, 80. Subsequently, miRNAs have been identified for several cancers, including lung, ovarian and breast cancers, and glioblastomas 81. In addition, there are many diseases where it has been shown that there is significantly greater expression of miRNA in diseased tissue than in normal tissue. These include diabetes, obesity, Alzheimer's and Parkinson's disease and cardiovascular and autoimmune disorder 82. MicroRNA therapeutics can be broadly classified into two types – the use of miRNA antagonists and miRNA mimics, with the latter also being called ‘miRNA replacement therapy’. In cancer, they can be targeted in two ways. Direct approaches involve the use of oligonucleotides or virus‐based constructs to block their expression or introduce a tumour suppressor miRNA that has been lost due to the disease process. Indirect approaches involve use of drug therapy that modulates miRNA expression by targeting their transcription and their processing 83.

One miRNA‐based agent that is in advanced clinical development is actually for use in an infectious disease and not cancer. miRNA‐122 is expressed in hepatocytes and interacts with hepatitis C virus (HCV) leading to proliferation of HCV 84. This seminal discovery paved the way for the discovery of the first anti‐viral agent that targets a miRNA, called miraversen (formerly SPC3649) 85.

Miravirsen

Miravirsen is a β‐d‐oxy‐locked nucleic acid‐modified phosphorothioate antisense oligonucleotide that targets the liver‐specific microRNA‐122 (miR‐122) and is one of a class of miRNA inhibitors or antagonists that has undergone clinical trials in patients with hepatitis C. Studies with the drug have been conducted in healthy participants (n = 82) and patients (n = 37) with chronic HCV infection in studies lasting 12 weeks. No dose‐limiting toxicities or participant discontinuations were seen 86. Miravirsen has also demonstrated broad spectrum antiviral activity and a relatively high genetic barrier to resistance which is slow to emerge 76. The drug should prove to be a useful therapeutic option and also as an addition alongside agents such as pegylated interferon, telepravir or bocepravir and ribavarin. The clinical trials registry clinicaltrials.gov lists seven studies with miravirsen of which two have been completed and the remaining five are active. The latter studies include long‐term safety, pharmacokinetics and evaluation of drug interactions. Another miRNA anatagonist RG‐101 has entered Phase II trials for the same indication. An miRNA mimic that targets miRNA‐34 is likely to be the first candidate mimic to reach the clinic for the treatment of hepatocellular carcinoma 87. Other potential roles of miRNA include targeting malignant plasma cells in multiple myeloma and modifying insulin release and resistance in diabetes mellitus 88, 89.

RNA aptamers and RNA decoys

A facet of RNAs that makes them promising as therapeutic agents is the ability of some small RNAs to fold into three‐dimensional structures which can then bind to proteins and thereby inhibit them in a manner similar to protein antagonists. This is the logic behind the use of RNA ‘decoys’ or RNA aptamers. They can bind viral proteins and thus can also be used as vehicles to transport siRNAs. RNA aptamers are developed by methods similar to those described above for DNA aptamers. RNA aptamers are highly susceptible to exonuclease degradation and so require capping with modified or inverted nucleotides such as 2′‐O‐modified pyrimidines, 2′‐amino pyrimidines or 2′‐fluoro pyrimidines to prevent terminal degradation 90. The advantage of RNA aptamers lies in both their ability to reach intracellular targets and also to bind directly to extracellular targets, unlike other RNA‐based therapeutics that must first enter the cell to carry out their functions 91.

An early clinical trial was conducted in patients with HIV using the RNA decoy strategy 92 against the HIV Tat protein. The approach was shown to be safe but disappointing in four children with HIV infection. It is likely that the RNA decoy was insufficiently expressed for effective anti‐viral activity. Just a few years later, in December 2004, pegaptanib (Macugen, Eyetech Pharmaceutics/Pfizer), an aptamer against vascular endothelial growth factor (VEGF), become the first RNA aptamer to receive approval from the US FDA for use in age‐related macular degeneration (AMD) 93. NOX‐A12 (olaptesed pegol) is a 45‐nucleotide‐long RNA aptamer that specifically antagonizes the CXC chemokine ligand 12/stromal cell derived factor‐1 (CXCL 12/SDF‐1) which is a key regulatory chemokine for the migration of leukaemic stem cells into the bone marrow 94. This inhibition of binding of SDF‐1 to its receptors can lead the leukaemic stem cells to re‐enter the cell cycle and become vulnerable to chemotherapeutic attack. A Phase 1 study with NOX‐A12 proved its safety in 48 normal, healthy participants. A Phase II study with the aptamer in combination with bendamustine and rituximab (NCT01486797) in relapsed chronic lymphocytic leukaemia is ongoing. Another aptamer NOX‐E36 (ematicap pegol) has undergone studies in both normal, healthy participants and patients with Type‐2 diabetes. NOX‐E36 interacts with the chemokine ligand 2 (CCL2), a protein involved in the recruitment of monocytes and T cells to sites of injury and inflammation. CCL2 has recently been identified as an ‘adipokine’ that plays an important role in obesity and complications of Type 2 diabetes 95. Both the NOX aptamers belong to a special class of aptamers called spiegelmers (from the German Spiegel, a mirror) as they are composed of l‐ribose units, not the naturally occurring d‐enantiomers, and are highly resistant to degradation by nucleases 96. Another distinguishing feature of spiegelmers is that they are not synthesized by SELEX technology but by mirrored target molecules 97. Olaptesed pegol is tried in clinical trials as an add‐on drug with other chemotherapeutic agents, while ematicap pegol is used alone for patients with diabetic nephropathy. REG‐1 (RB006/RB007, pegnivacogin/anivamirsen) is an RNA aptamer along with a single‐stranded RNA nucleotide that is given subcutaneously and which targets the coagulation factor IXa. A Phase 1 study in 36 healthy participants (NCT01872572) has been completed without serious adverse effects emerging. It has also been studied in patients with stable cardiovascular disease taking dual antiplatelet therapy with aspirin and clopidogrel 98, 99 and in patients with acute coronary syndromes undergoing percutaneous coronary intervention with no catheter or guide wire thrombosis.

Ribozymes

A subset of RNAs called catalytic RNAs or ribozymes can act as enzymes in the complete absence of protein 100. Because of their high specificity, wide range of target selection, and action before protein translation, both naturally occurring and artificially synthesized ribozymes can be used to specifically suppress gene function. They can also be used to validate disease‐related genes as potential targets for new therapeutic interventions. Their ability to cleave mRNA to prevent protein synthesis enables them to find applications in cancer and virology. Of the several different types of ribozymes, the hammerhead and hairpin ribozyme have been studied extensively. The former is a small ribozyme that has both specificity and catalytic efficacy and thus likely the most widely studied 101.

The first synthesized ribozyme to be evaluated in clinical trials was ANGIOZYMETM. It was designed to combat angiogenesis by cleaving the mRNA that produces VEGF. The anti‐Flt‐1 ribozyme was designed specifically to cleave the mRNAs for the primary VEGF receptors FLt‐ and KDR 102. This ribozyme was well tolerated after intravenous infusion or subcutaneous bolus administration to healthy individuals. A significant increase in serum von Willebrand factor antigen, a marker for endothelial cell dysfunction, was also seen after use of angiozyme 102. A nuclease‐resistant hammerhead ribozyme, Heptazyme (LY466700), completed a Phase I/II clinical trial in 28 patients with chronic HCV and was reported safe and well tolerated. Similarly, in Phase II studies, it was found safe and well tolerated. Clinical evaluation of this agent was eventually discontinued due to safety concerns in primates 103.

Circular RNAs

Normally, RNA is linear with both 3′ and 5′ ends open. But circular RNAs (circRNA) have a covalent bond between the ends so that they are closed. This confers on them resistance to degradation by exonucleases so they tend to be more stable; as a result there may be ten times as much circRNA as linear RNA in the cytoplasm 104. Another property of circRNA distinguishing it from linear RNA is that it is mostly non‐coding 105. The transcripts of linear mRNA typically are capped and polyadenylated and so can be detected by reverse transcriptase‐polymerase chain reaction, unlike circRNAs 106. CircRNAs are detected by either scrambled exon enrichment in an RNAse‐treated sample or by ‘TRAP electrophoresis’ or 2D electrophoresis 107, 108. Recently, circRNAs have been found to bind to RNA‐binding proteins or ribonucleprotein complexes acting as ‘sponges’, as so have also been called competing endogenous RNAs 109. Although no therapeutic molecule related to circRNA has been produced or approved by any regulatory agency, it has been implicated to play a crucial role in some human diseases. For example, cANRIL, a circRNA for a long non‐coding RNA, has been found to be a risk factor for atherosclerosis 110. CiRS‐7 is a circRNA that can act as a miRNA sponge to adsorb and quench normal miRNA‐7 which has been implicated in the pathophysiology of Parkinson's disease, Alzheimer's disease and cancers including colorectal cancer and pancreatic ductal adenocarcinoma 111. Additionally, reports indicate that circRNAs can also act as potential biomarkers for aging and gastric cancer 112. Studies are underway to explore the varied physiological and pathological roles of circRNAs and their therapeutic potential.

Conclusions

Though nucleic acids have been recognized as potential therapeutic agents for more than 40 years, it was the decoding of the human genome coupled with technological advances that has led to the realization of their therapeutic applications. Numerous studies, both in the laboratory and clinic, in the last two decades have demonstrated the enormous potential of nucleic acids as therapeutic agents, particularly with their promise of specificity, functional diversity and limited toxicity. Of the various nucleic acid therapies, gene therapy has been widely investigated clinically followed by ASOs but only few agents have been approved by the regulatory agencies so far. Among the several hundred studies that have been carried out, there have been two deaths 51 and four instances of malignancy. Overall, however, the agents appear acceptably safe. The spectrum of diseases that can potentially be addressed by nucleic acids is broad and ranges from infectious diseases on one end to those like diabetes and cancer at the other. Key challenges that need to be addressed for the successful translation of nucleic acids will be their delivery to the site of action, choice between direct delivery and the use of a vehicle, mass production at low cost, more clearly defined pharmacokinetics, ability to produce sustained long‐term effects, immunogenicity and toxicity (including inappropriate or excessive expression). Studies addressing these areas are needed to improve success in clinical trials. There is no doubt that an improved understanding of the biology of the various nucleic acid pathways will lead to novel therapies in future.

Competing Interest

All authors have completed the Unified Competing Interest form at www.icmje.org/coi_disclosure.pdf (available on request from the corresponding author) and declare: no support from any organization for the submitted work; no financial relationships with any organizations that might have an interest in the submitted work in the previous 3 years; no other relationships or activities that could appear to have influenced the submitted work.

Sridharan, K. , and Gogtay, N. J. (2016) Therapeutic nucleic acids: current clinical status. Br J Clin Pharmacol, 82: 659–672. doi: 10.1111/bcp.12987.

References

  • 1. Breaker RR, Joyce GF. The expanding view of RNA and DNA function. Chem Biol 2014; 21: 1059–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Khakshoor O, Kool ET. Chemistry of nucleic acids: impacts in multiple fields. Chem Commun 2011; 47: 7018–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Opalinska JB, Gewirtz AM. Nucleic‐acid therapeutics: basic principles and recent applications. Nat Rev Drug Discov 2002; 1: 503–14. [DOI] [PubMed] [Google Scholar]
  • 4. Muralidhara BK, Baid R, Bishop SM, Huang M, Wang W, Nema S. Critical considerations for developing nucleic acid macromolecule based drug products. Drug Discov Today 2016; 21: 430–44. [DOI] [PubMed] [Google Scholar]
  • 5. Ho D, Wang C‐H K, Chow EK‐H. Nanodiamonds: the intersection of nanotechnology, drug development and personalized medicine. Sci Adv 2015; 1: e1500439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Wraight CJ, White PJ. Antisense oligonucleotides in cutaneous therapy. Pharmacol Ther 2001; 90: 89–104. [DOI] [PubMed] [Google Scholar]
  • 7. Cavagnari BM. Gene therapy: nucleic acids as drugs. Action, mechanisms and delivery into the cell. Arch Argent Pediatr 2011; 109: 237–44. [DOI] [PubMed] [Google Scholar]
  • 8. Martinez T, Jimenez AI, Paneda C. Short‐interference RNAs: becoming medicines. EXCLI J 2015; 14: 714–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Lucas‐Samuel S, Ferry N, Trouvin JH. Overview of the regulatory oversight implemented by the French regulatory authorities for the clinical investigation of gene therapy and cell therapy products. Adv Exp Med Biol 2015; 871: 73–85. [DOI] [PubMed] [Google Scholar]
  • 10. Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov 2012; 11: 125–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Andronescu M, Zhang ZC, Condon A. Secondary structure prediction of interacting RNA molecules. J Mol Biol 2005; 345: 987–1001. [DOI] [PubMed] [Google Scholar]
  • 12. Sun H, Zhu X, Lu PY, Rosato RR, Tan W, Zu Y. Oligonucleotide aptamers: tools for targeted cancer therapy. Mol Ther Nucleic Acids 2014; 3: e182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ni X, Castanares M, Mukherjee A, Lupold SE. Nucleic acid aptamers: clinical applications and promising new horizons. Curr Med Chem 2011; 18: 4206–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Blind M, Blank M. Aptamer selection technology and recent advances. Mol Ther Nucleic Acids 2015; 4: e223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Berezovski MV, Lechmann M, Musheev MU, Mak TW, Krylov SN. Aptamer‐facilitated biomarker discovery (AptaBiD). J Am Chem Soc 2008; 130: 9137–43. [DOI] [PubMed] [Google Scholar]
  • 16. Syed M, Pervaiz S. Advances in sptamers. Oligonucleotides 2010; 20: 215–24. [DOI] [PubMed] [Google Scholar]
  • 17. Santosh B, Yadava PK. Nucleic acid aptamers: research tools in disease diagnostics and therapeutics. BioMed Res Int 2014; 2014: 540451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Shum KT, Zhou J, Rossi JJ. Nucleic acid aptamers as potential therapeutic and diagnostic agents for lymphoma. J Cancer Ther 2013; 4: 872–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Que‐Gewirth NS, Sullenger BA. Gene therapy progress and prospects: RNA aptamers. Gene Ther 2007; 14: 283–91. [DOI] [PubMed] [Google Scholar]
  • 20. Chan JHP, Lim S, Wong WSF. Antisense oligonucleotides: from design to therapeutic application. Clin Exp Pharmacol Physiol 2006; 33: 533–40. [DOI] [PubMed] [Google Scholar]
  • 21. Stanton R, Sciabola S, Salatto C, Weng Y, Moshinsky D, Little J, et al. Chemical modification study of antisense gapmers. Nucleic Acid Ther 2012; 22: 344–59. [DOI] [PubMed] [Google Scholar]
  • 22. Wan WB, Migawa MT, Vasquez G, Murray HM, Nichols JG, Gaus H, et al. Synthesis, biophysical properties and biological activity of second generation antisense oligonucleotides containing chiral phosphorothioate linkages. Nucleic Acids Res 2014; 42: 13456–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bennett CF, Swayze EE. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu Rev Pharmacol Toxicol 2010; 50: 259–93. [DOI] [PubMed] [Google Scholar]
  • 24. Geary RS. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin Drug Metab Toxicol 2009; 5: 381–91. [DOI] [PubMed] [Google Scholar]
  • 25. Evers MM, Toonen LJA, van Roon‐Mom WMC. Antisense oligonucleotides in therapy for neurodegenerative disorders. Adv Drug Deliv Rev 2015; 87: 90–103. [DOI] [PubMed] [Google Scholar]
  • 26. Yu RZ, Grundy JS, Geary RS. Clinical pharmacokinetics of second generation antisense oligonucleotides. Expert Opin Drug Metab Toxicol 2013; 9: 169–82. [DOI] [PubMed] [Google Scholar]
  • 27. Mansoor M, Melendez AJ. Advances in antisense oligonucleotide development for target identification, validation, and as novel therapeutics. Gene Regul Syst Bio 2008; 2: 275–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Tse MT. Antisense approval provides boost to the field. Drug Discov 2013; 12: 179. [Google Scholar]
  • 29. Grillone LR, Lanz R. Fomivirsen. Drugs Today (Barc) 2001; 37: 245–55. [DOI] [PubMed] [Google Scholar]
  • 30. Wong E, Goldberg T. Mipomersen (Kynamro). A novel antisense oligonucleotide inhibitor for the management of homozygous familial hypercholesterolemia. Pharm Therapeut 2014; 39: 119–22. [PMC free article] [PubMed] [Google Scholar]
  • 31. Orr RM. Technology evaluation: fomivirsen, Isis Pharmaceuticals Inc./CIBA vision. Curr Opin Mol Ther 2001; 3: 288–94. [PubMed] [Google Scholar]
  • 32. Henry SP, Miner RC, Drew WL, Fitchett J, York‐Defalco C, Rapp LM, et al. Antiviral activity and ocular kinetics of antisense oligonucleotides designed to inhibit CMV replication. Invest Ophthalmol Vis Sci 2001; 42: 2646–51. [PubMed] [Google Scholar]
  • 33. Geary RS, Henry SP, Grillone LR. Fomivirsen: clinical pharmacology and potential drug interactions. Clin Pharmacokinet 2002; 41: 255–60. [DOI] [PubMed] [Google Scholar]
  • 34. The Vitravene study group . Randomized dose‐comparison studies of intravitreous fomivirsen for treatment of cytomegalovirus retinitis that has reactivated or is persistently active despite other therapies in patients with AIDS. Am J Ophthalmol 2002; 133: 475–83. [DOI] [PubMed] [Google Scholar]
  • 35. Vitravene Study Group 2 . Safety of intravitreous fomivirsen for treatment of cytomegalovirus retinitis in patients with AIDS. Am J Ophthalmol 2002; 133: 484–98. [DOI] [PubMed] [Google Scholar]
  • 36. Stone TW, Jaffe GJ. Reversible bull's‐eye maculopathy associated with intravitreal fomivirsen therapy for cytomegalovirus retinitis. Am J Ophthalmol 2000; 130: 242–3. [DOI] [PubMed] [Google Scholar]
  • 37. Kastelein JJ, Wedel MK, Baker BF, Su J, Bradley JD, Yu RZ, et al. Potent reduction of apolipoprotein B and low‐density lipoprotein cholesterol by short‐term administration of an antisense inhibitor of apolipoprotein B. Circulation 2006; 114: 1729–35. [DOI] [PubMed] [Google Scholar]
  • 38. Goldberg AC, Hopkins PN, Toth PP, Ballantyne CM, Rader DJ, Robinson JG, et al. Familial hypercholesterolemia: screening, diagnosis and management of pediatric and adult patients: clinical guidance from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol 2011; 5: 133–40. [DOI] [PubMed] [Google Scholar]
  • 39. Raal FJ, Santos RD, Blom DJ, Marais AD, Charng MJ, Cromwell WC, et al. Mipomersen, an apolipoprotein B synthesis inhibitor, for lowering of LDL cholesterol concentrations in patients with homozygous familial hypercholesterolemia: a randomised, double‐blind, placebo‐controlled trial. Lancet 2010; 375: 998–1006. [DOI] [PubMed] [Google Scholar]
  • 40. McGowan MP, Tardif JC, Ceska R, Burgess LJ, Soran H, Gouni‐Berthold I, et al. Randomized, placebo‐controlled trial of mipomersen in patients with severe hypercholesterolemia receiving maximally tolerated lipid‐lowering therapy. PLoS One 2012; 7: 133–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Stein EA, Dufour R, Gagne C, Gaudet D, East C, Donovan JM, et al. Apolipoprotein B synthesis inhibition with mipomersen in heterozygous familial hypercholesterolemia: results of a randomized, double‐blind, placebo‐controlled trial to assess efficacy and safety as add‐on therapy in patients with coronary artery disease. Circulation 2012; 126: 2283–92. [DOI] [PubMed] [Google Scholar]
  • 42. Büller HR, Bethune C, Bhanot S, Gailani D, Monia BP, Raskob GE, et al. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N Engl J Med 2015; 372: 232–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Thomas M, Ge Q, Lu JJ, Chen J, Klibanov AM. Cross‐linked small polyethylenimines: while still nontoxic, deliver DNA efficiently to mammalian cells in vitro and in vivo . Pharm Res 2005; 22: 373–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Mastrobattista E, Hennink WE, Schiffelers RM. Delivery of nucleic acids. Pharm Res 2007; 24: 1561–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Kjems J, Howard KA. Oligonucleotide delivery to the lung: waiting to inhale. Mol Ther Nucleic Acids 2012; 1: e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Nishina T, Numata J, Nishina K, Yoshida‐Tanaka K, Nitta K, Piao W, et al. Chimeric antisense oligonucleotide conjugated to alpha‐tocopherol. Mol Ther Nucleic Acids 2015; 4: e220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Misra S. Human gene therapy: a brief overview of the genetic revolution. JAPI 2013; 61: 127–33. [PubMed] [Google Scholar]
  • 48. Papadakis ED, Nicklin SA, Baker AH, White SJ. Promoters and control elements: designing expression cassettes for gene therapy. Curr Gene Ther 2004; 4: 89–113. [DOI] [PubMed] [Google Scholar]
  • 49. Nayerossadat N, Maedeh T, Ali PA. Viral and nonviral delivery systems for gene delivery. Adv Biomed Res 2012; 1: 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Mathews QL, Curiel DT. Gene therapy: human gemline genetics modifications – assessing the scientific, socioethical, and religious issues. South Med J 2007; 100: 98–100. [DOI] [PubMed] [Google Scholar]
  • 51. Sibbald B. Death but one unintended consequence of gene therapy trial. CMAJ 2001; 164: 1612. [PMC free article] [PubMed] [Google Scholar]
  • 52. Ginn SL, Alexander IE, Edelstein ML, Abedi MR, Wixon J. Gene therapy clinical trials worldwide to 2012 – an update. J Gene Med 2013; 15: 65–77. [DOI] [PubMed] [Google Scholar]
  • 53. Peng H. Current status of Gendicine in China: recombinant human Ad‐p53 agent for treatment of cancers. Hum Gene Ther 2005; 16: 1016–27. [DOI] [PubMed] [Google Scholar]
  • 54. Krimsky S. China's gene therapy drug. Genewatch 2005; 18: 10–3. [PubMed] [Google Scholar]
  • 55. Li J, Pan J, Zhu X, Su Y, Bao L, Qiu S, et al. Recombinant adenovirus‐p53 (Gencidine) sensitizes a pancreatic carcinoma cell line to radiation. Chin J Can Res 2013; 25: 715–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yla‐Herttuala S. Endgame: Glybera finally recommended for approval as the first gene therapy drug in the European Union. Mol Ther 2012; 20: 1831–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. United States Food and Drug Administration . Guidance for industry. Gene therapy for clinical trials – observing subjects for delayed adverse events. Available at: http://www.fda.gov/downloads/BiologicsBloodVaccines/GuidanceComplianceRegulatoryInformation/Guidances/CellularandGeneTherapy/ucm078719.pdf (last accessed 22 December 2015).
  • 58. Foldvari M, Chen DW, Nafissi N, Calderon D, Narsineni L, Rafiee A. Non‐viral gene therapy: gains and challenges of non‐invasive administration methods. J Control Release 2015; p1130264–9. [DOI] [PubMed] [Google Scholar]
  • 59. Correa S, Dreaden EC, Gu L, Hammond PT. Engineering nanolayered particles for modular drug delivery. J Control Release 2016. doi:10.1016/j.jconrel.2016.01.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Kundu SK, Sharma AR, Lee S‐S, Sharma G, Doss CGP, Yagihara S, et al. Recent trends of polymer mediated liposomal gene delivery system. BioMed Res Int 2014; 2014: 934605. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 61. Ndeboko B, Lemamy GJ, Nielsen PE, Cova L. Therapeutic potential of cell penetrating peptides (CPPs) and cationic polymers for chronic hepatitis B. Int J Mol Sci 2015; 16: 28230–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Altman S. The RNA–protein world. RNA 2013; 19: 589–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Miele E, Spinelli GP, Miele E, Fabrizio ED, Ferretti E, Tomao S, et al. Nanoparticle‐based delivery of small interfering RNA: challenges for cancer therapy. Int J Nanomedicine 2012; 7: 3637–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double‐stranded RNA in Caenorhabditis elegans . Nature 1998; 391: 806–11. [DOI] [PubMed] [Google Scholar]
  • 65. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21‐nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001; 411: 494–8. [DOI] [PubMed] [Google Scholar]
  • 66. Dornseifer S, Willkomm S, Far RK, Liebschwager J, Beltsiou F, Frank K, et al. RNAi revised – target mRNA‐dependent enhancement of gene silencing. Nucleic Acids Res 2015; 43: 10623–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Lee JM, Yoon TJ, Cho YS. Recent developments in nanoparticle‐based siRNA delivery for cancer therapy. BioMed Res Int 2013; 2013: 782041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Roskams T, Libbrecht L, Van Damme B, Desmet V. Fas and Fas ligand: strong co‐expression in human hepatocytes surrounding hepatocellular carcinoma; can cancer induce suicide in peritumoural cells? J Pathol 2000; 191: 150–3. [DOI] [PubMed] [Google Scholar]
  • 69. Song E, Lee SK, Wang J, Ince N, Ouyang N, Min J, et al. RNA interference targeting Fas protects mice from fulminant hepatitis. Nat Med 2003; 9: 347–51. [DOI] [PubMed] [Google Scholar]
  • 70. Watts JK, Corey DR. Gene silencing by siRNAs and antisense oligonucleotides in the laboratory and the clinic. J Pathol 2012; 226: 365–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. DeVincenzo J, Cehelsky JE, Alvarez R, Elbashir S, Harborth J, Toudjarska I, et al. Evaluation of the safety, tolerability and pharmacokinetics of ALN‐RSV01, a novel RNAi antiviral therapeutic directed against respiratory syncytial virus (RSV). Antiviral Res 2008; 77: 225–31. [DOI] [PubMed] [Google Scholar]
  • 72. Zamora MR, Budev M, Rolfe M, Gottlieb J, Humar A, Devincenzo J, et al. RNA interference therapy lung transplant patients infected with respiratory syncytial virus. Am J Respir Crit Care Med 2011; 183: 531–8. [DOI] [PubMed] [Google Scholar]
  • 73. Astor TL. RNA interference, RSV and lung transplantation – a promising future for siRNA therapeutics. Am J Respir Crit Care Med 2011; 183: 427–8. [DOI] [PubMed] [Google Scholar]
  • 74. Almeida ML, Reis RM, Calin GA. MicroRNa history: discovery, recent applications, and next frontiers. Mutat Res Fund Mol Mech Mutagen 2011; 717: 1–8. [DOI] [PubMed] [Google Scholar]
  • 75. Mendell JT, Olson EN. MicroRNAs in stress signaling and human disease. Cell 2012; 148: 1172–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Ottosen S, Parsley TB, Yang L, Zeh K, van Doorn LJ, van der Veer E, et al. In vitro antiviral activity and preclinical and clinical resistance profile of Miravirsen, a novel anti‐hepatitis C virus therapeutic targeting the human factor miR‐122. Antimicrob Agents Chemother 2015; 59: 599–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Lorio MV, Croce CM. MicroRNA dysregulation in cancer: diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol Med 2012; 4: 143–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down‐regulation of micro‐ RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2002; 99: 15524–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. He L, Thomson JM, Hemann MT, Hernando‐Monge E, Mu D, Goodson S, et al. A microRNA polycistron as a potential human onco‐gene. Nature 2005; 435: 828–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Costinean S, Zanesi N, Pekarsky Y, Tili E, Volinia S, Heerema N, et al. Pre‐B cell proliferation and lymphoblastic leukemia/high‐grade lymphoma in E(mu)‐miR155 transgenic mice. Proc Natl Acad Sci U S A 2006; 103: 7024–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Jin D, Lee H. A computational approach to identifying gene‐microRNA modules in cancer. PLoS Comput Biol 2015; 11: e1004042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Bader AG. miR‐34 – a microRNA replacement therapy is headed to the clinic. Front Genet 2012; 3: 120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yu HW, Cho WC. The emerging role of miRNAs in combined cancer therapy. Expert Opin Biol Ther 2015; 15: 923–5. [DOI] [PubMed] [Google Scholar]
  • 84. Luna JM, Scheel TKH, Danino T, Shaw KS, Mele A, Fak JJ, et al. Hepatitis C virus RNA functionally sequesters miR‐122. Cell 2015; 160: 1099–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Jopling C, Schütz S, Sarnow P. Position‐dependent function for a tandem microRNA miR‐122‐binding site located in the hepatitis C virus RNA genome. Cell Host Microbe 2008; 4: 77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez‐Torres M, Patel K, et al. Treatment of HCV infection by targeting microRNA. N Engl J Med 2013; 368: 1685–94. [DOI] [PubMed] [Google Scholar]
  • 87. Van Rooij E, Kauppinen S. Development of microRNA therapeutics is coming of age. EMBO Mol Med 2014; 6: 851–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Rossi M, Amodio N, Di Martino MT, Tagliaferri P, Tassone P, Cho WC. MicroRNA and multiple myeloma: from laboratory findings to translational therapeutic approaches. Curr Pharm Biotechnol 2014; 15: 459–67. [DOI] [PubMed] [Google Scholar]
  • 89. Chen H, Lan HY, Roukos DH, Cho WC. Application of microRNAs in diabetes mellitus. J Endocrinol 2014; 222: R1–R10. [DOI] [PubMed] [Google Scholar]
  • 90. Lee JW, Kim HJ, Heo K. Therapeutic aptamers: developmental potential as anticancer drugs. BMB Rep 2015; 48: 234–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Germer K, Leonard M, Zhang X. RNA aptamers and their therapeutic and diagnostic applications. Int J Biochem Mol Biol 2013; 4: 27–40. [PMC free article] [PubMed] [Google Scholar]
  • 92. Kohn DB, Bauer G, Rice CR, Rothschild JC, Carbonaro DA, Valdez P, et al. A clinical trial of retroviral‐mediated transfer of a rev‐responsive element decoy gene into CD34(+) cells from the bone marrow of human immunodeficiency virus‐1‐infected children. Blood 1999; 94: 368–71. [PubMed] [Google Scholar]
  • 93. Ng EW, Adamis AP. Anti‐VEGF aptamer (pegaptanib) therapy for ocular vascular diseases. Ann N Y Acad Sci 2006; 1082: 151–71. [DOI] [PubMed] [Google Scholar]
  • 94. Duda DG, Kozin V, Kirkpatrick ND, Xu L, Fukumura D, Jain RK. CXCL 12 (SDF1 alpha)‐ CXCR4/CXCR7 pathway inhibition: an emerging sensitizer for anticancer therapies? Clin Cancer Res 2011; 8: 2074–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Rull A, Camps J, Alonso‐Villaverde C, Joven J. Insulin resistance, inflammation, and obesity: role of monocyte chemoattractant protein‐1 (or CCL2) in the regulation of metabolism. Mediators Inflamm 2010; 2010: 326580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Hoffmann S, Hoos J, Klussmann S, Vonhoff S. RNA aptamers and spiegelmers: synthesis, purification, and post‐synthetic PEG conjugation. Curr Protoc Nucleic Acid Chem 2011; 46: 1–30. [DOI] [PubMed] [Google Scholar]
  • 97. Vater A, Klussmann S. Turning mirror‐image oligonucleotides into drugs: the evolution of spiegelmer therapeutics. Drug Discov Today 2015; 1: 147–55. [DOI] [PubMed] [Google Scholar]
  • 98. Becker RC, Chan MY. REG‐1, a regimen comprising RB‐006, a Factor IXa antagonist, and its oligonucleotide active control agent RB‐007 for the potential treatment of arterial thrombosis. Curr Opin Mol Ther 2009; 11: 707–15. [PubMed] [Google Scholar]
  • 99. Povsic TJ, Cohen MG, Mehran R, Buller CE, Bode C, Cornel JH, et al. A randomized, partially blinded, multicenter, active‐controlled, dose‐ranging study assessing the safety, efficacy, and pharmacodynamics of the REG1 anticoagulation system in patients with acute coronary syndromes: design and rationale of the RADAR Phase IIb trial. Am Heart J 2011; 161: 261–8. [DOI] [PubMed] [Google Scholar]
  • 100. Abera G, Berhanu G, Tekewe A. Ribozymes: nucleic acid enzymes with potential pharmaceutical applications: a review. Pharmacophore 2012; 3: 164–78. [Google Scholar]
  • 101. Puerta‐Fernández E, Romero‐López C, Barroso‐delJesus A, Berzal‐Herranz A. Ribozymes: recent advances in the development of RNA tools. FEMS Microbiol Rev 2003; 27: 75–97. [DOI] [PubMed] [Google Scholar]
  • 102. Weng DE, Usman N. Angiozyme: a novel angiogenesis inhibitor. Curr Oncol Rep 2001; 3: 141–6. [DOI] [PubMed] [Google Scholar]
  • 103. Tong M, Schiff E, Jensen DM. Preliminary analysis of a phase II study of HEPTAZYMETM, a nuclease resistant ribozyme targeting hepatitis C virus (HCV) RNA. Hepatology 2002; 36: 360A. [Google Scholar]
  • 104. Enuka Y, Lauriola M, Feldman ME, Sas‐Chen A, Ulitsky I, Yarden Y. Circular RNAs are long‐lived and display only minimal early alterations in response to a growth factor. Nucleic Acids Res 2016; 44: 1370–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, et al. Exon‐intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol 2015; 22: 256–64. [DOI] [PubMed] [Google Scholar]
  • 106. Lasda E, Parker R. Circular RNAs: diversity of form and function. RNA 2014; 20: 1829–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, et al. miRNA‐dependent gene silencing involving Ago2‐mediated cleavage of a circular antisense RNA. EMBO J 2011; 30: 4414–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nat Biotechnol 2014; 32: 453–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Denzler R, Agarwal V, Stefano J, Bartel DP, Stoffel M. Assessing the ceRNA hypothesis with quantitative measurements of miRNA and target abundance. Mol Cell 2014; 54: 766–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z, Sharpless NE. Expression of linear and novel circular forms of an INK4/ARF‐associated non‐coding RNA correlates with atherosclerosis risk. PLoS Genet 2010; 6: e1001233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Qu S, Yang X, Li X, Wang J, Gao Y, Shang R, et al. Circular RNA: a new star of noncoding of RNAs. Cancer Lett 2015; 365: 141–8. [DOI] [PubMed] [Google Scholar]
  • 112. Li P, Chen S, Chen H, Mo X, Li T, Shao Y, et al. Using circular RNA as a novel type of biomarker in the screening of gastric cancer. Clin Chim Acta 2015; 444: 132–6. [DOI] [PubMed] [Google Scholar]

Articles from British Journal of Clinical Pharmacology are provided here courtesy of British Pharmacological Society

RESOURCES