Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW Human gene therapy – the future of health care Milad Soleimani, Maxime Merheb and Rachel Matar Department of Biotechnology, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates Abstract Gene therapy is the use of genes to heal diseases at the genetic level. It involves insertion, correction or inactivation of specific genes in organisms suffering from genetic disorders. Ever since it was introduced as a hypothesis in the 1960s, gene therapy has gone on to become a medical wonder that promises relief from the likes of Parkinson’s disease and multiple sclerosis. Today, the technique is used to manipulate the genome of not only somatic cells but also the germline cells of early embryos and gametes with a vast range of viral and non-viral vectors. The diseases targeted by gene therapy are among the deadliest on the planet: they are categorized into cancers, genetic deficiencies and autoimmune diseases. On the one hand, gene therapy is deemed by some to be an ethically and technically problematic technology because of its cost, its treatment of human dignity and its questionable accuracy and safety. In contrast to what its opponents say, the pro-gene therapy arguments support the technique by highlighting its remarkable efficiency, long-term cost-effectiveness and ability to cure unique diseases, among others. Introduction ‘I am sorry. There is nothing more we can do.’ That is what a doctor would say to a patient or the parents of a fetus suffering from, what might seem at this point, an incurable genetic disorder. The string of similar clichéd apologies was broken for the first time in 1990 by two brave souls, Dr Anderson and Dr Blaese, who took the initiative by treating a 4-year-old girl lacking an adenosine deaminase (ADA) gene with a role in immune responses.1 Gene therapy is a technique to alter aberrant phenotypic characteristics caused by mutations through approaches such as gene insertion, modification or regulation.2 The technique is particularly advantageous over conventional Correspondence: Rachel Matar, Department of Biotechnology, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates. Email: [email protected] medications in that it aims to correct the underlying cause of the disease, rather than its symptoms. Cancer, cardiovascular diseases and multiple sclerosis are some of the conditions with prospects for treatment with gene therapy. In addition, there have been attempts to tackle inherent genetic disorders at the embryonic stage. This form of gene therapy, however, has given rise to enormous controversy arising from the fear of misusing the technique for non-medical purposes, uncertainties regarding its success and other ethical issues. To address the issues, stringent regulations may be necessary; however, standing in the way of gene therapy merely out of fear is deemed unreasonable. Historical perspectives The idea of manipulating the genome emerged in the 1960s as the antisense concept, following the discovery of the DNA double helix and the explanation of its replication, expression and regulation.3 Joshua Lederberg was one of the pioneers who envisaged the possibility of controlling nucleotide sequences in human chromosomes and grafting DNA segments onto viral DNA.4 The hypothesis was reinforced during the 1970s, mainly as a result of the discovery of reverse transcriptase and the comprehensive studies on restriction enzymes.3 Soon after came two unapproved clinical trials: one to treat the arginase deficiency syndrome using the Shope papilloma virus, and the other to counteract β-thalassemia with bone marrow cells treated with a β-globin-containing plasmid.5 Despite the success of treating ADA in 1990, the decade that followed was full of disappointments, the biggest of which occurred in 1999 with the death of a teenager who had undergone gene therapy for a rare liver © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences 101 Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW disease.6 With improvements in our understanding of vectors, gene therapy has taken a sharp turn for the better, promising (science-grounded) miracles in the future. A concise timeline of the most salient events that have shaped gene therapy is shown in Table 1. Gene therapy: approaches, types and vectors In brief, gene-based therapeutics (gene therapy) may be generally defined as altering the expression of specific genes or correcting defective genes by introducing exogenous nucleic acids in order to prevent, arrest or reverse a pathological process.13 There are three chief approaches that are adopted to carry out gene therapy. Gene addition/insertion is the most common approach and involves introducing into cells a normal copy of a non-functional, mutated gene that is otherwise responsible for the production of a key protein. This method has been shown to tackle genetic disorders such as haemophilia14 and cystic fibrosis,15 which are characterized by the lack of the clotting factor IX and a chloride channel, respectively. The genes encoding these proteins are delivered into cells where they are expressed to provide the patient with the protein that they lack. Gene knockdown is the second approach, which entails regulating and/or inhibiting the expression of a mutant gene with the help of double-stranded RNA (dsRNA) via the process of RNA interference (RNAi). RNAi is a biological process whereby endogenously produced or artificially introduced dsRNA binds to the target messenger RNA (mRNA) and causes its degradation and, therefore, silences the target gene.13,16 Being a biological response, RNAi can be triggered in patients by the administration of short synthetic dsRNAs or by the expression of short hairpin RNAs (shRNAs). Its mechanism involves a series of events during which dsRNAs and shRNAs are processed mainly by an enzyme called Dicer to generate short interfering RNAs (siRNAs) and microRNAs (miRNAs), respectively. siRNAs bring about cleavage of their target mRNAs and can, therefore, be used to completely suppress disease-causing genes. miRNAs, on the other hand, follow a non-cleavage-dependent pathway, which renders them useful in modulating gene expression.17 The therapeutic applications of RNAi include, but are not limited to, the treatment of cancer,18–20 Alzheimer’s disease,21 rheumatic diseases22 and eye diseases.23,24 Gene correction/ modification is another approach wherein zinc-finger nucleases and DNA recombination techniques are applied to either repair a mutation or mutate a gene, encoding a virus receptor in order to thwart infection.25 Zinc-finger nucleases are powerful gene modification tools composed of a sequence-specific, customizable DNA-binding domain and a nuclease domain that functions only when bound to the target site.26 The cleavage of DNA evokes a damage response that is prominently in the form of non-homologous end joining which, despite being inaccurate, can be used to introduce small insertions and/or deletions to inactivate mutant genes.25,27 To augment the accuracy of the response, a synthetic donor DNA template homologous to the target is TABLE 1 Timeline of the most important events in the history of gene therapy 1959 1968 1970–1973 1974 1980 1983 1984 1990 1997 Physician Stanfield Rogers observed that the warts caused by the Shope papilloma virus on the skin of rabbits were rich in arginase, which he attributed to the introduction of a Shope virus-encoded arginase gene into rabbit skin cells3 Nobelist Marshall Nirenberg discussed programming cells with synthetic messages7 Stanfield Rogers used the Shope papilloma virus in an unsuccessful attempt to treat two sisters suffering from hyperargininaemia, as he believed the virus would induce arginase activity in the patients8 Creation of the Recombinant DNA Advisory Committee (RAC) to stringently regulate all gene transfer experiments (http://osp.od.nih.gov/office-biotechnology-activities/biomedical-technology-assessment/hgt/rac) Martin Cline conducted an unapproved experiment to transfer the β-thalassemia gene into the bone marrow of two patients with hereditary blood disorders9 Miller et al.10 built a retroviral vector and used it to transfer a human hypoxanthine-guanine phosphoribosyltransferase gene into rodent cells in vitro10 The RAC created the Human Gene Therapy Working Group specifically to review gene therapy protocols8 Anderson and Blaese cured a young girl suffering from severe combined immunodeficiency by injecting her with T cells carrying an adenosine deaminase gene11 A HIV-positive child was treated using recombinant CD34+ cells carrying a HIV-1 PRE decoy gene12 HIV, human immunodeficiency virus; PRE, progesterone-responsive element. 102 © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW delivered along with zinc-finger nucleases to replace the target with desired genetic information through homologous recombination.27,28 The method has been utilized in the treatment of HIV29 and diseases such as haemophilia,30 sickle-cell anaemia31 and the X-linked chronic granulomatous disease,32 and also in the creation of knockout models.33 Alternatively, a donor template that has only one mismatch with the target gene can be used to induce a site-specific correction of a point mutation in a process referred to as site-directed mutagenesis. A more commonly practised approach involves the use of RNA/DNA oligonucleotides, as they are more active in homologous pairing than DNA alone.34 Alexeev et al.35 used this approach to demonstrate that injection of albino mice with RNA/DNA oligonucleotides tailored to correct a point mutation in the tyrosinase gene would result in permanent and inheritable restoration of tyrosinase activity and melanin synthesis. Other disorders against which this method has been employed include sickle cell anaemia,36 Fabry’s disease34 and haemophilia B.37 Depending on the type of target cell, gene therapy is broadly classified as somatic or germline. Clearly, the former refers to correcting anomalies in non-reproductive cells in the patient, whereas the latter is often performed in early embryos, such that the change will be passed on to posterity.38 Somatic gene therapy could be implemented either in vitro or in vivo. The in vitro method consists of three key steps: (1) transforming a virus and suppressing its ability to reproduce; (2) transfecting the virus into cells isolated from a patient; and (3) injecting the genetically modified cells into the patient.39 Conversely, the in vivo method involves direct administration of injectable self-eliminating vectors (e.g. adenoviral vectors) to patients.40 Germline gene therapy is relatively cumbersome and sensitive with a protocol that comprises eight major steps: (1) isolation of undifferentiated cells from an embryo or a gamete; (2) culturing the cells in vitro; (3) transfection of the cells; (4) selection of the transfected cells; (5) replacement of the targeted gene; (6) removal of marker genes; (7) nuclear transfer into an unfertilized ovum; and (8) reimplantation of the ovum in the uterus.38 Vectors play a crucial role in transferring DNA into cells. They fall under the two classes of viral and non-viral vectors. Viral vectors are essentially viral particles composed of nucleic acid contained within a capsid protein that is, more often than not, covered by a membrane envelope. Generally, some of the structural genes of these vectors are deleted in order to prevent them from replicating in host cells.41 Gene delivery by means of replicationincompetent viral vectors has been demonstrated to be more efficient and less demanding in terms of dose number because of the presence of strong promoters in viral vectors.42 The genetic material of each type of viral vector may be different from those of others, which confers unique features for the therapeutic exploitation of the virus (Table 2). Notwithstanding their high efficiency and specific uptake, viral vectors are plagued by their stimulation of the host immune system that recognizes the vector and the transgene product(s) as a foreign agent.44 Non-viral vectors are deemed superior to viral vectors in some respects as they are safer, can be more easily constructed and modified and display a higher packaging capacity.45 Typically, non-viral vectors are in the form of either naked DNA or a complex of DNA and nanoparticles.46 Liposomes are known to be the most popular type of nanoparticles composed of natural or synthetic phospholipids and steroids or other surfactants. Thanks to their ability to fuse with biological membranes, liposomes have been successfully employed to transfect DNA into cells.47 Polymers constitute another class of non-viral vector for therapeutic gene delivery. The most studied polymers are dendrimers that are highly branched macromolecules, consisting of a central atom (e.g. nitrogen), interior layers of polymers and an exterior, cationic functionality layer.48 Microprojectiles such as tungsten and gold nanoparticles coated with DNA also serve as effective non-viral vectors that are blasted into target cells using a gene gun.46 Other notable modes of delivery for non-viral vectors, in general, include electroporation (relies on electrical pulses to create pores in the cell membrane),49 ultrasound50 and hydroporation (uses a highly pressured aqueous solution to generate membrane pores).51 Applications: case studies As far as its applications are concerned, gene therapy is a versatile tool, potentially capable of treating a range of disorders that require gene correction or replacement. Cancer remains, by far, the most investigated potential application of gene therapy (Table 3). The most successful clinical trials, however, are those that address cardiovascular diseases.52 This review will discuss specific studies in © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences 103 Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW TABLE 2 Genetic material, advantages and disadvantages of viral vectors43 Viral vector Genetic material Retroviral vectors dsRNA Lentiviral vectors dsRNA Adenovirus vectors dsDNA Herpes virus vectors dsDNA Poxvirus vectors dsRNA Adeno-associated virus (AAV) dsDNA Advantages Disadvantages Insert capacity for transgene <7–8kb Stable integration into host DNA Low immunogenicity Broad cell tropism of infectivity Prolonged expression Infect proliferating and non-proliferating cells Stable gene expression High transfection efficiency Efficient targeted transfection Infects dividing and non-dividing cells Infects a wide variety of cell types Insertion capacity of up to 50kb Generation of high virus titres High insertion capacity High expression levels Infect dividing and non-dividing cells Very prolonged expression Low immunogenicity Non-pathogenic Difficult targeting of viral infection Transfect only proliferating cells Concern of insertional mutagenesis Potential insertional mutagenesis No clinical trials Immune response to viral proteins Insert size limit of 7.5kb Transient gene expression Possible toxicities Risk of recombination Possible cytopathic effects Insert size limit of 4.5kb Insufficient clinical trials Requirement of adenovirus or herpes virus for AAV replication dsDNA, double-stranded DNA. the application of gene therapy against colon cancer, heart failure, multiple sclerosis and type 1 diabetes mellitus. Given their high death toll, cancers, both benign and malignant, are relentlessly investigated for the possibility of using gene therapy as a form of treatment. Researchers are constantly improving the technique for the purpose of presenting it as a safe and superior alternative to chemotherapy, which has the potential to cause systemic toxicity and/or, in the long run, lead to other types of cancer.53 TABLE 3 Proportions of gene therapy clinical trials for various diseases52 Disease/indication Proportion of trials (%) Cancer Monogenic diseases Cardiovascular diseases Infectious diseases Neurological diseases Ocular diseases Inflammatory diseases Other diseases Gene marking Healthy volunteers 64.4 8.7 8.4 8.0 2.0 1.5 0.7 1.4 2.7 2.3 104 The studies in this regard are largely focused on two strategies, namely immunotherapy and oncolytic virotherapy. Briefly, immunotherapy involves use of cells transfected with cytokine genes in an attempt to provoke a robust immune response against existing tumour(s).54 Oncolysis, on the other hand, uses viruses that replicate within tumours, destroying them in the process. As a case study on oncolysis, Yoon and collaborators55 assessed the efficacy of a herpes simplex virus (HSV) mutant hrR3, containing a β-galactosidase insert within its ribonucleotide reductase gene for cancer therapy. The virus was expected to proliferate in tumours, depending on the ribonucleotide reductase supply by cancer cells. In cell culture studies, human, as well as murine, colon carcinoma and hepatocytes were infected with hrR3, and subsequently subjected to viral cytotoxicity assays. The number of surviving cells was greater in the case of hepatocytes, indicating the effectiveness of the virus as an antitumour agent. Results of animal studies were equally favourable, as mouse models suffering from carcinoma showed tumour lysis with blue (β-galactosidase) spots in the cleared regions upon infection with hrR3.55 Similar to cancer, cardiovascular diseases are frequently cited among those that gene therapy is © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW expected to cure. These diseases include, among others, heart failure, arrhythmia and coronary artery disease.56 Both the in vivo (percutaneous) and the in vitro approaches to gene delivery may involve non-viral and/or viral vectors.57 That said, viral vectors are given preference, as they have proven more efficient and some of them (e.g. adenoviruses) can even infect the non-dividing cells of the cardiovascular system.56 Jessup et al.58 specifically exploited adeno-associated viruses in a study on 39 patients to examine the efficacy of sarcoplasmic reticulum-calcium (Ca2+)-adenosine triphosphatase (ATPase) (SERCA2a) in alleviating the symptoms of heart failure. It is worth noting that reduced calcium uptake, which has been observed in failing hearts, is closely associated with a decrease in the expression and activity of SERCA2a. To normalize intracellular calcium cycling and correct cardiac metabolism, the authors randomly administered the patients with either one of the three different doses of SERCA2a-transformed adeno-associated virus type 1 or a placebo by coronary artery infusion over a 12-month period. In the end, their results attested to the effectiveness of the method, as the patients who had received higher doses exhibited considerable improvement in functional capacity, symptoms and cardiac structure.58 Multiple sclerosis (MS) is an autoimmune disease with good prospects for a gene therapy-based cure. Patients afflicted by this chronic, neurological disease of the central nervous system experience episodes of neurological deficits in the earlier stages, followed by progressive neurological deterioration.59 Currently, a wide range of disease-modifying therapies, such as those using interferon-β (IFN-β) (down-regulating T-helper cells), glatiramer acetate (Copaxone®, Teva UK, Essex, UK; preventing lymphocyte proliferation) and mitoxantrone (inhibiting repair by topoisomerase), are in use or under study.60 However, the fact that most of these drugs do not elicit an optimal response in many patients has directed attention towards gene therapy. Ryu and colleagues61 reported the use of IFN-β as a therapeutic gene to reduce the autoimmune effects of MS. Initially, human bone marrow-derived mesenchymal cells (MSCs) were cultured and infected with adenoviral vectors transformed with either green fluorescent protein (GFP) or murine IFN-β. To induce experimental allergic encephalomyelitis (an animal model for MS), the mice were given the MOG35–55 peptide emulsified in Freund’s adjuvant containing Mycobacterium tuberculosis. Later, the models were divided into two groups, each receiving either MSCs–GFP or MSCs–IFN-β. The impact of the therapy was evaluated, taking into account its various aspects and likely consequences. Histological analysis of spinal cord sections taken from mice containing MSCs–GFP (control) and MSCs–IFN-β revealed that demyelination and infiltration were more severe in the former. In addition, the results of an enzyme-linked immunosorbent assay confirmed the immunomodulatory effect of the therapy by indicating that cytokine levels were far lower in MSCs–IFN-β recombinants. To those, one has to add the relatively high expression levels of a regeneration stimulant called the neurotrophic factor in the MSCs–IFN-β models that further added weight to the reliability of the method.61 Type I diabetes mellitus (T1DM) is another autoimmune disease, among several others, that has been shown to be a viable candidate for treatment by gene therapy. T1DM occurs as the result of the autoimmune destruction of insulin-secreting β-cells that induce the removal and metabolism of glucose by muscle and liver cells, respectively.62 T1DM is a polygenic disease, as it is caused by the combined effect of more than one underlying susceptibility gene. About 18 loci (regions) of the genome, besides environmental factors such as diet and infection(s), are known to contribute to the onset of T1DM. Most of these loci harbour multiple genes and are labelled insulin-dependent diabetes mellitus 1 (IDDM1) to 18 (IDDM18). The two most researched loci, IDDM1 and IDDM2, contain genes that encode immune response proteins and preproinsulin, respectively.63 Currently, the sole treatment option for diabetic patients is insulin administration. However, exogenous insulin is not physiologically regulated. Moreover, the occasional lack of patient adherence exacerbates the condition, resulting in suboptimal control and secondary complications.64 These setbacks can be avoided with gene therapy; nevertheless, ectopic insulin expression alone cannot cure T1DM unless it is accompanied by a regulatory system that responds to variations in glucose concentration in the blood.65 In a recent effort to counteract T1DM using gene therapy, Callejas and coworkers66 aimed at increasing the muscular uptake of glucose through the coexpression of insulin and glucokinase (GK) in skeletal muscle cells. Insulin was required to ensure transport of glucose by insulin-stimulated glucose transporter 4 (GLUT4) and GK was made available to © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences 105 Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW facilitate glucose uptake by muscle cells in the event of hyperglycaemia (high blood glucose). Five beagle dogs induced with streptozotocin and alloxan to exhibit symptoms of T1DM were injected in the leg with adeno-associated viral vectors that carried either an insulin- or a GK-encoding gene. The immunohistochemical analysis and radioimmunoassay of insulin in conjunction with the measurement of blood glucose using a Glucometer Elite analyser (Bayer, Leverkuser, Germany) indicated the normalization of insulin and glucose levels. Further, northern blot analysis confirmed the expression of the two transgenes in the skeletal muscle biopsies of the models. The long-term efficacy of this approach was established as the models, so far, have lived in good health for more than four years. The prospects of gene therapy go well beyond the current scope of its applications. Researchers are constantly pursuing new strategies that tap into the therapeutic potentials of various cellular components and machineries. Pseudogenes, for instance, can be thought of as suitable tools for gene therapy. In simple terms, a pseudogene is a (usually) non-functional DNA sequence that bears a resemblance to one or two functional parent genes in the genome. The lack of function is because of either the presence of mutations in the pseudogene or the loss of regulatory sequences, resulting in the disruption of transcription or translation.67,68 Dewannieux et al.69 showed that the human endogenous retrovirus HERV-K (HML2) (a non-functional remnant of ancestral retroviral infections) could regain its infectiousness and repeat its life cycle following a multistep recombination event with functional alleles.69 This suggests the possibility of resuscitating beneficial pseudogenes such as ψ connexin 43 (ψCx43), which tends to suppress the growth of breast cancer cells,70 for therapeutic purposes. Arguments: for and against Although everyone more or less acknowledges the tremendous potential of gene therapy to revolutionize clinical sciences, some sceptics with backgrounds in both science and ethics have raised questions that not only pose a challenge to its technical aspects but also underline the moral issues provoked by gene therapy. The vast majority of these questions concern the germline mode of gene therapy, as its outcomes are, contrary to the somatic mode, not confined to the patient under treatment. 106 Yet some of the opponents even revile somatic gene therapy, despite its wide public acceptance. Among the critics, some have gone so far as to argue against gene therapy in its entirety. One of their arguments is that gene therapy is still in its infancy, with a scope that is too limited to overcome its limitations and prevent its possible adverse consequences. These consequences usually arise from flawed gene transfer that may result in mutations (insertional mutagenesis), up-regulation of oncogenes or inactivation of tumour suppressor genes.71 In addition, the present shortcomings of gene therapy are pointed out as major obstacles to its success. Some of the most common genetic disorders such as T1DM, hypertension and Alzheimer’s disease are polygenic in nature. Such disorders, unlike their monogenic counterparts, are extremely complicated to treat effectively and permanently using gene therapy.72 The opponents also assert that the very high cost of gene therapy renders it against fairness and equality, as it would allow only the affluent minority to benefit from treatment.73 Yet another concern raised is that altering the human genome, which is perceived as the blueprint of life, in any form or fashion is an act of ‘playing God’, with serious threats to human dignity.74 As for somatic gene therapy in particular, its opponents express their fear of the probable technical errors and the slippery slope that could lead to crossing the barrier between the somatic mode and the germline mode of therapy. From a technical point of view, they state that the incorrect choice of vectors and target cells could pave the way for a therapeutic transgene to spread to the gonads and affect the genotype of the offspring.73 Socioethically, they claim that permitting somatic gene therapy is a preliminary step on a slippery slope towards what is described as the horrendous manipulation of humankind that is made possible by germline gene therapy.75 When it comes to germline gene therapy, the debate get more intense and the naysayers’ tone more resentful. Their argument is essentially based upon the consequences of deviating from a treatment-centred use of the therapy. The most frequently mentioned concern is employing the method for genetic enhancement. This is often illustrated with healthy individuals such as athletes who seek physiological improvements6 and ‘designer babies’ possessing their parents’ favourite © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW traits.76 Such activities may, as the commentators suggest, set in motion a chain of events leading to the re-emergence of eugenics. Another concern surrounding germline gene therapy is that any accidental mistakes in the process will undoubtedly create a lineage of people whose genomes have been not only manipulated without their consent but also disrupted before they were born.77 On the other hand, the pro-gene therapy arguments hail the technique as a highly efficient and absolutely ethical saviour of human beings in the face of otherwise incurable diseases. People holding this perspective view gene therapy as a continuation of other conventional therapies with bigger, life-saving promises and exaggerated drawbacks. They contend that germline gene therapy is exceptionally efficient, as its application in one individual can preclude the reappearance of a disorder in succeeding generations. Evidently, it would also be less costly for patients and useful in conserving future health care resources.73,78 Furthermore, the advocates put forth the rationale that gene therapy is necessary owing to its ability to cure unique diseases such as Parkinson’s disease79 and retinoblastoma80 that other therapies have consistently failed to beat. On ethical grounds, gene therapy is considered pro-life by the likes of French Anderson, a leading National Institutes of Health investigator, who believes ‘it would be unethical to delay human trials’, as he thinks that ‘the patients have little other hope’.81 In addition, germline gene therapy provides the opportunity for couples to save their genetically challenged embryos, rather than discard them outright.78 those against gene therapy overstate a few points, such as the lack of morality in modifying genes and the possibility of the non-medical use of gene therapy, without appreciating its very real prospects. First, the notion that altering DNA poses a threat to human dignity is subjective, and may vary from one belief system or theory of ethics to another. This raises the question of whose beliefs should be preferred to those of others who might possibly be in favour of gene therapy. Second, the fear that allowing one form of gene therapy could facilitate the emergence of the other, which in turn could be exploited for non-medical purposes, is merely justified by a rhetorical slippery slope that an action will ultimately culminate in havoc via a chain of uncontrolled events. Although regulation is essential, the slippery slope argument, as it is put forth, would call a halt to most of the ongoing medical and non-medical research across the globe. Third, the fact that gene therapy could be error prone holds true for most scientific developments. However, rigorous research on the technique to verify its safety could significantly minimize the likely errors. In addition, devising and enforcing such regulations as prohibition of non-medical uses of gene therapy, requirement of the patient’s informed consent and proof of the necessity of gene therapy to treat a disease can further help address the issue. In the end, bringing mankind’s creativity, which fuels developments in gene therapy, to a standstill is taking the easy way out, rather than dealing with the issue. References 1 Conclusions 2 The premise of gene therapy as a novel therapeutic technique, capable of introducing new genes and/or correcting existing defective genes in an organism, is as ethically controversial as it is scientifically ground breaking. Both the somatic and germline modes of gene therapy have spurred vehement debates among those who consider it immoral and unreliable and others who admire it on account of its unique and medically plausible potentials. An overall look at the issue and the positions on either side reveals that some aspects or others are being undermined by either the supporters or the critics. On the one hand, those that support gene therapy usually downgrade the fact that it could lead to unexpected repercussions, and, on the other hand, 3 © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences 4 5 6 7 8 Schmeck J, Harold M. A new era of gene therapy. FDA Consumer 1991; 25:14. Kelly EB. Gene Therapy. Westport, CT: Greenwood Press; 2007, pp. x, 198. Judson H. The glimmering promise of gene therapy. MIT Technology Review 2006; 109:40–7. Cotrim AP, Baum BJ. Gene therapy: some history, applications, problems, and prospects. J Toxicol Pathol 2008; 36:97–103. http://dx.doi.org/10.1177/ 0192623307309925 Scollay R. Gene therapy. Ann N Y Acad Sci 2001; 953a:26–30. http://dx.doi.org/10.1111/j.1749-6632. 2001.tb11357.x Macrina FL. Scientific Integrity: Text and Cases in Responsible Conduct of Research, 3rd ed. Washington, DC: ASM Press; 2005, pp. xxv, 402. Nirenberg M. Genetic memory. J Am Med Assoc 1968; 206:1973–7. http://dx.doi.org/10.1001/jama. 1968.03150090049012 Bauer G, Anderson J. History of gene therapy. Gene therapy for HIV. New York, NY: Springer; 2014, pp. 9–15. http://dx.doi.org/10.1007/978-1-4939-0434-1_2 107 Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 108 Martin A, Thompson AA. Thalassemias. Pediat Clin North Am 2013; 60:1383–91. http://dx.doi.org/10.1016/ j.pcl.2013.08.008 Miller AD, Eckner RJ, Jolly DJ, Friedmann T, Verma IM. Expression of a retrovirus encoding human HPRT in mice. Science 1984; 225:630–2. http://dx.doi.org/ 10.1126/science.6377498 Blaese RM, Culver KW, Chang L, et al. Treatment of severe combined immunodeficiency disease (SCID) due to adenosine deaminase deficiency with CD34+ selected autologous peripheral blood cells transduced with a human ADA gene. Amendment to clinical research project, Project 90-C-195, January 10, 1992. Hum Gene Ther 1993; 4:521–7. http://dx.doi.org/ 10.1089/hum.1993.4.4-521 Kohn DB, Parkman R. Gene therapy for newborns. FASEB J 1997; 11:635–9. Kay MA. State-of-the-art gene-based therapies: the road ahead. Nat Rev Genet 2011; 12:316–28. http://dx. doi.org/10.1038/nrg2971 Tuddenham E. Gene therapy for haemophilia B. Haemophilia 2012; 18(Suppl. 4):13–17. http://dx.doi. org/10.1111/j.1365-2516.2012.02823.x Hyde SC, Gill DR, Higgins CF, et al. Correction of the ion transport defect in cystic fibrosis transgenic mice by gene therapy. Nature 1993; 362:250–5. http://dx. doi.org/10.1038/362250a0 Milhavet O, Gary DS, Mattson MP. RNA interference in biology and medicine. Pharmacol Rev 2003; 55:629–48. http://dx.doi.org/10.1124/pr.55.4.1 Pushparaj PN, Aarthi JJ, Manikandan J, Kumar SD. siRNA, miRNA, and shRNA: in vivo applications. J Dent Res 2008; 87:992–1003. http://dx.doi.org/10.1177/ 154405910808701109 Felipe AV, Oliveira J, Chang PY, et al. RNA interference: a promising therapy for gastric cancer. Asian Pac J Cancer Prev 2014; 15:5509–15. http://dx.doi.org/ 10.7314/APJCP.2014.15.14.5509 Shi Y, Tian Y, Zhou YQ, Ju JY, Liu Y, Zhu LP. Reduction of CD44 expression results in growth inhibition of human nasopharyngeal carcinoma cell CNE-2L2 in vitro. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2007; 29:67–72. Zeng H, Wei W, Xu X. Chemokine (C-X-C motif) receptor 4 RNA interference inhibits bone metastasis in breast cancer. Oncol Lett 2014; 8:77–81. Orlacchio A, Bernardi G, Orlacchio A, Martino S. RNA interference as a tool for Alzheimer’s disease therapy. Mini Rev Med Chem 2007; 7:1166–76. http://dx.doi. org/10.2174/138955707782331678 de Franca NR, Mesquita Júnior D, Lima AB, Pucci FV, Andrade LE, Silva NP. RNA interference: a new alternative for rheumatic diseases therapy. Rev Bras Reumatol 2010; 50:695–702. Ramachandran PS, Bhattarai S, Singh P, et al. RNA interference-based therapy for spinocerebellar ataxia type 7 retinal degeneration. PLOS ONE 2014; 9:e95362. http://dx.doi.org/10.1371/journal.pone.0095362 Jiang L, Frederick JM, Baehr W. RNA interference gene therapy in dominant retinitis pigmentosa and cone-rod dystrophy mouse models caused by GCAP1 mutations. Front Mol Neurosci 2014; 7:25. http://dx. doi.org/10.3389/fnmol.2014.00025 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 2010; 11:636–46. http://dx. doi.org/10.1038/nrg2842 Rahman SH, Maeder ML, Joung JK, Cathomen T. Zinc-finger nucleases for somatic gene therapy: the next frontier. Hum Gene Ther 2011; 22:925–33. http://dx.doi.org/10.1089/hum.2011.087 Carroll D. Progress and prospects: zinc-finger nucleases as gene therapy agents. Gene Ther 2008;15:1463–8. http://dx.doi.org/10.1038/gt.2008.145 Gellhaus K, Cornu TI, Heilbronn R, Cathomen T. Fate of recombinant adeno-associated viral vector genomes during DNA double-strand break-induced gene targeting in human cells. Hum Gene Ther 2010; 21:543–53. http://dx.doi.org/10.1089/hum.2009.167 Perez EE, Wang J, Miller JC, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 2008; 26:808–16. http://dx.doi.org/10.1038/nbt1410 Li H, Haurigot V, Doyon Y, et al. In vivo genome editing restores haemostasis in a mouse model of haemophilia. Nature 2011; 475:217–21. http://dx.doi. org/10.1038/nature10177 Sebastiano V, Maeder ML, Angstman JF, et al. In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem Cells 2011; 29:1717–26. http://dx.doi.org/10.1002/stem.718 Zou J, Sweeney CL, Chou BK, et al. Oxidase-deficient neutrophils from X-linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease-mediated safe harbor targeting. Blood 2011; 117:5561–72. http://dx.doi.org/10.1182/ blood-2010-12-328161 Yang D, Zhang J, Xu J, et al. Production of apolipoprotein C-III knockout rabbits using zinc finger nucleases. J Vis Exp 2013; 81: e50957. http://dx.doi. org/10.3791/50957 Lai LW, Lien YH. Therapeutic application of chimeric RNA/DNA oligonucleotide based gene therapy. Expert Opin Biol Ther 2001; 1: 41–7. http://dx.doi.org/ 10.1517/14712598.1.1.41 Alexeev V, Yoon K. Gene correction by RNA–DNA oligonucleotides. Pigment Cell Melanoma Res 2000; 13:72–9. http://dx.doi.org/10.1034/j.1600-0749. 2000.130205.x Cole-Strauss A, Yoon K, Xiang Y, et al. Correction of the mutation responsible for sickle cell anemia by an RNA–DNA oligonucleotide. Science 1996; 273:1386–9. http://dx.doi.org/10.1126/science.273.5280.1386 Nichols TC, Dillow AM, Franck HW, et al. Protein replacement therapy and gene transfer in canine models of hemophilia A, hemophilia B, von Willebrand disease, and factor VII deficiency. ILAR J 2009; 50:144–67. http://dx.doi.org/10.1093/ilar.50.2.144 Nielsen T. Human germline gene therapy. McGill J Med 1997; 3:126–32. Patil SS, Gentschev I, Adelfinger M, et al. Virotherapy of canine tumors with oncolytic vaccinia virus GLV-1h109 expressing an anti-VEGF single-chain antibody. PLOS ONE 2012; 7:e47472. http://dx.doi.org/ 10.1371/journal.pone.0047472 © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 Reynolds PN, Zinn KR, Gavrilyuk VD, et al. A targetable, injectable adenoviral vector for selective gene delivery to pulmonary endothelium in vivo. Mol Ther 2000; 2:562–78. http://dx.doi.org/10.1006/ mthe.2000.0205 Lundstrom K, Boulikas T. Viral and non-viral vectors in gene therapy: technology development and clinical trials. Technol Cancer Res Treat 2003; 2:471–86. http://dx.doi.org/10.1177/153303460300200513 Kay MA, Liu D, Hoogerbrugge PM. Gene therapy. Proc Natl Acad Sci USA 1997; 94:12744–6. http://dx.doi.org/ 10.1073/pnas.94.24.12744 Mountain A. Gene therapy: the first decade. Trends Biotechnol 2000; 18:119–28. http://dx.doi.org/10.1016/ S0167-7799(99)01416-X Thomas CE, Ehrhardt A, Kay MA. Progress and problems with the use of viral vectors for gene therapy. Nat Rev Genet 2003; 4:346–58. http://dx.doi. org/10.1038/nrg1066 Razi Soofiyani S, Baradaran B, Lotfipour F, Kazemi T, Mohammadnejad L. Gene therapy, early promises, subsequent problems, and recent breakthroughs. Adv Pharma Bull 2013; 3:249–55. Mali S. Delivery systems for gene therapy. Indian J Hum Genet 2013; 19:3–8. http://dx.doi.org/10.4103/ 0971-6866.112870 Abhilash M. Nanorobots. Int J Pharma Bio Sci 2010; 1:1–10. Pushkar S, Philip A, Pathak K, Pathak D. Dendrimers: nanotechnology derived novel polymers in drug delivery. Indian J Pharm Educ Res 2006; 40:153–58. Brown PA, Bodles-Brakhop AM, Pope MA and Draghia-Akli R. Gene therapy by electroporation for the treatment of chronic renal failure in companion animals. BMC Biotechnol 2009; 9:4. http://dx.doi.org/ 10.1186/1472-6750-9-4 Zhou Z, Zhang P, Ren J, et al. Synergistic effects of ultrasound-targeted microbubble destruction and TAT peptide on gene transfection: an experimental study in vitro and in vivo. J Control Release 2013; 170:437–44. http://dx.doi.org/10.1016/j.jconrel. 2013.06.005 Zhang G, Gao X, Song Y, et al. Hydroporation as the mechanism of hydrodynamic delivery. Gene Ther 2004; 11:675–82. http://dx.doi.org/10.1038/sj.gt.3302210 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. http://dx.doi.org/10.1002/jgm.2698 Cross D, Burmester JK. Gene therapy for cancer treatment: past, present and future. Clin Med Res 2006; 4:218–27. http://dx.doi.org/10.3121/cmr.4.3.218 Kowalczyk DW, Wysocki PJ, Mackiewicz A. Cancer immunotherapy using cells modified with cytokine genes. Acta Biochim Pol 2003; 50:613–24. Yoon SS, Nakamura H, Carroll NM, Bode BP, Chiocca EA, Tanabe KK. An oncolytic herpes simplex virus type 1 selectively destroys diffuse liver metastases from colon carcinoma. FASEB J 2000; 14:301–11. Wolfram JA and Donahue JK. Gene therapy to treat cardiovascular disease. Journal of the American Heart Association. 2013; 2: e000119. http://dx.doi.org/ 10.1161/JAHA.113.000119 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences Baumgartner I, Isner JM. Somatic gene therapy in the cardiovascular system. Annu Rev Physiol 2001; 63:427–50. http://dx.doi.org/10.1146/annurev. physiol.63.1.427 Jessup M, Greenberg B, Mancini D, et al. Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID): a phase 2 trial of intracoronary gene therapy of sarcoplasmic reticulum Ca2+-ATPase in patients with advanced heart failure. Circulation 2011; 124:304–13. http://dx.doi.org/ 10.1161/CIRCULATIONAHA.111.022889 Goldenberg MM. Multiple sclerosis review. PT 2012; 37:175–84. Castro-Borrero W, Graves D, Frohman TC, et al. Current and emerging therapies in multiple sclerosis: a systematic review. Ther Adv Neurol Disord 2012; 5:205–20. http://dx.doi.org/10.1177/ 1756285612450936 Ryu CH, Park KY, Hou Y, Jeong CH, Kim SM, Jeun S-S. Gene therapy of multiple sclerosis using interferon β-secreting human bone marrow mesenchymal stem cells. BioMed Res Int 2013; 2013:696738. http://dx.doi. org/10.1155/2013/696738 Lee DD, Grossman E, Chong AS, Scherbaum WA. Cellular therapies for type I diabetes. Horm Metab Res 2008; 40: 147–54. http://dx.doi.org/10.1055/s-20081042430 Dean L, McEntyre J. Introduction to Diabetes. In: Dean L, McEntyre J (eds). The Genetic Landscape of Diabetes. Bethesda, MD: National Center for Biotechnology Information (US), National Library of Medicine, National Institutes of Health; 2004. Yechoor V, Chan L. Gene therapy progress and prospects: gene therapy for diabetes mellitus. Gene Ther 2004; 12:101–7. http://dx.doi.org/10.1038/ sj.gt.3302412 Gavriliuc OI, Tatu CA, Paunescu V. The race to cure diabetes: how far are we from a breakthrough? Fiziologia 2010; 20:4. Callejas D, Mann CJ, Ayuso E, et al. Treatment of diabetes and long-term survival following insulin and glucokinase gene therapy. Diabetes 2013; 62:1718–29. http://dx.doi.org/10.1098/rstb.2013.0507 Roberts TC, Morris KV, Wood MJ. The role of long non-coding RNAs in neurodevelopment, brain function and neurological disease. Phil Trans R Soc B Biol Sci 2014; 369:20130507. http://dx.doi.org/10.1016/ S0014-5793(00)01199-6 Mighell A, Smith N, Robinson P, Markham A. Vertebrate pseudogenes. FEBS Lett 2000; 468:109–14. http://dx.doi.org/10.1101/gr.5565706 Dewannieux M, Harper F, Richaud A, et al. Identification of an infectious progenitor for the multiple-copy HERV-K human endogenous retroelements. Genome Res 2006; 16:1548–56. http://dx.doi.org/10.1158/1535-7163.MCT-08-0930 Bier A, Oviedo-Landaverde I, Zhao J, Mamane Y, Kandouz M, Batist G. Connexin43 pseudogene in breast cancer cells offers a novel therapeutic target. Mol Cancer Ther 2009; 8:786–93. http://dx.doi.org/ 10.1182/asheducation-2007.1.460 Dunbar CE. The yin and yang of stem cell gene therapy: insights into hematopoiesis, leukemogenesis, 109 Hamdan Medical Journal 2015; 8:101–110 (http://dx.doi.org/10.7707/hmj.304) REVIEW 72 73 74 75 76 110 and gene therapy safety. Hematology Am Soc Hematol Educ Program 2007: 460–5. Misra S. Human gene therapy: a brief overview of the genetic revolution. J Assoc Phys India 2013; 61:127–33. Sade RM, Khushf G. Gene therapy: ethical and social issues. J SC Med Assoc 1998; 94:406–10. http://dx.doi. org/10.1097/SMJ.0b013e31802e645f Matthews QL, Curiel DT. Gene therapy: human germline genetic modifications–assessing the scientific, socioethical, and religious issues. South Med J 2007; 100:98–100. http://dx.doi.org/10.1136/ jme.21.6.350 McGleenan T. Human gene therapy and slippery slope arguments. J Med Ethics 1995; 21:350–5. http://dx.doi.org/10.1038/nrg2317 Kimmelman J. The ethics of human gene transfer. Nat Rev Genetics 2008; 9:239–44. http://dx.doi.org/ 10.1093/jmp/10.3.275 77 78 79 80 81 Anderson WF. Human gene therapy: scientific and ethical considerations. J Med Philos 1985; 10:275–91. Elias S, Annas GJ. Somatic and germline gene therapy. In: Elias S, Annas G (eds). Gene Mapping: Using Law and Ethics as Guides. Oxford, UK: Oxford University Press; 1992, pp. 151–165. http://dx.doi.org/ 10.1101/cshperspect.a009431 Coune PG, Schneider BL, Aebischer P. Parkinson’s disease: gene therapies. Cold Spring Harb Perspect Med 2012; 2:a009431. http://dx.doi.org/10.1038/ nm1296-1316 Riley DJ, Nikitin AY, Lee WH. Adenovirus-mediated retinoblastoma gene therapy suppresses spontaneous pituitary melanotroph tumors in Rb+/– mice. Nat Med 1996; 2:1316–21. http://dx.doi.org/10.1093/jmp/ 14.6.681 Anderson WF. Human gene therapy: why draw a line? J Med Philos 1989; 14:681–93. © 2015 The Author(s) Journal Compilation © 2015 Sheikh Hamdan Bin Rashid Al Maktoum Award for Medical Sciences