Gene therapy

From Infogalactic: the planetary knowledge core
(Redirected from Genetic enhancement)
Jump to: navigation, search
File:Gene therapy.jpg
Gene therapy using an adenovirus vector. In some cases, the adenovirus will insert the new gene into a cell. If the treatment is successful, the new gene will make a functional protein to treat a disease.

Gene therapy is the therapeutic delivery of nucleic acid polymers into a patient's cells as a drug to treat disease.

The origins of gene therapy can be traced back to the first live attenuated vaccines in the 1950s.[1] Although attenuated vaccines do not alter extant human genes, viruses are RNA polymers with their own genetic code that acts upon human cells, thus live vaccines can be considered a primitive form of gene therapy, albeit not in the sense that is generally implied today.

The first attempt at modifying human DNA was performed in 1980 by Martin Cline, but the first successful and approved nuclear gene transfer in humans was performed in May 1989.[2] The first therapeutic use of gene transfer as well as the first direct insertion of human DNA into the nuclear genome was performed by French Anderson in a trial starting in September 1990.

Between September 1990 and January 2014 some 2,000 clinical trials had been conducted or approved.[3][4]

It should be noted that not all medical procedures that introduce alterations to a patient's genetic makeup can be considered gene therapy. Bone marrow transplantation, and organ transplants in general have been found to introduce foreign DNA into patients.[5] Gene therapy is defined by the precision of the procedure and the intention of direct therapeutic effects.<templatestyles src="Template:TOC limit/styles.css" />

Background

Gene therapy was conceptualized in 1972, by authors who urged caution before commencing human gene therapy studies.

The first attempt, albeit an unsuccessful one, at gene therapy (as well as the first case of medical transfer of foreign genes into humans not counting organ transplantation) was performed by Martin Cline on 10 July 1980.[6][7] Cline claimed that one of the genes in his patients was active six months later, though he never published this data or had it verified [8] and even if he is correct, it's unlikely it produced any significant beneficial effects treating beta-thalassemia.[9]

After extensive research on animals throughout the 1980s and a 1989 bacterial gene tagging trial on humans, the first gene therapy widely accepted as a success was demonstrated in a trial that started on September 14, 1990, when Ashi DeSilva was treated for ADA-SCID.[10]

The first somatic treatment that produced a permanent genetic change was performed in 1993.[11]

The first germ line gene therapy consisted of producing a genetically engineered embryo in October 1996. The baby was born on July 21, 1997 and was produced by taking a donor's egg with healthy mitochondria, removing its nuclear DNA and filling it with the nuclear DNA of the biological mother - a procedure known as cytoplasmic transfer.[12]

This procedure was referred to sensationally and somewhat inaccurately in the media as a "three parent baby", though mtDNA is not the primary human genome and has little effect on an organism's individual characteristics beyond powering their cells.

Gene therapy is a way to fix a genetic problem at its source. The polymers are either expressed as proteins, interfere with protein expression, or possibly correct genetic mutations.

The most common form uses DNA that encodes a functional, therapeutic gene to replace a mutated gene. The polymer molecule is packaged within a "vector", which carries the molecule inside cells.

Early clinical failures led to dismissals of gene therapy. Clinical successes since 2006 regained researchers' attention, although as of 2014, it was still largely an experimental technique.[13] These include treatment of retinal disease Leber's congenital amaurosis,[14][15][16][17] X-linked SCID,[18] ADA-SCID,[19][20] adrenoleukodystrophy,[21] chronic lymphocytic leukemia (CLL),[22] acute lymphocytic leukemia (ALL),[23] multiple myeloma,[24] haemophilia[20] and Parkinson's disease.[25] Between 2013 and April 2014, US companies invested over $600 million in the field.[26]

The first commercial gene therapy, Gendicine, was approved in China in 2003 for the treatment of certain cancers.[27] In 2011 Neovasculgen was registered in Russia as the first-in-class gene-therapy drug for treatment of peripheral artery disease, including critical limb ischemia.[28] In 2012 Glybera, a treatment for a rare inherited disorder, became the first treatment to be approved for clinical use in either Europe or the United States after its endorsement by the European Commission.[13][29]

Approaches

Following early advances in genetic engineering of bacteria, cells and small animals, scientists started considering how to apply it to medicine. Two main approaches were considered – replacing or disrupting defective genes.[30] Scientists focused on diseases caused by single-gene defects, such as cystic fibrosis, haemophilia, muscular dystrophy, thalassemia and sickle cell anemia. Glybera treats one such disease, caused by a defect in lipoprotein lipase.[29]

DNA must be administered, reach the damaged cells, enter the cell and express/disrupt a protein.[31] Multiple delivery techniques have been explored. The initial approach incorporated DNA into an engineered virus to deliver the DNA into a chromosome.[32][33] Naked DNA approaches have also been explored, especially in the context of vaccine development.[34]

Generally, efforts focused on administering a gene that causes a needed protein to be expressed. More recently, increased understanding of nuclease function has led to more direct DNA editing, using techniques such as zinc finger nucleases and CRISPR. The vector incorporates genes into chromosomes. The expressed nucleases then "edit" the chromosome. As of 2014 these approaches involve removing cells from patients, editing a chromosome and returning the transformed cells to patients.[35]

Future of CRISPR-Cas 9

A duplex of crRNA and tracrRNA acts as guide RNA to introduce a specifically located gene modification based on the RNA 5’ upstream of the crRNA. Cas9 binds the tracrsRNA and needs a DNA binding sequence (5’NGG3’), which is called protospacer adjacent motif (PAM). After binding, Cas9 introduces a DNA double strand break, which is then followed by gene modification via homologous recombination (HDR) or non-homologous end joining (NHEJ).

Gene editing has been a potential therapy for many genetic diseases. Targeted genome editing using nucleases provides a general method for inducing deletions or insertion. An earlier method for targeting relies on protein-DNA interactions; however, the most recent one, using CRISPR – associated protein 9 (Cas9), provides better specificity, simplicity, speed and pricing. The CRISPR System was first identified in single cell archaea (Prokaryotes). It is now widely used in different cell types and organisms including human cells (HEK293T, HeLa, iPSC), mouse, fruit fly, rice, wheat etc. CRISPR –Cas9 genome editing has potential applications in human gene therapy, screening drug targets, synthetic biology, agriculture, programmable RNA targeting, and viral gene disruption.

Other technologies employ antisense, small interfering RNA and other DNA. To the extent that these technologies do not alter DNA, but instead directly interact with molecules such as RNA, they are not considered "gene therapy" per se.[citation needed]

Cell types

Gene therapy may be classified into two types:

Somatic cell

In somatic cell gene therapy (SCGT), the therapeutic genes are transferred into any of any cell other than a gamete, germ cell, gametocyte or undifferentiated stem cell. Any such modifications affect the individual patient only, and are not inherited by offspring. Somatic gene therapy represents mainstream basic and clinical research, in which therapeutic DNA (either integrated in the genome or as an external episome or plasmid) is used to treat disease.

Over 600 clinical trials utilizing SCGT are underway in the US. Most focus on severe genetic disorders, including immunodeficiencies, haemophilia, thalassaemia and cystic fibrosis. Such single gene disorders are good candidates for somatic cell therapy. The complete correction of a genetic disorder or the replacement of multiple genes is not yet possible. Only a few of the trials are in the advanced stages.[36]

Germline

In germline gene therapy (GGT), germ cells (sperm or eggs) are modified by the introduction of functional genes into their genomes. Modifying a germ cell causes all the organism's cells to contain the modified gene. The change is therefore heritable and passed on to later generations. Australia, Canada, Germany, Israel, Switzerland and the Netherlands[37] prohibit GGT for application in human beings, for technical and ethical reasons, including insufficient knowledge about possible risks to future generations[37] and higher risks versus SCGT.[38] The US has no federal controls specifically addressing human genetic modification (beyond FDA regulations for therapies in general).[37][39][40][41]

Vectors

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

The delivery of DNA into cells can be accomplished by multiple methods. The two major classes are recombinant viruses (sometimes called biological nanoparticles or viral vectors) and naked DNA or DNA complexes (non-viral methods).

Viruses

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

In order to replicate, viruses introduce their genetic material into the host cell, tricking the host's cellular machinery into using it as blueprints for viral proteins. Scientists exploit this by substituting a virus's genetic material with therapeutic DNA. (The term 'DNA' may be an oversimplification, as some viruses contain RNA, and gene therapy could take this form as well.) A number of viruses have been used for human gene therapy, including retrovirus, adenovirus, lentivirus, herpes simplex, vaccinia and adeno-associated virus.[3] Like the genetic material (DNA or RNA) in viruses, therapeutic DNA can be designed to simply serve as a temporary blueprint that is degraded naturally or (at least theoretically) to enter the host's genome, becoming a permanent part of the host's DNA in infected cells.

Non-viral

Non-viral methods present certain advantages over viral methods, such as large scale production and low host immunogenicity. However, non-viral methods initially produced lower levels of transfection and gene expression, and thus lower therapeutic efficacy. Later technology remedied this deficiency[citation needed].

Methods for non-viral gene therapy include the injection of naked DNA, electroporation, the gene gun, sonoporation, magnetofection, the use of oligonucleotides, lipoplexes, dendrimers, and inorganic nanoparticles.

Hurdles

Some of the unsolved problems include:

  • Short-lived nature – Before gene therapy can become a permanent cure for a condition, the therapeutic DNA introduced into target cells must remain functional and the cells containing the therapeutic DNA must be stable. Problems with integrating therapeutic DNA into the genome and the rapidly dividing nature of many cells prevent it from achieving long-term benefits. Patients require multiple treatments.
  • Immune response – Any time a foreign object is introduced into human tissues, the immune system is stimulated to attack the invader. Stimulating the immune system in a way that reduces gene therapy effectiveness is possible. The immune system's enhanced response to viruses that it has seen before reduces the effectiveness to repeated treatments.
  • Problems with viral vectors – Viral vectors carry the risks of toxicity, inflammatory responses, and gene control and targeting issues.
  • Multigene disorders – Some commonly occurring disorders, such as heart disease, high blood pressure, Alzheimer's disease, arthritis, and diabetes, are affected by variations in multiple genes, which complicate gene therapy.
  • Some therapies may breach the Weismann barrier (between soma and germ-line) protecting the testes, potentially modifying the germline, falling afoul of regulations in countries that prohibit the latter practice.[42]
  • Insertional mutagenesis – If the DNA is integrated in a sensitive spot in the genome, for example in a tumor suppressor gene, the therapy could induce a tumor. This has occurred in clinical trials for X-linked severe combined immunodeficiency (X-SCID) patients, in which hematopoietic stem cells were transduced with a corrective transgene using a retrovirus, and this led to the development of T cell leukemia in 3 of 20 patients.[43][44] One possible solution is to add a functional tumor suppressor gene to the DNA to be integrated. This may be problematic since the longer the DNA is, the harder it is to integrate into cell genomes. CRISPR technology allows researchers to make much more precise genome changes at exact locations.[45]
  • Cost – Alipogene tiparvovec or Glybera, for example, at a cost of $1.6 million per patient, was reported in 2013 to be the world's most expensive drug.[46][47]

Deaths

Three patients' deaths have been reported in gene therapy trials, putting the field under close scrutiny. The first was that of Jesse Gelsinger in 1999.[48] One X-SCID patient died of leukemia in 2003.[10] In 2007, a rheumatoid arthritis patient died from an infection; the subsequent investigation concluded that the death was not related to gene therapy.[49]

History

1970s and earlier

In 1972 Friedmann and Roblin authored a paper in Science titled "Gene therapy for human genetic disease?"[50] Rogers (1970) was cited for proposing that exogenous good DNA be used to replace the defective DNA in those who suffer from genetic defects.[51]

1980s

In 1984 a retrovirus vector system was designed that could efficiently insert foreign genes into mammalian chromosomes.[52]

1990s

The first approved gene therapy in the US took place on 14 September 1990, at the National Institutes of Health (NIH), under the direction of William French Anderson.[53] Four-year-old Ashanti DeSilva received treatment for a genetic defect that left her with ADA-SCID, a severe immune system deficiency. The effects were temporary, but successful.[54]

Cancer gene therapy was introduced in 1992/93.[55] The treatment of glioblastoma multiforme, the malignant brain tumor whose outcome is always fatal, was done using a vector expressing antisense IGF-I RNA (clinical trial approved by NIH n˚ 1602, and FDA in 1994). The therapy proved to be effective due to the anti-tumor mechanism of IGF-I antisense, which is related to strong immune and apoptotic phenomena. For this reason this strategy can be considered also as immunotherapy.[56]

In 1992 Claudio Bordignon, working at the Vita-Salute San Raffaele University, performed the first gene therapy procedure using hematopoietic stem cells as vectors to deliver genes intended to correct hereditary diseases.[57] In 2002 this work led to the publication of the first successful gene therapy treatment for adenosine deaminase-deficiency (SCID). The success of a multi-center trial for treating children with SCID (severe combined immune deficiency or "bubble boy" disease) from 2000 and 2002, was questioned when two of the ten children treated at the trial's Paris center developed a leukemia-like condition. Clinical trials were halted temporarily in 2002, but resumed after regulatory review of the protocol in the US, the United Kingdom, France, Italy and Germany.[58]

In 1993 Andrew Gobea was born with SCID following prenatal genetic screening. Blood was removed from his mother's placenta and umbilical cord immediately after birth, to acquire stem cells. The allele that codes for adenosine deaminase (ADA) was obtained and inserted into a retrovirus. Retroviruses and stem cells were mixed, after which the viruses inserted the gene into the stem cell chromosomes. Stem cells containing the working ADA gene were injected into Andrew's blood. Injections of the ADA enzyme were also given weekly. For four years T cells (white blood cells), produced by stem cells, made ADA enzymes using the ADA gene. After four years more treatment was needed.[citation needed]

Jesse Gelsinger's death in 1999 impeded gene therapy research in the US.[59][60] As a result, the FDA suspended several clinical trials pending the reevaluation of ethical and procedural practices.[61]

2000s

The modified cancer gene therapy strategy of antisense IGF-I RNA (NIH n˚ 1602)[62] using antisense / triple helix anti IGF-I approach was registered in 2002 by Wiley gene therapy clinical trial - n˚ 635 and 636. The approach has shown promising results in the treatment of six different malignant tumors: glioblastoma, cancers of liver, colon, prostate, uterus and ovary (Collaborative NATO Science Programme on Gene Therapy USA, France, Poland n˚ LST 980517 conducted by J. Trojan) (Trojan et al., 2012). This anti–gene antisense/triple helix therapy has proven to be efficient, due to the mechanism stopping simultaneously IGF-I expression on translation and transcription levels, strengthening anti-tumor immune and apoptotic phenomenons (Trojan et al., 2013).[63]

2002

Sickle-cell disease can be treated in mice.[64] The mice – which have essentially the same defect that causes human cases – used a viral vector to induce production of fetal hemoglobin (HbF), which normally ceases to be produced shortly after birth. In humans, the use of hydroxyurea to stimulate the production of HbF temporarily alleviates sickle cell symptoms. The researchers demonstrated this treatment to be a more permanent means to increase therapeutic HbF production.[65]

A new gene therapy approach repaired errors in messenger RNA derived from defective genes. This technique has the potential to treat thalassaemia, cystic fibrosis and some cancers.[66]

Researchers created liposomes 25 nanometers across that can carry therapeutic DNA through pores in the nuclear membrane.[67]

2003

In 2003 a research team inserted genes into the brain for the first time. They used liposomes coated in a polymer called polyethylene glycol, which, unlike viral vectors, are small enough to cross the blood–brain barrier.[68]

Short pieces of double-stranded RNA (short, interfering RNAs or siRNAs) are used by cells to degrade RNA of a particular sequence. If a siRNA is designed to match the RNA copied from a faulty gene, then the abnormal protein product of that gene will not be produced.[69]

Gendicine is a cancer gene therapy that delivers the tumor suppressor gene p53 using an engineered adenovirus. In 2003, it was approved in China for the treatment of head and neck squamous cell carcinoma.[27]

2006

In March researchers announced the successful use of gene therapy to treat two adult patients for X-linked chronic granulomatous disease, a disease which affects myeloid cells and damages the immune system. The study is the first to show that gene therapy can treat the myeloid system.[70]

In May a team reported a way to prevent the immune system from rejecting a newly delivered gene.[71] Similar to organ transplantation, gene therapy has been plagued by this problem. The immune system normally recognizes the new gene as foreign and rejects the cells carrying it. The research utilized a newly uncovered network of genes regulated by molecules known as microRNAs. This natural function selectively obscured their therapeutic gene in immune system cells and protected it from discovery. Mice infected with the gene containing an immune-cell microRNA target sequence did not reject the gene.

In August scientists successfully treated metastatic melanoma in two patients using killer T cells genetically retargeted to attack the cancer cells.[72]

In November researchers reported on the use of VRX496, a gene-based immunotherapy for the treatment of HIV that uses a lentiviral vector to deliver an antisense gene against the HIV envelope. In a phase I clinical trial, five subjects with chronic HIV infection who had failed to respond to at least two antiretroviral regimens were treated. A single intravenous infusion of autologous CD4 T cells genetically modified with VRX496 was well tolerated. All patients had stable or decreased viral load; four of the five patients had stable or increased CD4 T cell counts. All five patients had stable or increased immune response to HIV antigens and other pathogens. This was the first evaluation of a lentiviral vector administered in a US human clinical trial.[73][74]

2007

In May researchers announced the first gene therapy trial for inherited retinal disease. The first operation was carried out on a 23-year-old British male, Robert Johnson, in early 2007.[75]

2008

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

Leber's congenital amaurosis is an inherited blinding disease caused by mutations in the RPE65 gene. The results of a small clinical trial in children were published in April.[14] Delivery of recombinant adeno-associated virus (AAV) carrying RPE65 yielded positive results. In May two more groups reported positive results in independent clinical trials using gene therapy to treat the condition. In all three clinical trials, patients recovered functional vision without apparent side-effects.[14][15][16][17]

2009

In September researchers were able to give trichromatic vision to squirrel monkeys.[76] In November 2009, researchers halted a fatal genetic disorder called adrenoleukodystrophy in two children using a lentivirus vector to deliver a functioning version of ABCD1, the gene that is mutated in the disorder.[77]

2010s

2010

An April paper reported that gene therapy addressed achromatopsia (color blindness) in dogs by targeting cone photoreceptors. Cone function and day vision were restored for at least 33 months in two young specimens. The therapy was less efficient for older dogs.[78]

In September it was announced that an 18-year-old male patient in France with beta-thalassemia major had been successfully treated.[79] Beta-thalassemia major is an inherited blood disease in which beta haemoglobin is missing and patients are dependent on regular lifelong blood transfusions.[80] The technique used a lentiviral vector to transduce the human ß-globin gene into purified blood and marrow cells obtained from the patient in June 2007.[81] The patient's haemoglobin levels were stable at 9 to 10 g/dL. About a third of the hemoglobin contained the form introduced by the viral vector and blood transfusions were not needed.[81][82] Further clinical trials were planned.[83] Bone marrow transplants are the only cure for thalassemia, but 75% of patients do not find a matching donor.[82]

2011

In 2007 and 2008, a man was cured of HIV by repeated Hematopoietic stem cell transplantation (see also Allogeneic stem cell transplantation, Allogeneic bone marrow transplantation, Allotransplantation) with double-delta-32 mutation which disables the CCR5 receptor. This cure was accepted by the medical community in 2011.[84] It required complete ablation of existing bone marrow, which is very debilitating.

In August two of three subjects of a pilot study were confirmed to have been cured from chronic lymphocytic leukemia (CLL). The therapy used genetically modified T cells to attack cells that expressed the CD19 protein to fight the disease.[22] In 2013, the researchers announced that 26 of 59 patients had achieved complete remission and the original patient had remained tumor-free.[85]

Human HGF plasmid DNA therapy of cardiomyocytes is being examined as a potential treatment for coronary artery disease as well as treatment for the damage that occurs to the heart after myocardial infarction.[86][87]

n 2011 Neovasculgen was registered in Russia as the first-in-class gene-therapy drug for treatment of peripheral artery disease, including critical limb ischemia.[28] Neovasculogen is a plasmid encoding the CMV promoter and the 165 amino acid form of VEGF.[88][89]

2012

The FDA approved Phase 1 clinical trials on thalassemia major patients in the US for 10 participants in July.[90] The study was expected to continue until 2015.[91]

In July 2012, the European Medicines Agency recommended approval of a gene therapy treatment for the first time in either Europe or the United States. The treatment used Alipogene tiparvovec (Glybera) to compensate for lipoprotein lipase deficiency, which can cause severe pancreatitis.[92] The recommendation was endorsed by the European Commission in November 2012[13][29][93][94] and commercial rollout began in late 2014.[95]

In December 2012, it was reported that 10 of 13 patients with multiple myeloma were in remission "or very close to it" three months after being injected with a treatment involving genetically engineered T cells to target proteins NY-ESO-1 and LAGE-1, which exist only on cancerous myeloma cells.[24]

2013

In March researchers reported that three of five subjects who had acute lymphocytic leukemia (ALL) had been in remission for five months to two years after being treated with genetically modified T cells which attacked cells with CD19 genes on their surface, i.e. all B-cells, cancerous or not. The researchers believed that the patients' immune systems would make normal T-cells and B-cells after a couple of months. They were also given bone marrow. One patient relapsed and died and one died of a blood clot unrelated to the disease.[23]

Following encouraging Phase 1 trials, in April, researchers announced they were starting Phase 2 clinical trials (called CUPID2 and SERCA-LVAD) on 250 patients[96] at several hospitals to combat heart disease. The therapy was designed to increase the levels of SERCA2a protein in heart muscles, improving muscle function.[97] The FDA granted this a Breakthrough Therapy Designation to accelerate the trial and approval process.[98]

In July researchers reported promising results for six children with two severe hereditary diseases had been treated with a partially deactivated lentivirus to replace a faulty gene and after 7–32 months. Three of the children had metachromatic leukodystrophy, which causes children to lose cognitive and motor skills.[99] The other children had Wiskott-Aldrich syndrome, which leaves them to open to infection, autoimmune diseases and cancer.[100] Follow up trials with gene therapy on another six children with Wiskott-Aldrich syndrome were also reported as promising.[101][102]

In October researchers reported that two children born with adenosine deaminase severe combined immunodeficiency disease (ADA-SCID) had been treated with genetically engineered stem cells 18 months previously and that their immune systems were showing signs of full recovery. Another three children were making progress.[20] In 2014 a further 18 children with ADA-SCID were cured by gene therapy.[103] ADA-SCID children have no functioning immune system and are sometimes known as "bubble children."[20]

Also in October researchers reported that they had treated six haemophilia sufferers in early 2011 using an adeno-associated virus. Over two years later all six were producing clotting factor.[20][104]

Data from three trials on Topical cystic fibrosis transmembrane conductance regulator gene therapy were reported to not support its clinical use as a mist inhaled into the lungs to treat cystic fibrosis patients with lung infections.[105]

2014

In January researchers reported that six choroideremia patients had been treated with adeno-associated virus with a copy of REP1. Over a six-month to two-year period all had improved their sight. Choroideremia is an inherited genetic eye disease with no approved treatment, leading to loss of sight.[106][107]

In March researchers reported that 12 HIV patients had been treated since 2009 in a trial with a genetically engineered virus with a rare mutation (CCR5 deficiency) known to protect against HIV with promising results.[108][109]

Clinical trials of gene therapy for sickle cell disease were started in 2014[110][111] although one review failed to find any such trials.[112]

2015

In February LentiGlobin BB305, a gene therapy treatment undergoing clinical trials for treatment of beta thalassemia gained FDA "breakthrough" status after several patients were able to forgo the frequent blood transfusions usually required to treat the disease.[113]

In March researchers delivered a recombinant gene encoding a broadly neutralizing antibody into monkeys infected with simian HIV; the monkey's cells produced the antibody, which cleared them of HIV. The technique is named immunoprophylaxis by gene transfer (IGT). Animal tests for antibodies to ebola, malaria, influenza and hepatitis are underway.[114][115]

In March scientists, including an inventor of CRISPR, urged a worldwide moratorium on germline gene therapy, writing “scientists should avoid even attempting, in lax jurisdictions, germline genome modification for clinical application in humans” until the full implications “are discussed among scientific and governmental organizations”.[116][117][118][119]

Also in 2015 Glybera was approved for the German market.[120]

In October, researchers announced that they had treated a baby girl, Layla Richards, with an experimental treatment using donor T-cells genetically engineered to attack cancer cells. Two months after the treatment she was still free of her cancer (a highly aggressive form of acute lymphoblastic leukaemia [ALL]). Children with highly aggressive ALL normally have a very poor prognosis and Lalya's disease had been regarded as terminal before the treatment.[121]

In December, scientists of major world academies called for a moratorium on inheritable human genome edits, including those related to CRISPR-Cas9 technologies[122] but that basic research including embryo gene editing should continue.[123]

Speculative uses

Speculated uses for gene therapy include:

Fertility

Gene Therapy techniques have the potential to provide alternative treatments for those with infertility. Recently, successful experimentation on mice has proven that fertility can be restored by using the gene therapy method, CRISPR.[124] Spermatogenical stem cells from another organism were transplanted into the testes of an infertile male mouse. The stem cells re-established spermatogenesis and fertility.[125] Gene Therapy research on fertility is promising, and scientists anticipate that this method can be further utilized to help humans in the near future.

Life extension

Aging has much to do with the wearing out of telomeres in the DNA. Biogerontologist Aubrey De Grey of the Methuselah Foundation and SENS Research Foundation has proposed a gene therapy and stem cell research program called Strategies for Engineered Negligible Senescence, or SENS for short.[126]

In September 15, 2015, Elizabeth Parrish became the first person to undergo anti-aging gene therapy - almost exactly 25 years after the first authorized and successful use of gene therapy.[127]

Gene doping

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

Athletes might adopt gene therapy technologies to improve their performance.[128] Gene doping is not known to occur, but multiple gene therapies may have such effects. Kayser et al. argue that gene doping could level the playing field if all athletes receive equal access. Critics claim that any therapeutic intervention for non-therapeutic/enhancement purposes compromises the ethical foundations of medicine and sports.[129]

Human genetic engineering

<templatestyles src="Module:Hatnote/styles.css"></templatestyles>

Genetic engineering could be used to change physical appearance, metabolism, and even improve physical capabilities and mental faculties such as memory and intelligence. Ethical claims about germline engineering include beliefs that every fetus has a right to remain genetically unmodified, that parents hold such rights, and that every child has the right to be born free of preventable diseases.[130][131][132] For adults, genetic engineering could be seen as another enhancement technique to add to diet, exercise, education, cosmetics and plastic surgery.[133][134] Another theorist claims that moral concerns limit but do not prohibit germline engineering.[135]

Possible regulatory schemes include a complete ban, provision to everyone, or professional self-regulation. The American Medical Association’s Council on Ethical and Judicial Affairs stated that "genetic interventions to enhance traits should be considered permissible only in severely restricted situations: (1) clear and meaningful benefits to the fetus or child; (2) no trade-off with other characteristics or traits; and (3) equal access to the genetic technology, irrespective of income or other socioeconomic characteristics."[136]

As early in the history of biotechnology as 1990, the scientific community was opposed to attempts to modify the human germline using these new tools,[137] and such cautions continued as technology progressed.[138] With the advent of new techniques like CRISPR, in March 2015 scientists urged a worldwide ban on clinical use of gene editing technologies to edit the human genome in a way that can be inherited.[116][117][118][119] In April 2015, researchers sparked controversy when they reported results of basic research to edit the DNA of non-viable human embryos using CRISPR.[124][139] Additionally, it has been suggested that 3D printing can be implemented in the rapid development of "bioresorbable scaffolds" capable of implanting stem cells.[140]

Regulations

Regulations covering genetic modification are part of general guidelines about human-involved biomedical research.

The Helsinki Declaration (Ethical Principles for Medical Research Involving Human Subjects) was amended by the World Medical Association's General Assembly in 2008. This document provides principles physicians and researchers must consider when involving humans as research subjects. The Statement on Gene Therapy Research initiated by the Human Genome Organization (HUGO) in 2001 provides a legal baseline for all countries. HUGO’s document emphasizes human freedom and adherence to human rights, and offers recommendations for somatic gene therapy, including the importance of recognizing public concerns about such research.[141]

United States

No federal legislation lays out protocols or restrictions about human genetic engineering. This subject is governed by overlapping regulations from local and federal agencies, including the Department of Health and Human Services, the FDA and NIH's Recombinant DNA Advisory Committee. Researchers seeking federal funds for an investigational new drug application, (commonly the case for somatic human genetic engineering), must obey international and federal guidelines for the protection of human subjects.[142]

NIH serves as the main gene therapy regulator for federally funded research. Privately funded research is advised to follow these regulations. NIH provides funding for research that develops or enhances genetic engineering techniques and to evaluate the ethics and quality in current research. The NIH maintains a mandatory registry of human genetic engineering research protocols that includes all federally funded projects.

An NIH advisory committee published a set of guidelines on gene manipulation.[143] The guidelines discuss lab safety as well as human test subjects and various experimental types that involve genetic changes. Several sections specifically pertain to human genetic engineering, including Section III-C-1. This section describes required review processes and other aspects when seeking approval to begin clinical research involving genetic transfer into a human patient.[144]

The FDA regulates the quality and safety of gene therapy products and supervises how these products are used clinically. Therapeutic alteration of the human genome falls under the same regulatory requirements as any other medical treatment. Research involving human subjects, such as clinical trials, must be reviewed and approved by the FDA and an Institutional Review Board.[145][146]

Popular culture

Gene therapy is the basis for the plotline of the film I Am Legend[147] and the TV show Will Gene Therapy Change the Human Race?[148]

See also

References

  1. http://vaccine-safety-training.org/live-attenuated-vaccines.html
  2. Lua error in package.lua at line 80: module 'strict' not found.
  3. 3.0 3.1 Gene Therapy Clinical Trials Worldwide Database. The Journal of Gene Medicine. Wiley (January 2014)
  4. http://archive.hhs.gov/news/press/1995pres/951019a.html
  5. http://www.nytimes.com/2013/09/17/science/dna-double-take.html?pagewanted=all&_r=1
  6. https://books.google.com/books?id=FxGjBqEL-3kC&pg=PA45&lpg=PA45&dq=%22martin+cline%22+%22gene+therapy%22&source=bl&ots=Goj5oCAVBp&sig=KbB9EHE4ucGkS1C2AUVltD-A4-U&hl=en&sa=X&ved=0CD8Q6AEwBWoVChMIyczEkPrRyAIVTcNjCh1NBg3U#v=onepage&q=%22martin%20cline%22%20%22gene%20therapy%22&f=false
  7. Lua error in package.lua at line 80: module 'strict' not found.
  8. https://books.google.com/books?id=FzuwRwb--X4C&pg=PA30&lpg=PA30&dq=%22gene+therapy%22+martin+cline&source=bl&ots=L0oOWFnD-i&sig=k751o-g-TgVgJnECRIRMcM4EcFU&hl=en&sa=X&ved=0CDQQ6AEwBjgKahUKEwja2rCXtvfIAhVP5GMKHVPWDWQ#v=onepage&q=%22gene%20therapy%22%20martin%20cline&f=false
  9. http://cshmonographs.org/index.php/monographs/article/viewFile/4773/3874
  10. 10.0 10.1 Lua error in package.lua at line 80: module 'strict' not found.
  11. http://www.asgct.org/UserFiles/kohnslides.pdf
  12. http://www.docguide.com/healthy-baby-born-after-worlds-first-successful-cytoplasmic-transfer
  13. 13.0 13.1 13.2 Lua error in package.lua at line 80: module 'strict' not found.
  14. 14.0 14.1 14.2 Lua error in package.lua at line 80: module 'strict' not found.
  15. 15.0 15.1 Lua error in package.lua at line 80: module 'strict' not found.
  16. 16.0 16.1 Lua error in package.lua at line 80: module 'strict' not found.
  17. 17.0 17.1 Lua error in package.lua at line 80: module 'strict' not found.
  18. Lua error in package.lua at line 80: module 'strict' not found.
  19. Lua error in package.lua at line 80: module 'strict' not found.
  20. 20.0 20.1 20.2 20.3 20.4 Geddes, Linda (30 October 2013) 'Bubble kid' success puts gene therapy back on track' The New Scientist. Retrieved 2 November 2013
  21. Lua error in package.lua at line 80: module 'strict' not found.
  22. 22.0 22.1 Lua error in package.lua at line 80: module 'strict' not found.
  23. 23.0 23.1 Coghlan, Andy (26 March 2013) Gene therapy cures leukaemia in eight days. The New Scientist. Retrieved 15 April 2013
  24. 24.0 24.1 Coghlan, Andy (11 December 2013) Souped-up immune cells force leukaemia into remission. New Scientist. Retrieved 15 April 2013
  25. Lua error in package.lua at line 80: module 'strict' not found.
  26. Herper, Matthew (26 March 2014) Gene Therapy's Big Comeback Forbes. Retrieved 28 April 2014
  27. 27.0 27.1 Lua error in package.lua at line 80: module 'strict' not found.
  28. 28.0 28.1 Lua error in package.lua at line 80: module 'strict' not found.
  29. 29.0 29.1 29.2 Gallagher, James. (2 November 2012) BBC News – Gene therapy: Glybera approved by European Commission. BBC. Retrieved 15 December 2012.
  30. U.S. National Library of Medicine, Genomics Home Reference. What is gene therapy?
  31. U.S. National Library of Medicine, Genomics Home Reference. How does gene therapy work?
  32. Lua error in package.lua at line 80: module 'strict' not found.
  33. Lua error in package.lua at line 80: module 'strict' not found.
  34. Lua error in package.lua at line 80: module 'strict' not found.
  35. Lua error in package.lua at line 80: module 'strict' not found.
  36. Lua error in package.lua at line 80: module 'strict' not found.
  37. 37.0 37.1 37.2 Lua error in package.lua at line 80: module 'strict' not found.
  38. Strachnan, T. and Read, A. P. (2004) Human Molecular Genetics, 3rd Edition, Garland Publishing, p. 616, ISBN 0815341849.
  39. Hanna, K., 2006, Germline Gene Transfer, National Human Genome Research Institute, [1]
  40. 2013, Human Cloning and Genetic Modification, Association of Reproductive Health Officials, [2]
  41. Lua error in package.lua at line 80: module 'strict' not found.
  42. Lua error in package.lua at line 80: module 'strict' not found.
  43. Lua error in package.lua at line 80: module 'strict' not found.
  44. Lua error in package.lua at line 80: module 'strict' not found.
  45. Young, Susan (11 February 2014) Genome Surgery MIT Technology Review. Retrieved 17 February 2014
  46. (31 October 2013) Gene therapy needs a hero to live up to the hype The New Scientist. Retrieved 2 November 2012
  47. Crasto, Anthony Melvin (2013) Glybera – The Most Expensive Drug in the world & First Approved Gene Therapy in the West All About Drug. Retrieved 2 November 2013
  48. ORNL.gov. ORNL.gov. Retrieved 15 December 2012.
  49. Lua error in package.lua at line 80: module 'strict' not found.
  50. Lua error in package.lua at line 80: module 'strict' not found.
  51. Rogers S, New Scientist 1970, p. 194
  52. Lua error in package.lua at line 80: module 'strict' not found.
  53. Lua error in package.lua at line 80: module 'strict' not found.
  54. Lua error in package.lua at line 80: module 'strict' not found.
  55. Lua error in package.lua at line 80: module 'strict' not found.
  56. Lua error in package.lua at line 80: module 'strict' not found.
  57. Lua error in package.lua at line 80: module 'strict' not found.
  58. Lua error in package.lua at line 80: module 'strict' not found.
  59. Lua error in package.lua at line 80: module 'strict' not found.
  60. Lua error in package.lua at line 80: module 'strict' not found.
  61. Lua error in package.lua at line 80: module 'strict' not found.
  62. Trojan J, Pan YX, Wei MX, LyA, Shevelev A, Bierwagen M, Ardourel M-Y, Trojan L, Alvarez A, Andres C, Noguera MC, Briceño I, Aristizabal BH, Kasprzak H, Duc HT, Anthony DD. Methodology for anti - gene anti - IGF-I therapy of malignant tumours. Chemother. Res. Pract., doi:10.1155/2012/721873; 2012
  63. Trojan J. and Briceno I. IGF-I Antisense and Triple-Helix Gene Therapy of Glioblastoma In: A. Pantar "Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications"
  64. Lua error in package.lua at line 80: module 'strict' not found.
  65. Lua error in package.lua at line 80: module 'strict' not found.
  66. Lua error in package.lua at line 80: module 'strict' not found.
  67. Lua error in package.lua at line 80: module 'strict' not found.
  68. Lua error in package.lua at line 80: module 'strict' not found.
  69. Lua error in package.lua at line 80: module 'strict' not found.
  70. Lua error in package.lua at line 80: module 'strict' not found.
  71. Lua error in package.lua at line 80: module 'strict' not found.
  72. Lua error in package.lua at line 80: module 'strict' not found.
  73. Lua error in package.lua at line 80: module 'strict' not found.
  74. Lua error in package.lua at line 80: module 'strict' not found.
  75. Lua error in package.lua at line 80: module 'strict' not found.
  76. Lua error in package.lua at line 80: module 'strict' not found.
  77. Lua error in package.lua at line 80: module 'strict' not found.
  78. Lua error in package.lua at line 80: module 'strict' not found.
  79. Lua error in package.lua at line 80: module 'strict' not found.
  80. Lua error in package.lua at line 80: module 'strict' not found.
  81. 81.0 81.1 Beals, Jacquelyn K. (16 September 2010). Gene Therapy Frees Beta-Thalassemia Patient From Transfusions for 2+ Years. Medscape.com (16 September 2010). Retrieved 15 December 2012.
  82. 82.0 82.1 Lua error in package.lua at line 80: module 'strict' not found.
  83. (11 July 2012) ß-Thalassemia Major With Autologous CD34+ Hematopoietic Progenitor Cells Transduced With TNS9.3.55 a Lentiviral Vector Encoding the Normal Human ß-Globin Gene ClinicalTrials.gov, Clinical trial NCT01639690 at the Memorial Sloan-Kettering Cancer Center. Retrieved 12 February 2014
  84. Rosenberg, Tina (29 May 2011) The Man Who Had HIV and Now Does Not, New York.
  85. Lua error in package.lua at line 80: module 'strict' not found.
  86. Lua error in package.lua at line 80: module 'strict' not found.
  87. Lua error in package.lua at line 80: module 'strict' not found.
  88. Eurolab. Neovasculogen listing in Eurolab Page accessed August 4, 2015
  89. Lua error in package.lua at line 80: module 'strict' not found.
  90. On Cancer: Launch of Stem Cell Therapy Trial Offers Hope for Patients with Inherited Blood Disorder | Memorial Sloan-Kettering Cancer Center. Mskcc.org (16 July 2012). Retrieved 15 December 2012.
  91. (4 September 2014) ß-Thalassemia Major With Autologous CD34+ Hematopoietic Progenitor Cells Transduced With TNS9.3.55 a Lentiviral Vector Encoding the Normal Human ß-Globin Gene ClinicalTrials.gov, US National Institutes of Health. Retrieved 17 December 2014.
  92. Pollack, Andrew (20 July 2012) European Agency Backs Approval of a Gene Therapy, The New York Times.
  93. First Gene Therapy Approved by European Commission. UniQure (2 November 2012). Retrieved 15 December 2012.
  94. Lua error in package.lua at line 80: module 'strict' not found.
  95. Lua error in package.lua at line 80: module 'strict' not found.
  96. Bosely, Sarah (30 April 2013) Pioneering gene therapy trials offer hope for heart patients The Guardian. Retrieved 28 April 2014
  97. First gene therapy trial for heart failure begins in UK. The Physicians Clinic (8 September 2013) Archived April 29, 2014 at the Wayback Machine
  98. Celladon Receives Breakthrough Therapy Designation From FDA for MYDICAR(R), Novel, First-in-Class Therapy in Development to Treat Heart Failure. New York Times (10 April 2014)
  99. Lua error in package.lua at line 80: module 'strict' not found.
  100. Lua error in package.lua at line 80: module 'strict' not found.
  101. Gallagher, James (21 April 2015) Gene therapy: 'Tame HIV' used to cure disease BBC News, Health, Retrieved 21 April 2015
  102. Lua error in package.lua at line 80: module 'strict' not found.
  103. Gene therapy cure for children with 'bubble baby' disease Science Daily (18 November 2014)
  104. Gene therapy provides safe, long-term relief for patients with severe hemophilia B Science Daily (20 November 2014)
  105. Lua error in package.lua at line 80: module 'strict' not found.
  106. Lua error in package.lua at line 80: module 'strict' not found.
  107. Beali, Abigail (25 January 2014) Gene therapy restores sight in people with eye disease The New Scientist. Retrieved 25 January 2014
  108. Lua error in package.lua at line 80: module 'strict' not found.
  109. Dvorsky, George (6 March 2014) Scientists Create Genetically Modified Cells That Protect Against HIV io9, Biotechnology. Retrieved 6 March 2014
  110. (15 December 2014) Stem Cell Gene Therapy for Sickle Cell Disease, ClinicalTrials.gov Identifier: NCT02247843 ClinicalTrials.gov, U.S. National Institutes of Health. Retrieved 17 December 2014
  111. Collection and Storage of Umbilical Cord Stem Cells for Treatment of Sickle Cell Disease; ClinicalTrials.gov Identifier: NCT00012545 ClinicalTrials.gov, U.S. National Institutes of Health (15 December 2014)
  112. Lua error in package.lua at line 80: module 'strict' not found.
  113. Lua error in package.lua at line 80: module 'strict' not found.
  114. Lua error in package.lua at line 80: module 'strict' not found.
  115. Lua error in package.lua at line 80: module 'strict' not found.
  116. 116.0 116.1 Lua error in package.lua at line 80: module 'strict' not found.
  117. 117.0 117.1 Lua error in package.lua at line 80: module 'strict' not found.
  118. 118.0 118.1 Lua error in package.lua at line 80: module 'strict' not found.
  119. 119.0 119.1 Lua error in package.lua at line 80: module 'strict' not found.
  120. Die 1-Million-Euro-Spritze (4 April 2015)
  121. Lua error in package.lua at line 80: module 'strict' not found.
  122. Lua error in package.lua at line 80: module 'strict' not found.
  123. Lua error in package.lua at line 80: module 'strict' not found.
  124. 124.0 124.1 Lua error in package.lua at line 80: module 'strict' not found.
  125. Lua error in package.lua at line 80: module 'strict' not found.
  126. http://www.lifeextension.com/magazine/2006/2/profile/page-01
  127. http://www.technologyreview.com/news/542371/a-tale-of-do-it-yourself-gene-therapy/
  128. Lua error in package.lua at line 80: module 'strict' not found.
  129. Lua error in package.lua at line 80: module 'strict' not found.
  130. Lua error in package.lua at line 80: module 'strict' not found.
  131. Lua error in package.lua at line 80: module 'strict' not found.
  132. Lua error in package.lua at line 80: module 'strict' not found.
  133. Gene Therapy and Genetic Engineering. The Center for Health Ethics, University of Missouri School of Medicine. 25 April 2013.
  134. Lua error in package.lua at line 80: module 'strict' not found.
  135. Lua error in package.lua at line 80: module 'strict' not found.
  136. AMA Council on Ethical and Judicial Affairs, Report on Ethical Issues Related to Prenatal Genetic Tests, 3 Archives Fam. Med. 633, 637–39 (1994), Archived September 28, 2011 at the Wayback Machine
  137. The Declaration of Inuyama: Human Genome Mapping, Genetic Screening and Gene Therapy
  138. Smith KR, Chan S, Harris J. Human germline genetic modification: scientific and bioethical perspectives. Arch Med Res. 2012 Oct;43(7):491-513. doi: 10.1016/j.arcmed.2012.09.003. PMID 23072719
  139. Lua error in package.lua at line 80: module 'strict' not found.
  140. Lua error in package.lua at line 80: module 'strict' not found.
  141. Human Genome Organization. HUGO Ethics Committee. Statement on Gene Therapy Research. April 2001.
  142. Isasi, R. M. et al. (October 2006) National Regulatory Frameworks Regarding Human Genetic Modification Technologies (Somatic and Germline Modification). Genetics & Public Policy Center.
  143. National Institutes of Health. NIH Guidelines for Research Involving Recombinant or Synthetic Nucleic Acid Molecules. Revised March 2013.
  144. U.S. Department of Health & Human Services. The National Commission for the Protection of Human Subjects of Biomedical and Behavioral Research. The Belmont Report: Ethical Principles and Guidelines for the Protection of Human Subjects of Research. 18 April 1979.
  145. Lua error in package.lua at line 80: module 'strict' not found.
  146. U.S. Department of Health and Human Services. Food and Drug Administration. Center for Biologics Evaluation and Research. Guidance for Industry: Guidance for Human Somatic Cell Therapy and Gene Therapy. March 1998.
  147. Lua error in package.lua at line 80: module 'strict' not found.
  148. Gene Therapy Change the Human Race?/Lua error in Module:WikidataCheck at line 28: attempt to index field 'wikibase' (a nil value). Gene therapy at IMDb

Further reading

  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.
  • Lua error in package.lua at line 80: module 'strict' not found.

External links