Gene therapy

from Wikipedia, the free encyclopedia

As gene therapy refers to the insertion of nucleic acids such as DNA or RNA in the body cells of a subject to treat a disease for example. Classically, an intact gene should be inserted into the genome of the target cell in order to replace a defective gene that is the cause of the disease. In humans, gene therapies have been and are partially successful z. B. carried out in the context of clinical studies. Gendicine ( rAD-p53 ) was introduced in China in 2003 as the world's first finished medicinal product .

Within Europe, gene therapeutics belong to the group of advanced therapy drugs (ATMPs). An overview of the gene therapeutics approved in the USA can be found on the website of the Food and Drug Administration (FDA) .

principle

Usually some cells are removed from the body in order to insert the corresponding nucleic acids in the laboratory ( ex vivo ) . The cells can then be multiplied, for example, and then reintroduced into the body. Gene therapy can also take place directly in the body ( in vivo ). Depending on the type of gene therapy and the technology used, the nucleic acid can be integrated into the cell genome or only remain in the cell temporarily. Accordingly, the therapeutic effect can be permanent or temporary.

A distinction must be made between the following mechanisms:

  • Gene replacement therapy: Typical areas of application are recessive diseases. Affected patients have two defective copies of a certain gene, which leads to the outbreak of disease. In gene replacement therapy, the therapeutic nucleic acid codes for that very gene, so that treated cells can have and use an intact copy of this gene after therapy.
  • Gene editing, e.g. E.g. using Crispr-Cas9 , also known as genome surgery: The aim of this approach is to correct defects in the genome of a cell. This approach can potentially also be used to treat dominant diseases.
  • Prosthetic gene therapy: While the two aforementioned methods rely on largely structurally intact target cells, prosthetic gene therapy is intended to enable other cells to take over the function of cells that have died, similar to a prosthesis. An example of this is the "vision restoration" approach to gene therapy.

Methods

There are various methods of transferring a therapeutic nucleic acid into a cell:

  • Transduction : This is the most widely used method, when a viral vector (a modified virus ) brings the therapeutic sequence into the cell.
  • Transfection (chemical): The nucleic acid and an electrically charged compound (e.g. calcium phosphate) are added to the cells. The electrically charged compound binds to the cell membrane and is endocytosed , allowing the vector to enter the cytosol after the endosomal membrane has been perforated .
  • Transfection (physical): During electroporation , a current surge makes the cell membrane temporarily permeable so that the vector can penetrate the cell.
  • Transfection (physical): The microinjection offers high chances for a successful integration of the gene (approx. 1: 5), but each cell has to be treated individually.
  • Sperm-mediated gene transfer in egg cells

Limitations and Risks

The replacement and permanent insertion of an intact gene in the form of DNA has a chance of success only in so-called monogenic diseases. Diseases that are triggered by more complex genetic damage, such as cancer , cannot be treated causally with gene therapy. In Germany and some other countries, gene therapy may only be carried out in the somatic (not affecting the germline ) cells so that the new genetic information cannot be passed on to the children of the person being treated. This legal restriction is based on ethical and safety aspects (see also germ line therapy ). The greatest possible risk of somatic gene therapy is an undirected integration of the donor DNA at an inappropriate location within the genome of the host cell. Since the point of integration cannot yet be foreseen, other previously intact genes can be disturbed in their function. In the worst case, the therapeutic benefit of the new gene could be nullified by a new, possibly more serious disease caused by the disruption of a previously intact gene.

In practice, gene therapy approaches are currently limited to two different cell types : accessible stem cells and long-lived, differentiated, postmitotic cells. Depending on the cell type, different methods of gene therapy are used.

Somatic cells that can be used as vectors for gene therapy with retroviruses must meet certain requirements:

  • They have to be tough enough to survive the "infection", but especially the removal from and re-implantation in the body
  • They must be easily removable and reusable
  • They should be long-lived so that they can produce the new protein for a long time

The following cell types have proven to be suitable:

  • Skin cells: fibroblasts from the dermis (out of date)
  • Liver cells
  • T cells: T lymphocytes (circulating white blood cells) are responsible for the cellular immune response. The lack of the gene for adenosine deaminase (ADA), which leads to a “ severe combined immunodeficiency ” (SCID), is treated by treating these cells accordingly. Another possible therapy is a defect in the common chain of some interleukin receptors, X-SCID .
  • Bone marrow stem cells: They produce the red and white blood cells. Genetic diseases of the blood and the immune system can be treated by gene therapy of the rare stem cells. So z. For example, the beta-thalassemia (a lack of β-globin leads to anemia ) by incorporating an "enhancer sequence" treat stem cells (s. U .: Zynteglo).

Applications on the patient

The Gene Therapy Clinical Trials Worldwide database provided by The Journal of Gene Medicine lists over 1500 clinical studies that have been approved to date (as of 2010).

Purification and molecular characterization of cytokines

The purification and molecular characterization of cytokines was researched by Roland Mertelsmann's team . Both interleukin-2 and G-CSF were displayed homogeneously, first translational and clinical studies on cytokines followed. Since experiments in murine models demonstrated that the local secretion of immunity-stimulating cytokines such as interleukin-2 led to the strong activation of the immune system with therapeutic effectiveness, this approach was also followed in humans. These investigations provided one of the foundations for the later, clinically successful gene therapy strategies.

Therapy of SCID

On September 14, 1990, doctors from the US Federal Institute of Health performed the world's first gene therapy treatment on a four-year-old girl. Ashanti DeSilva suffered from severe combined immunodeficiency (SCID), a very rare disease ( incidence 1: 100,000) caused by a severe defect in both the T and B lymphocyte systems . In patients affected by this defect, the function of the immune system is considerably or completely impaired, i. E. In other words, there is little or no immune response - even a cold can kill children. The gene therapy, which has to be repeated several times a year due to the limited lifespan of the leukocytes , enables patients to live without strict quarantine . The gene therapy at Ashanti DeSilva was preceded by a three-year approval process.

The disease X-SCID , which is identical in terms of symptoms and occurs due to mutations in the common chain of some interleukin receptors (γc, CD132 ), was also treated with a gene therapy approach by Alain Fischer in Paris. After the treatment was initially largely successful, leukemia developed in some patients after a while (for details see X-SCID).

Jesse Gelsinger case

In 1999 gene therapy research suffered a setback. Serious complications arose in a series of experiments conducted by the University of Pennsylvania and directed by James M. Wilson.

18-year-old Jesse Gelsinger suffered from a congenital ornithine transcarbamylase deficit . He took part in the last of six test stages as a test subject. The dose of the adenovirus used as a carrier injected into him on September 13 was 38 trillion particles. That is significantly more than is transmitted in natural infections. His condition deteriorated rapidly, so that he died of multiple organ failure on September 17th . At that time, Jesse Gelsinger was the sixth officially reported death worldwide from an artificial adenovirus infection used in gene therapy experiments. In the six cases, according to the director of the experiment, the underlying disease was the cause of death.

The US health authorities then prohibited senior physician James M. Wilson from carrying out any research on humans, among other things because he deliberately violated predetermined requirements. Gelsinger's state of health was stable before the experiment, but his liver values ​​exceeded the maximum limit set by the authorities.

Therapy of HIV-1 infection

By partially removing the co-receptor CCR5 from the genome of CD4-positive T cells using adoptive cell transfer and genome editing , the decrease in the concentration of CD4-positive T cells in the event of an HIV infection can be slowed down.

The end of Glybera

In October 2012, Glybera ( Alipogentiparvovec ) was the first gene therapeutic agent in the western world to receive approval for the treatment of the rare condition of familial lipoprotein lipase deficiency (LPLD) in adults. In November 2015, the National Association of Statutory Health Insurance Funds and Chiesi GmbH, as the licensee of the Dutch company uniQure, agreed on a reimbursement amount for the medicine. In the disease, which is usually diagnosed in childhood, the body does not produce the enzyme lipoprotein lipase. Because fat particles build up in the blood, it looks white. Patients have to abstain from alcohol and sugar for their entire life and suffer from pain caused by inflammation of the pancreas. In April 2017, uniQure announced that it would not extend the approval in the EU. The approval ended on October 25, 2017.

Glybera had been unsuccessfully applied for in the USA . In order to help patients nonetheless, attempts by individual independent biologists began to make the therapy available for around 7,000 instead of 1,000,000 US dollars.

Imlygic

Talimogen laherparepvec (trade name: Imlygic , manufacturer: Amgen ) is used for the oncolytic therapy of malignant melanoma. It is a genetically modified herpes simplex virus type 1 (replication competent). It was approved in the USA in October 2015 and in the EU in December 2015.

Strimvelis

Strimvelis (trade name: Strimvelis , manufacturer: GSK ) is an ex vivo gene therapy for ADA-SCID (severe combined immunodeficiency). Autologous (derived from the patient himself), CD34 + -enriched blood stem cells are transduced with a retroviral vector which codes for the human ADA cDNA sequence. The cells, which henceforth express a working version of adenosine deaminase (ADA), are re-injected (transplanted) into the patient. The therapy costs around 600,000 euros per patient, which on the other hand only corresponds to enzyme replacement therapy in around 2 years. It was approved in the EU in May 2016.

Kymriah

Tisagenlecleucel (CTL019) (trade name: Kymriah ; manufacturer Novartis ) is the first CAR-T-cell therapy agent to be approved in the USA for adoptive immunotherapy, specifically against acute lymphoblastic leukemia (ALL). Since this is the first representative of a completely new therapy, it is called first in class . It is the first gene therapy approved in the USA. Kymriah was approved in the EU on August 27, 2018, and subsequently also in Switzerland on October 22, 2018.

On April 22, 2020, the American health authority FDA granted Kymriah the special status of “Regenerative Medicine Advanced Therapy” (RMAT) for drugs that can be used against previously untreatable diseases in the approval process for the treatment of recurrent or therapy-resistant follicular lymphoma.

Novartis set the treatment price at $ 475,000. The Federal Office of Public Health is responsible for setting prices in Switzerland . In March 2019, Novartis and GWQ ServicePlus agreed on a new reimbursement model: Novartis will reimburse part of the drug costs for Kymriah to GWQ if the therapy result "survival" is not achieved within a defined period of time. This " Pay for Outcome " reimbursement is already being used in other indications. However, the patents on Kymriah are controversial; After two NGOs objected, the manufacturer Novartis withdrew a patent.

Luxturna

Voretigen neparvovec (trade name: Luxturna , manufacturer: Spark Therapeutics ) received a unanimous approval recommendation for a hereditary retinal disease for the USA in October 2017 . Luxturna was approved in the United States in December 2017.

It has also been approved in the EU since November 2018. Novartis acquired the therapy from Spark Therapeutics in January 2018 through a license agreement. Novartis is authorized to distribute Luxturna® in all countries outside the United States.

The cost of the Luxturna treatment is estimated at $ 850,000.

Zolgensma

Onasemnogene abeparvovec-xioi (trade name: Zolgensma , manufacturer: AveXis , a subsidiary of Novartis ) is an adeno-associated virus vector-based gene therapy that is used for the treatment of pediatric patients up to two years of age with spinal muscular atrophy (SMA) type 1 , with bi-allelic mutations in the survival motor neuron 1 (SMN1) gene. Zolgensma received FDA approval in May 2019. The high-priced gene therapeutic agent was approved in the EU at the end of May 2020. In March 2020, the European Medicines Agency issued a positive opinion (ie a recommendation for approval) for the EU.

In August 2019, the US health authority accused Novartis of having withheld test results in the approval process. In January 2020, Novartis announced that it would be giving away 100 treatments. "There are no reasons to contradict the compassionate use program ," the Paul Ehrlich Institute is quoted as saying on SPON , although Zolgensma was not yet approved in Europe at the time.

Comparative studies with the approved therapy with Nusinersen (trade name: Spinraza, Biogen ) regarding effectiveness and safety do not exist. In Spinraza it is not a gene therapy. Therefore - in contrast to Zolgensma - it has to be given quarterly. Gene therapy sees the one-time treatment; H. Healing before. In May 2020 the authors of the Süddeutsche Zeitung Astrid Viciano and Michele Catanzaro dealt with the pricing of Zolgensma .

Yescarta

Axicabtagene Ciloleucel (trade name: Yescarta ; manufacturer Kite Pharma , a subsidiary of Gilead Sciences ) is the second CAR-T-cell therapy agent that is used for treatment in both the US (October 2017) and the EU (August 2018) of B-cell lymphomas . In March 2020, the pharmaceutical company Bristol-Myers Squibb (as legal successor to Juno Therapeutics ) was awarded a claim for damages in the amount of 1.2 billion US dollars due to patent infringements . Gilead announced that it would object to this.

Zynteglo

Since January 2020, it has been possible to treat a form of the rare hereditary disease transfusion-dependent β-thalassemia (TDT) with the gene therapy Zynteglo (autologous CD34 + cells that code for the β A-T87Q -globin gene). The manufacturer is the US pharmaceutical company bluebird bio . Germany is the first country where this treatment is available.

Indications

Numerous indications are currently being investigated in gene therapy, e.g. B .:

  • Hemophilia : In an audio interview in the NEJM : “Most will agree that therapies that only have to be administered once are preferable to repeated medication. Therefore, the expectation is that the long-term benefits of these one-off therapies will justify the high costs. ”The interview partners also cite haemophilia as an example: the standard treatment“ can cost US $ 400,000 and more per year and patient. ” BioMarin launched one at the end of 2019 Approval for their gene therapy ( valoctocogene roxaparvovec ) requested from the FDA. According to the Wallstreet Journal , BioMarin is aiming for a price of between USD 2 and 3 million for Valrox on the grounds that this cure eliminates future therapy costs.

Companies

See also

literature

  • Gene Therapy Clinical Trials Worldwide Overview database of the clinical gene therapy studies carried out and ongoing
  • Inder M. Verma: Gene Therapy. In: Scientific American. November 1990.
  • DT Suzuki et al.: Genetics. VCH Verlag, 1991.
  • Benjamin Lewin: Genes V. Oxford University Press, 1994.
  • Deutsches Ärzteblatt . 100, No. 6, 2003, pp. A 314-318.
  • James M. Wilson: Gendicine : The First Commercial Gene Therapy Product; Chinese Translation of Editorial . In: Human Gene Therapy . tape September 16 , 2005, p. 1014-1015 , doi : 10.1089 / hum.2005.16.1014 .
  • AM Raem among others: genetic medicine, an inventory. Springer-Verlag, 2001.
  • DFG statement on the development of gene therapy, December 2006 (PDF) (236 kB)
  • Christopher Baum et al .: Side effects of retroviral gene transfer into hematopoietic stem cells. In: Blood. 101/2003, pp. 2099-2114.
  • PA Horn et al .: Stem cell gene transfer - efficacy and safety in large animal studies. In: Molecular Therapy. 10/2004, pp. 417-431.
  • Boris Fehse, Silke Domasch (ed.): Gene therapy in Germany. An interdisciplinary inventory. 2nd, updated and expanded edition. Dornburg, 2011, ISBN 978-3-940647-06-1 . ( Short version as PDF )
  • Christopher Baum, Gunnar Duttge , Michael Fuchs: Gene therapy. Medical and scientific, legal and ethical aspects. (= Ethics in the life sciences. Volume 5). Karl Alber Verlag, Freiburg i. Br./ Munich 2013, ISBN 978-3-495-48593-4 .

Web links

Individual evidence

  1. Directive 2009/120 / EC of the Commission of September 14, 2009 amending Directive 2001/83 / EC of the European Parliament and of the Council on the creation of a Community code for medicinal products for human use with regard to advanced therapy medicinal products.
  2. a b c Medicines for Advanced Therapies (ATMP) - Gene Therapeutics , website of the Paul Ehrlich Institute (PEI), accessed on November 27, 2019.
  3. Approved Cellular and Gene Therapy Products , FDA website, accessed November 27, 2019.
  4. ^ Robert E. MacLaren, Markus Groppe, Alun R. Barnard, Charles L. Cottriall, Tanya Tolmachova: Retinal gene therapy in patients with choroideremia: initial findings from a phase 1/2 clinical trial . In: Lancet (London, England) . tape 383 , no. 9923 , March 29, 2014, ISSN  1474-547X , p. 1129–1137 , doi : 10.1016 / S0140-6736 (13) 62117-0 , PMID 24439297 , PMC 4171740 (free full text).
  5. Moritz Lindner, Michael J. Gilhooley, Stuart N. Peirson, Steven Hughes, Mark W. Hankins: The functional characteristics of optogenetic gene therapy for vision restoration . In: Cellular and Molecular Life Sciences . July 29, 2020, ISSN  1420-682X , doi : 10.1007 / s00018-020-03597-6 ( online [accessed July 29, 2020]).
  6. International Law. (No longer available online.) The Genetics and Public Policy Center, Johns Hopkins University Berman Institute of Bioethics, 2010, archived from the original September 2, 2014 ; accessed on September 22, 2018 .
  7. M. Cavazzana-Calvo, A. Fischer: Gene therapy for severe combined immunodeficiency: are we there yet? . In: J Clin Invest. 117 (6), Jun 2007, pp. 1456-1465. Review: PMID 17549248
  8. The Journal of Gene Medicine Clinical Trial site ( Memento of November 14, 2007 in the Internet Archive )
  9. K. Welte, CY Wang, R. Mertelsmann, S. Venuta, SP Feldman, MA Moore: Purification of human interleukin 2 to apparent homogeneity and its molecular heterogeneity. In: J Exp Med. 156 (2), Aug 1, 1982, pp. 454-464. PMID 6980256 , PMC 2186775 (free full text)
  10. K. Welte, E. Platzer, L. Lu, JL Gabrilove, E. Levi, R. Mertelsmann, MA Moore: Purification and biochemical characterization of human pluripotent hematopoietic colony-stimulating factor. In: Proc Natl Acad Sci U.S.A. 82 (5), Mar 1985, pp. 1526-1530. PMID 3871951 , PMC 397296 (free full text)
  11. ^ R. Mertelsmann, K. Welte, C. Sternberg, R. O'Reilly, MA Moore, BD Clarkson, HF Oettgen: Treatment of immunodeficiency with interleukin-2: initial exploration. In: J Biol Response Mod. 3 (5), Oct 1984, pp. 483-490. PMID 6334136
  12. H. Glimm, K. Flügge, D. Möbest, VM Hofmann, J. Postmus, R. Henschler, W. Lange, J. Finke, HP Kiem, G. Schulz, F. Rosenthal, R. Mertelsmann, C. von Kalle: Efficient serum-free retroviral gene transfer into primitive human hematopoietic progenitor cells by a defined, high-titer, nonconcentrated vector-containing medium. In: Hum Gene Ther. 9 (6), Apr 10, 1998, pp. 771-778. PMID 9581900 .
  13. H. Veelken, A. Mackensen, M. Lahn, G. Köhler, D. Becker, B. Franke, U. Brennscheidt, P. Kulmburg, FM Rosenthal, H. Keller, J. Hasse, W. Schultze-Seemann, EH Farthmann, R. Mertelsmann, A. Lindemann: A phase-I clinical study of autologous tumor cells plus interleukin-2-gene-transfected allogeneic fibroblasts as a vaccine in patients with cancer. In: Int J Cancer. 70 (3), Jan 27, 1997, pp. 269-277. PMID 9033626
  14. G. Stingl, EB Brŏcker, R. Mertelsmann, K. Wolff, S. Schreiber, E. Kămpgen, A. Schneeberger, W. Dummer, U. Brennscheid, H. Veelken, ML Birnstiel, K. Zatloukal, W. Schmidt , G. Maass, E. Wagner, M. Baschle, M. Giese, ER Kempe, HA Weber, T. Voigt: Phase I study on the immunotherapy of metastatic malignant melanoma by a cancer vaccine consisting of autologous cancer cells transfected with the human IL-2 genes. In: Hum Gene Ther. 7 (4), Mar 1, 1996, pp. 551-563. PMID 8800750
  15. ^ Mary Carrington Coutts: Human Gene Therapy. In: Kennedy Institute of Ethics Journal. Volume 4, Number 1, March 1994, pp. 63-83. doi: 10.1353 / ken.0.0148
  16. In: Nature Biotechnology . December 17, 1999, p. 1153. (nature.com)
  17. P. Tebas, D. Stein, WW Tang, I. Frank, SQ Wang, G. Lee, SK Spratt, RT Surosky, MA Giedlin, G. Nichol, MC Holmes, PD Gregory, DG Ando, ​​M. Kalos, RG Collman, G. Binder-Scholl, G. Plesa, WT Hwang, BL Levine, CH June: Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. In: The New England Journal of Medicine . Volume 370, number 10, March 2014, pp. 901-910, doi: 10.1056 / NEJMoa1300662 . PMID 24597865 , PMC 4084652 (free full text).
  18. Summary of the European public assessment report (EPAR) for Glybera , der EMA, accessed on November 24, 2015.
  19. Summary of the EPAR for the public of the EMA (German), accessed on November 24, 2015.
  20. Adeno-associated viral vector expressing lipoprotein lipase. Community register of orphan medicinal products, accessed June 8, 2013 .
  21. Agreement for the first approved gene therapy against enzyme deficiency is available , PM of the National Association of Statutory Health Insurance Funds from November 23, 2015, accessed on November 24, 2015.
  22. uniQure Announces It Will Not Seek Marketing Authorization Renewal for Glybera in Europe , PM uniQure from April 20, 2017, accessed on October 15, 2017.
  23. Goodbye Glybera! The World's First Gene Therapy will be Withdrawn ; Labiotech.eu of April 20, 2017, accessed October 15, 2017.
  24. Biohackers want to recreate withdrawn gene therapy , accessed on October 14, 2019.
  25. Summary of the European public assessment report (EPAR) for Imlygic , the EMA, accessed on October 15, 2017.
  26. Summary of the EPAR for the public of the EMA (German), accessed on October 15, 2017.
  27. Summary of the European public assessment report (EPAR) for Strimvelis , the EMA, accessed on October 15, 2017.
  28. Summary of the EPAR for the public of the EMA (German), accessed on October 15, 2017.
  29. KYMRIAH , FDA Status, accessed September 9, 2017.
  30. Novartis receives first ever FDA approval for a CAR-T cell therapy, Kymriah (TM) (CTL019), for children and young adults with B-cell ALL that is refractory or has relapsed at least twice , PM Novartis of August 30, 2017 , accessed October 15, 2017.
  31. Novartis gene therapy for blood cancer approved in the EU. Reuters, August 27, 2018.
  32. Novartis receives European Commission approval of its CAR-T cell therapy, Kymriah® (tisagenlecleucel) , Novartis from August 27, 2018.
  33. Swissmedic gives the green light for Novartis hope In: handelszeitung.ch . October 22, 2018, accessed October 22, 2018.
  34. Press release submission: NOVARTIS: Novartis Kymriah® receives FDA Regenerative Medicine Advanced Therapy designation in follicular lymphoma. Retrieved April 29, 2020 .
  35. First gene therapy is approved in the USA. In: NZZ. August 30, 2017 ( nzz.ch , accessed September 9, 2017)
  36. Novartis Pharma GmbH and GWQ ServicePlus AG conclude an agreement on an innovative reimbursement model for CAR-T cell therapy , PM GWQ of March 6, 2019, accessed on March 7, 2019.
  37. Novartis Pharma GmbH and GWQ ServicePlus AG sign an agreement on an innovative reimbursement model for CAR-T cell therapy. Novartis press release March 6, 2019, accessed March 7, 2019.
  38. Cancer drug Kymriah: If the patient dies, the health insurance company pays less. In: Spiegel online. March 6, 2019, accessed on March 7, 2019 (spiegel.de)
  39. Isabel Strassheim: Novartis gives in to an NGO for the first time. In: bazonline.ch . December 17, 2019, accessed December 17, 2019 .
  40. Celine Müller, pharmacist, editor DAZ.online (cel): Novartis withdraws patent from Kymriah. December 19, 2019, accessed December 20, 2019 .
  41. FDA Advisory Committee Unanimously Recommends Approval of Investigational LUXTURNA ™ (formerly neparvovec) for Patients with Biallelic RPE65-mediated Inherited Retinal Disease , PM Spark Therapeutics, October 12, accessed September 24, 2018.
  42. Luxturna FDA , FDA website, accessed September 24, 2018.
  43. Luxturna - voretigene neparvovec , EMA, accessed on March 17, 2019.
  44. Gene therapy against hereditary eye diseases approved in the EU , DAZ of November 23, 2018, accessed on March 17, 2019.
  45. We may soon have our first $ 1 million drug. Who will pay for it? And how? , Statnews.com of October 13, 2017, accessed October 15, 2017.
  46. Novartis gene therapy before approval in Europe Handelsblatt dated September 21, 2018, accessed on March 17, 2019.
  47. ZOLGENSMA , FDA, accessed August 9, 2019.
  48. Markus Grill and Antonius Kempmann: Zolgensma - How the world's most expensive drug came about. In: tagesschau.de. May 19, 2020, accessed May 21, 2020 .
  49. Meeting highlights from the Committee for Medicinal Products for Human Use (CHMP) March 23-26, 2020 , PM EMA of March 27, 2020, accessed on March 30, 2020
  50. Statement on data accuracy issues with recently approved gene therapy , FDA statement from August 6, 2019, accessed on August 9, 2019.
  51. Novartis because of the manipulation of test data in the sights of the US supervisors , NZZ from August 7, 2019, accessed on August 9, 2019.
  52. What is behind the raffle for a drug worth millions , SPON from February 2, 2020, accessed on February 2, 2020
  53. German children can take part in the raffle for the most expensive drug in the world , SPON from February 3, 2020, accessed on February 3, 2020
  54. Thorsten Maybaum and Kathrin Gießelmann: Non-approved gene therapy: health insurance companies under pressure. In: Deutsches Ärzteblatt. November 19, 2019, accessed May 21, 2020 .
  55. The Value of Life , SZ from 23./24. May 2020, accessed on May 24, 2020
  56. Kite's Yescarta ™ (Axicabtagene Ciloleucel) Becomes First CAR T Therapy Approved by the FDA , PM Gilead, October 18, 2017, accessed February 18, 2020
  57. Yescarta® (Axicabtagene Ciloleucel) Receives European Marketing Authorization , PM Gilead, August 27, 2018, accessed February 18, 2020
  58. Yescarta Axicabtagene Ciloleucel , EPAR of the EMA, accessed on February 18, 2020
  59. Bristol-Myers Patent Win Over Gilead Grows to $ 1.2 Billion Bloomberg, April 10, 2020, accessed April 13, 2020
  60. Bristol-Myers' win in US patent case against Gilead boosted to $ 1.2 billion , Reuters April 10, 2020, accessed April 13, 2020
  61. Zynteglo , EPAR of the EMA, accessed April 10, 2020
  62. bluebird bio starts in Germany with the world's first market launch of the gene therapy Zynteglo , PM bluebird bio on January 13, 2020, accessed on April 10, 2020
  63. Bluebird Bio Launches First Gene Therapy for Beta Thalassemia in Germany , Labiotech.eu of January 14, 2020, accessed on April 10, 2020
  64. ^ NEJM : Gene Therapy
  65. ^ NEJM : Interview with Dr. Katherine A. High on gene therapy for genetic disease.
  66. WSJ : BioMarin Explores Pricing Experimental Gene Therapy at $ 2 million to $ 3 million
  67. Future therapy options
  68. Novartis enters agreement to acquire AveXis Inc. for USD 8.7 bn to transform care in SMA and expand position as a gene therapy and Neuroscience leader , PM Novartis, April 9, 2018, accessed December 4, 2019
  69. Novartis successfully completes acquisition of AveXis, Inc. , PM Novartis, May 15, 2018, accessed December 4, 2019
  70. Roche enters into definitive merger agreement to acquire Spark Therapeutics , Roche PM on February 25, 2019, accessed on March 18, 2019.
  71. ^ Roche Agrees to Buy Biotech Firm Spark Therapeutics , WSJ February 23, 2019, accessed March 18, 2019.
  72. How Jeff Marrazzo Chased Spark's Price , Handelszeitung March 16, 2019, accessed March 18, 2019.
  73. ^ Roche concludes acquisition of Spark Therapeutics, Inc. to strengthen presence in gene therapy ; Roche press release December 17, 2019, accessed December 17, 2019.
  74. Ferring and Blackstone Life Sciences Invest Over $ 570 Million USD in Novel Gene Therapy for Bladder Cancer Patients , Blackstone PM, November 25, 2019, accessed November 27, 2019
  75. Astellas Agrees to Buy Audentes in $ 3 Billion Gene-Therapy Deal , Bloomberg December 3, 2019, accessed December 4, 2019