throbber
E v o l u t i o n o f G e n e T h e r a p y,
`H i s t o r i c a l P e r s p e c t i v e
`
`Harry L. Malech, MDa,*, Elizabeth K. Garabedian, RN, MSLSb, Matthew M. Hsieh, MDc
`
`KEYWORDS
` Gamma retrovirus vector  Lentiviral vector  Gene editing
` Hematopoietic stem cells  Transduction  Apheresis  CD341 HSC
` Insertional mutagenesis
`
`KEY POINTS
` Gene therapy by the genetic modification of hematopoietic stem cells (HSC) has reached
`a stage of development that has resulted in substantial clinical benefits.
` This article explores the separate threads of knowledge, conceptual design, materials,
`and equipment required to reach our current era of clinically beneficial gene therapy.
` The history of gene therapy targeting hematopoietic stem cells include improvements in
`integrating vectors such as lentivectors and improvements in gene editing methods
`such as CRISPR/Cas9.
` Understanding the pathophysiology of adverse events such as insertional mutagenesis is
`important for seeking improvements in vector design that may enhance the safety of gene
`therapy.
`
`INTRODUCTION TO THEORETIC CONCEPTS AND EARLY BACKGROUND HISTORY
`IMPACTING HEMATOPOIETIC STEM CELL GENE THERAPY
`
`The history of gene therapy comprises the advance of theoretic concepts, under-
`standing of the human genome, availability of critical materials and instruments,
`design of vectors and chemical tools to manipulate and change genomic DNA, im-
`provements in the procurement and culture/maintenance of stemness of HSC in cul-
`ture, improvements in myeloid conditioning, the outcomes of conduct of clinical trials,
`observing successes and problems occurring in clinical trials, and deep study and
`
`Conflict of Interest: The authors declare no conflicts.
`a Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, Na-
`tional Institute of Allergy and Infectious Diseases, National Institutes of Health, 10 Center
`Drive, MSC1456, Bldg 10, Rm 5-3750, Bethesda, MD 20892-1456, USA; b National Human
`Genome Research Institute, NIH, 10 Center Dr, MSC1611, Bldg 10, Rm 10C-103, Bethesda, MD
`20892-1611, USA; c National Heart, Lung, and Blood Institute, NIH, 10 Center Drive, MSC1812,
`Bldg 10, Rm 9N119, Bethesda, MD 20892-1812, USA
`* Corresponding author.
`E-mail address: hmalech@niaid.nih.gov
`
`Hematol Oncol Clin N Am 36 (2022) 627–645
`https://doi.org/10.1016/j.hoc.2022.05.001
`0889-8588/22/Published by Elsevier Inc.
`
`hemonc.theclinics.com
`
`SKI Exhibit 2074
`Page 1 of 19
`
`

`

`628
`
`Malech et al
`
`elucidation of the mechanisms of problems that arise in clinical trials to seek and incor-
`porate corrective measures. The evolution of our understanding of ethical
`issues
`impacting gene therapy, and the logistics of access to and cost of successful gene
`therapy treatments are also important elements of this history.1
`In this broad brush and somewhat unconventional view of the history of gene
`therapy, we address general principles; key experiments, basic science, and clinical
`trials that illustrate some general principle; and the evolution of materials and instru-
`mentation that make current clinical approaches to gene therapy of HSC possible.
`We aim to complement rather than duplicate the extensive discussion of the back-
`ground studies of gene therapy and the march of the many published clinical trials
`in specific disorders or categories of disorders that are the subject of the other chap-
`ters in this series, as well as excellent recent reviews.2
`The earliest experiments that laid the foundation for gene therapy began with exper-
`iments on the transforming properties of bacteria.3 Alloway reported in 1932 that non-
`virulent (R type) pneumococci became lethal by adding cell-free extracts from virulent
`(S type) pneumococci. When injected with these “transformed” pneumococci, the
`mice developed pneumonia and died.
`In our view, the key conceptual background to all gene therapy emerged in the
`1940s with the seminal work by Avery and colleagues on bacterial transformation
`(which one could perhaps very loosely call gene therapy of bacteria). They identified
`DNA as the transforming factor that could change the physiology of a bacterial strain,4
`and more specifically, showed that the “transforming substance” was precipitated out
`by alcohol and later confirmed to be DNA. This was one of the key background ele-
`ments to Watson and Crick in identifying the structure of DNA,5 postulating its role
`as the genomic code of all prokaryotic and eukaryotic organisms, and thus demon-
`strating that nucleic acid sequences, rather than proteins, carry genetic information.
`The next critical discovery was that of Marshall Nirenberg, who in 1961 discovered
`the “triplet” code by which DNA encodes for the assembly of the 20 amino acids
`that serve as the building blocks of proteins.6
`In parallel with this elucidation of the biochemical basis of heredity, were emerging
`concepts from early transformation studies in mammalian cells, for example, the early
`reports that the transformation of 8-azaguanine sensitive cells with nuclei and chromo-
`somes from 8-azaguanine resistant cells rendered the transformants resistant due to
`transfer of a mutated hypoxanthine-guanine phosphoribosyltransferase gene.7,8 An
`early review of mammalian cell transformation studies conducted over the following
`18 years was reviewed in 1980 by Shows and Sakaguchi.9 This body of work further
`established that newly acquired biochemical traits from DNA transformation experi-
`ments in mammalian cells can be heritable.
`Many other key concepts that evolved into current methods of viral vector-
`mediated gene therapy were developed in the 1970s, during a period of the active
`investigation of viruses capable of transforming normal tissues into cancers. From
`this work, the concept emerged that perhaps these DNA and RNA tumor viruses
`known to insert into the genome of target cells could be modified in some way to
`remove the tumor causing elements, but retain their genome insertion capabilities
`to deliver a therapeutic payload. Some of the earliest published reviews of the history
`of gene therapy incorporating these essential concepts were those written in a series
`of reports over time by Theodore Friedmann10–13 who shared the 2015 Japan Prize
`with Alain Fischer for “For the Proposal of the Concept of Gene Therapy and its Clin-
`ical Applications.”
`More generally, the term “gene therapy” now broadly includes the introduction or
`manipulation of DNA or RNA sequences in human cells to treat disease. There is a
`
`SKI Exhibit 2074
`Page 2 of 19
`
`

`

`History of Gene Therapy
`
`629
`
`general consensus among the US Food and Drug Administration (FDA),14 the Euro-
`pean Medicines Agency (EMA),15 and the American Society of Gene and Cell Therapy
`(ASGCT)16 defining gene therapy as changes in gene expression, achieved by replac-
`ing or correcting a disease-causing gene, inactivating a target gene, or inserting a new
`or modified gene, using a vector or delivery system of genetic sequence or gene,
`genetically modified microorganisms, viruses, or cells.
`By the late 1970s, while our understanding of the molecular basis of human dis-
`eases was advancing through cloning and sequencing of genes, there were major
`technical challenges to implement gene transfer. Exogenous DNA could be introduced
`to target cells by transformation or transfection, but the overall efficiency was low.
`Additionally, if the introduced gene(s) did not provide a survival advantage, the dura-
`bility of gene transfer was also low. The resulting gene transfer efficiency at that time
`was about one in 100,000 cells, but nonetheless was proposed as a method to
`achieve genetic correction.17
`Intense interest in inherited hemoglobinopathies such as sickle cell disease and
`beta-thalassemia fueled work on beta-globin, one of the first genes to be cloned
`and then studied with the intent of gene transfer for clinical application. Mulligan
`and colleagues replaced the viral capsid protein (VP1) of the SV40 genome with
`complementary DNA of rabbit beta-globin in a monkey kidney cell line, which pro-
`duced large quantities of rabbit beta-globin mRNA and protein.18 As there was no
`inherent advantage for beta-globin gene transformed cells, several
`laboratories
`worked on selectable genes to be cotransferred. Pellicer and colleagues success-
`fully inserted beta-globin and thymidine kinase (TK) genes into murine teratocarci-
`noma cells.19 The Cline laboratory inserted dihydrofolate reductase (DHFR) or TK
`in murine marrow cells.20
`Cline and colleagues from UCLA then applied these results and tested them clin-
`ically.21–23 An experimental protocol to insert genetically modified marrow cells from
`patients with beta-thalassemia, inject the cells in the femur after local irradiation, and
`treat with a selecting agent was submitted to the human research review committee
`at their home institution. Because the first 2 patients to be treated were receiving
`their care in other countries (in a hospital in Naples, Italy and at Hadassah Hospital,
`Jerusalem, Israel), not covered by the UCLA review committee, the team sought in
`parallel and secured permission in Naples and Hadassah for the clinical study.
`Both patients were informed of the experimental nature and the low likelihood of suc-
`cess in this approach. After femur irradiation and infusion of modified marrow cells,
`the patients reported no adverse events, and selective agents were not used. Three
`months later, there was no demonstrable clinical benefit in both patients. Although
`safety of this clinical gene transfer was undebated, many controversial
`issues
`were brought forth.24–26 Can a clinical protocol proceed with permission from
`some but not all institutions? How many preclinical experiments (in vitro or animal),
`and what degree of “success” are needed to garner approval? While the responses
`to these issues are much more straightforward today, various review committees at
`that time were caught off guard and the consensus was that this was a rather prema-
`ture and in retrospect problematic initial attempt at the clinical application of gene
`therapy.27
`These first 2 attempts at human gene therapy generated much media attention and
`scrutiny by regulatory committees. The remainder of the decade into the early 1990s,
`scientists was quietly working on recombinant DNA methods, in vitro and animal
`models for testing, and strategies to enhance transgene expression. It quickly became
`clear that using viral vectors was more efficient in gene transfer than the previous
`methods of physical entry by transfection, fusion, or even electroporation. Much of
`
`SKI Exhibit 2074
`Page 3 of 19
`
`

`

`630
`
`Malech et al
`
`the gene transfer experiments then focused on vector optimization and design, and
`brought this background discussion into the early modern era of gene therapy.
`The following sections of this review will provide a historical background of a num-
`ber of parallel developments that provided the laboratory and clinical tools and mate-
`rials that facilitate our current approaches to gene therapy targeting blood cells
`including HSC.
`
`DESIGN OF INTEGRATING VECTORS USED FOR HEMATOPOIETIC STEM CELLS GENE
`THERAPY
`
`Vectors engineered from gamma retroviruses,28 long under study as the cause of a va-
`riety of cancers in mice, had the desired property of efficient insertion into the genome
`of target cells. Murine gamma retroviruses and their derivatives were the first of the
`genome integrating vectors to be applied to T lymphocytes and HSC in the clinical
`setting.
`Gamma retroviruses are RNA viruses, that on entry into a cell, are “reverse tran-
`scribed” (hence “retro”virus) into a DNA sequence. It is the DNA virus sequence
`that ultimately inserts itself into the host cell’s genomic DNA, becoming a “provirus”
`that in turn generates RNA virus sequences and viral mRNAs encoding virus proteins
`required for the replication phase of the virus life cycle. The critical issue was how to
`turn these viruses that efficiently insert provirus DNA genomic sequence into mamma-
`lian cell genomes, but are also efficient at causing tumors, into safe tools for gene ther-
`apy. The solution was to remove and/or inactivate as many elements of the virus
`genome as possible, while still retaining the ability of the highly engineered provirus
`sequence to insert efficiently into the mammalian cell genome. The goal was a func-
`tioning single-cycle virus capable of cell entry, uncoating, reverse transcription into
`provirus DNA, and insertion into the genome, but incapable of generating infectious
`virus. The solution involved separating the key elements required to generate
`replication-incompetent viral vector into 3 separate “production plasmids”: (1) an en-
`velope (env) producing element (the vector virus coat also serving the purpose of bind-
`ing to target cell and facilitating virus payload entry); (2) a gag-pol producing element
`(gag protein important for vector RNA packaging and polymerase for reverse tran-
`scribing the RNA); and (3) the vector sequence (retaining the psi element needed for
`packaging and the long terminal repeat (LTR) sequences at both ends of the vector
`sequence, which serves both as the internal strong promoter driving the production
`of a therapeutic protein and containing initiation elements binding the 2 ends of the
`vector for the circle formation required for reverse transcription). Where possible the
`env and gag-pol codons were changed to avoid recombination events that could
`reconstruct a replication-competent virus. To simplify the process of making different
`gamma retrovirus vectors, permanent packaging lines were devised that constitutively
`produce env and gag-pol, and when a specific vector sequence is added, clones
`could be assessed and chosen that constitutively produced vector in adequate titers.
`Many laboratories contributed to this technology and created a large array of different
`“flavors” of therapeutic gene therapy gamma retrovirus vectors. Many of these
`continue to be used for the production of some CAR-T lymphocytes or therapeutic
`cloned T cell receptors. This tour-de-force of engineering involving the contribution
`of many laboratories has served as the core technology used in the first generation
`of gene therapy targeting HSC or lymphocytes.
`The LTRs of gamma retroviruses were retained in the engineered vectors as conve-
`nient, very strong promoters to drive high levels of production of downstream inserted
`therapeutic protein-coding sequences. However, these same LTR elements contain
`
`SKI Exhibit 2074
`Page 4 of 19
`
`

`

`History of Gene Therapy
`
`631
`
`strong enhancer elements that can activate nearby genes. The engineered vectors by
`design retained the insertion targeting elements of the parent virus required to insert
`the DNA provirus into the mammalian genome. While the insertion of vector seems
`to be random, it is actually stochastic in that the mechanism used by the vector cou-
`ples to cellular elements, resulting in preferred sites of insertion into the genome.
`These preferred sites (also known as integration sites) are often located near the start
`of genes and in enhancer elements, and may in turn strongly interact with enhancer
`elements in the LTR.29,30 While the odds of any one insert occurring in a sensitive
`site are very low, gene therapy for a human subject may involve tens to hundreds of
`millions of insertions. Depending on the vector, the LTR and the host human subject
`disease substrate, we now know from adverse leukemic insertional mutagenesis
`events occurring in a number of clinical trials, that gamma retroviral vectors can trans-
`activate oncogenes such as LMO2, the MECOM complex, and other oncogene tar-
`gets to initiate the development of leukemia. These insertional mutagenesis events
`will be further discussed in greater detail in the last section of this historical review.
`Curiously, insertional mutagenesis leading to leukemic events has not been observed
`when the target of gamma retroviral vector gene therapy is T lymphocytes.
`Well before the first insertional mutagenesis, oncogenic events were observed in
`clinical trials of gene therapy using gamma retroviral vectors, certain limitations of
`this class of vectors (eg, limits of therapeutic payload size, limits on the use of alternate
`promoter elements instead of the LTR, absolute requirement for cell division for vector
`insertion into the genome) encouraged the development of gene therapy vectors
`derived from human immunodeficiency virus (HIV). HIV is part of a different group of
`retroviruses called lentiviruses and the vectors engineered from HIV are referred to
`here as “lentivectors.” HIV and other lentiviruses have a more complex structure,
`and have a number of required functional elements not present in gamma retroviruses,
`such as rev, that needed to be considered while engineering HIV into a safe gene ther-
`apy tool.31,32 As with gamma retroviruses, determining how much could be removed
`from the virus and whether the addition of elements from other viruses might enhance
`function and efficiency of the vector was an iterative discovery process. From a histor-
`ical perspective, some key advantages of lentivector function and engineering, and
`the insertional mutagenesis oncogenic events noted above have resulted for the
`most part in the abandonment of gamma retroviral vectors for the transduction of
`HSC for clinical trials.
`As with gamma retrovirus vectors, the production of lentivectors that are functional,
`but replication incompetent, required the separation of packaging elements into plas-
`mids separate from the transfer vector. Almost all lentivector production for clinical
`application uses the membrane fusion G protein derived from vesicular stomatitis virus
`(VSV-G) as the vector envelope element, rather than the natural env component of HIV.
`The cell membrane target of the VSV-G protein is ubiquitous to all cells with high ef-
`ficiency of binding and vector membrane fusion. Almost from the start, lentivector en-
`gineering strategies incorporated a self-inactivating (SIN) feature, modifying the LTR
`element that contains strong enhancers with transactivating potential and using safer
`promotors with little enhancer activity instead. This was accomplished by creating a
`0
`deletion in the 3
`LTR of the vector production plasmid. During vector production,
`0
`LTR assists in the important packaging biochemistry needed to produce
`the intact 5
`0
`infective but replication-incompetent lentivirus vector. During transduction, the SIN 3
`0
`LTR binds to the 5
`LTR in the circularization and priming step that retrotranscribes the
`0
`insertional provirus DNA from the lentivector RNA, and is incorporated into the 5
`end
`0
`LTR. This safety feature removes
`of the provirus DNA, thus “self-inactivating” the 5
`enhancer and activator elements, and allows the therapeutic payload transgene(s)
`
`SKI Exhibit 2074
`Page 5 of 19
`
`

`

`632
`
`Malech et al
`
`to be transcribed from a promoter of choice. The use of the SIN feature with an alter-
`nate internal promoter is not limited to lentivectors. A group of investigators incorpo-
`rated the SIN feature into the gamma retroviral vector that was efficacious in a clinical
`trial targeting hematopoietic stem cells (HSC) to treat infants with X-linked severe
`combined immunodeficiency (X-SCID). This SIN gamma retroviral vector showed clin-
`ical efficacy,33 with no insertional oncogenesis after median of 9 years of follow-up
`(personal communication S.-Y. Pai, 2022). Importantly, this maneuver does not alter
`the pattern of insertion characteristic of gamma retroviruses that tends to target
`0
`end of genes.33
`enhancer elements and the 5
`The larger payload capacity of lentivectors, SIN design, and potential to incorporate
`tissue-specific promoters and enhancers catalyzed concerted efforts by several in-
`vestigators to develop lentivectors that would drive high-level beta-globin expression
`at specific stages of erythroid precursor development. In the 1990s, the locus control
`region of beta-globin was discovered to contain hypersensitive sites (HS) that were
`important for high-level expression. After a series of in vitro experiments, a lentivector
`containing optimized regulatory elements, the TNS9 vector, was shown to drive high-
`level erythroid-specific expression of adult beta-globin in transduced murine HSC,
`successfully correcting a murine model of beta-thalassemia.34 This seminal work
`was followed shortly by the same group to correct another more severe phenotype
`of beta-thalassemia in Berkeley mice.35
`In parallel, another group designed a modified adult beta-globin for which the 87th
`amino acid was switched from threonine to glutamine, to mimic the antisickling effect
`of gamma-globin. This T87Q version successfully corrected 2 sickle mouse models36
`and was expressed at high levels when transduced human sickle cord blood stem
`cells were transplanted into immunodeficient mice.37 These reassuring preclinical
`studies ultimately led to the treatment of a patient with compound beta-E/beta-0 thal-
`assemia, who achieved transfusion independence.38
`From those early efforts, an increasing number of successful clinical trials have
`emerged to treat thalassemia and sickle cell disease, described in significant detail
`in other chapters in this series. Some of these studies will be revisited later in this
`article in comments relating to transduction enhancers and certain types of adverse
`events associated with gene therapy.
`
`GENE EDITING OF HEMATOPOIETIC STEM CELLS
`
`Gene editing is the most recently evolving technology to be applied to gene therapy for
`HSCs, having the capability of targeting a specific sequence within the genome, in
`contrast to the stochastic semirandom insertion of integrating viruses throughout
`the genome. The 4 main types of gene editing systems are meganucleases, zinc finger
`nucleases, transcription activator-like effector nucleases (TALENs),39 and clustered
`regularly interspaced short palindromic repeats-Cas (CRISPR-Cas).40 Of particular
`near term historical note are recent clinical reports of unequivocal clinical benefit
`from successful application of the CRISPR technology applied to HSCs for the correc-
`tion of hemoglobinopathies.41,42
`CRISPR is derived from an antivirus system that evolved in bacteria to “copy and
`memorize” virus sequence, thereby allowing the bacterium to target that same virus
`sequence for cleavage when subsequently infected by a similar virus.43 The discovery
`of the use of the CRISPR system for gene editing was the basis for the 2020
`Nobel Prize in Chemistry (https://www.nobelprize.org/prizes/chemistry/2020/press-
`release/). The CRISPR system has revolutionized the field, due to its ease of applica-
`tion and versatility. CRISPR-based derivative methods such as base editing and prime
`
`SKI Exhibit 2074
`Page 6 of 19
`
`

`

`History of Gene Therapy
`
`633
`
`editing use the core element of the CRISPR system to find the genome target
`sequence to enzymatically convert a single base pair or reverse transcribe a short
`sequence change into the genome, respectively.44
`Most current approaches to editing use electroporation to introduce the editing el-
`ements (CRISPR-Cas9 mRNA or protein, guide RNA, and any additional factors to
`augment editing) into HSC. There have been a number of small scale non-GMP
`research-grade electroporation instruments available for gene editing development
`work. Fortuitously, clinical scale and high throughput GMP compliant commercial in-
`struments in development in the past few years have become available just in time for
`the current initiatives in gene editing of HSC.
`
`HEMATOPOIETIC STEM CELLS: SOURCING, SELECTING, CULTURING, AND
`TRANSDUCING
`
`Parallel to the development of vectors and editing tools for gene modifying HSC, ad-
`vances in the procurement, culture, transduction, and engraftment of HSC have had
`an important impact on the field.
`HSC occupy niches in the marrow that facilitate the retention of pluripotent potential to
`give rise to all hematopoietic lineages and asymmetric proliferation of some progeny into
`lineage-specific progenitors.45 Sourcing HSC for gene therapy or conventional allogeneic
`transplants initially was restricted to the harvesting of bone marrow with needles. That HSC
`are constantly translocating at a slow rate from marrow to the circulation and back to the
`marrow was a key discovery that ultimately led to alternate sourcing of HSC.46 There is a
`steady state presence of CD341 HSC or progenitors in the peripheral blood of healthy
`humans of about 1400 cells per ml (1.4 per ml). This low baseline frequency of CD341
`HSC and progenitors in the circulation can be increased by daily injections of granulocyte
`colony-stimulating factor (G-CSF or filgrastim), which induces a transient release of
`CD341 HSC and progenitors from the marrow into the peripheral blood, peaking at 5 to
`6 days at an average of 76 per ml (a 50-fold increase), then declining, even with additional
`daily injections.47,48 Subsequent studies indicated that CXCR4 (the receptor for stromal
`cell-derived factor-1 [SDF-1], also known as CXCL12) tethers HSC within the marrow
`and that G-CSF breaks that tether by increasing granulocyte proliferation and release of
`granule enzymes in the marrow, thereby enhancing the release of HSC into the circulation.
`More recently a small molecule inhibitor of the binding site of CXCR4, plerixafor (previously
`called AMD3100), was also shown to release HSC from the marrow to the circulation,49
`and when administered in the combination with G-CSF results in a synergistic mobilization
`of HSC to the peripheral blood.50
`The ease of apheresis collection of stem cells using continuous flow instruments
`following mobilization has resulted in this method becoming the preferred sourcing
`of HSC for many gene therapy clinical studies. Some patients with some inherited
`blood disorders that impair marrow proliferation (Fanconi anemia for example) will
`not mobilize, and infants who are too small for standard apheresis procedures still
`require bone marrow aspiration for sourcing HSC for gene therapy. Patients with sickle
`cell disease are at high risk of adverse events including vaso-occlusive crises when
`treated with G-CSF. Fortunately, several groups have shown that efficient and safe
`mobilization of HSC from marrow to peripheral blood can be accomplished using
`plerixafor alone as the mobilization agent in patients with sickle cell.51–54
`
`Apheresis
`
`The first apheresis device for separating blood components was developed in a
`collaboration between the National Cancer Institute and IBM,55 and was appreciated
`
`SKI Exhibit 2074
`Page 7 of 19
`
`

`

`634
`
`Malech et al
`
`at the time as a critical breakthrough in instrumentation for the separation and collec-
`tion of blood components.56 A number of companies developed a series of progres-
`sively efficient continuous flow instruments to harvest of fractions enriched in HSC
`from G-CSF and/or plerixafor mobilized donors. Without this instrumentation now
`commonplace in blood collection centers, and simply viewed by the gene therapy
`community as “background” standard banking technology, much of the current prog-
`ress in gene therapy would not have been possible.
`
`Selection of Hematopoietic Stem Cells from Marrow or Apheresis Products
`
`Multipotent permanently repopulating HSC express the CD34 surface antigen that
`was originally called My-10 as detected by a murine monoclonal antibody, raised
`against the KG-1a human tumor cell line.57 Studies later demonstrated that immuno-
`magnetic beads coated with anti-CD34 antibody could form complexes with HSC in a
`marrow or apheresis product, which in turn could be purified with magnets. These
`initial studies used incubation with chymopapain to release HSC from the beads.58
`This magnetic bead selection approach became the basis for the commercial devel-
`opment of instruments for selective enrichment of HSC, 2 of which reached the late
`commercialization phase.
`Baxter International, Inc (then called Baxter Healthcare Corporation) together with
`its “spin-off” subsidiary Nexell, Inc was the first to develop a fully automated instru-
`ment called the Isolex 300i that relied on the binding of HSC to magnetic beads coated
`with the anti-CD34 antibody. Following magnetic separation, the washed product was
`exposed to an octapeptide that directly competed for the binding site of the anti-CD34
`antibody to the CD34 antigen, thus removing the antibody complexed magnetic beads
`from the cells. This allowed the beads to be retained by a second pass through the
`magnetic field, yielding a cell product free of the beads and antibody.59 A precommer-
`cial manual version of the Isolex system was used in one of the earliest clinical studies
`of gene therapy for CGD.60
`A very similar system developed in parallel by Miltenyi Biotec GmbH became their
`CliniMACS CD34 Reagent System. This system uses anti-CD34 antibody chemically
`conjugated to dextran beads with an iron oxide/hydroxide core. A binding column
`with a magnetic gradient is used to separate the HSC bound to the anti-CD34 mag-
`netic dextran bead from marrow or apheresis product. Unlike the Isolex system, there
`is no maneuver to separate the cells from the antibody-conjugated beads, which are
`presumably degraded in culture or in vivo following transplantation. In October 2003
`exclusive rights to market, the Isolex system was acquired by Miltenyi and not further
`developed by them leaving only the CliniMACS and its derivative devices available
`currently for the clinical selection of HSC for gene therapies.61
`
`Culture and Transduction of Hematopoietic Stem Cells
`
`As noted previously, successful gamma retrovirus vector transduction requires 1 cell
`division cycle to complete integration into the genome of a cell. While lentivector trans-
`duction does not absolutely require cell division, HSC must enter at least the G1 phase
`of the cell cycle, and exposure of HSC to growth factors for a period of time in culture
`seems to enhance transduction efficiency. While it was initially hoped that gene editing
`methods might not require the activation of HSC, a similar improvement of gene edit-
`ing when HSC are cultured with growth factors has been observed by many investiga-
`tors. Defining ex vivo culture conditions and growth factor combinations that enhance
`transduction or editing while minimizing loss of long-term marrow repopulating poten-
`tial has been intensely studied. The discovery of each of the many critical HSC growth
`factors important for both efficient vector transduction and gene editing approaches
`
`SKI Exhibit 2074
`Page 8 of 19
`
`

`

`History of Gene Therapy
`
`635
`
`will not be reviewed here; suffice it to say that without the work to identify, clone, and
`provide GMP compliant growth factors, gene therapy for HSC could not have
`advanced. While there is no consensus about “best” conditions, the minimum combi-
`nation of three growth factors, stem cell factor (SCF), FLT3-L, and thrombopoietin
`(TPO) is used by most of the investigators for clinical gene therapy for HSC applica-
`tions. Additional factors used by some investigators include interleukin (IL)-3 and/or
`IL-6. A key limiting issue is that culture in these growth conditions beyond 3 to
`4 days results in a significant loss of long-term repopulating potential. Outside the
`scope of this review is the discovery of other biochemical factors and conditions
`that may prolong the period during which HSC can be maintained in culture while
`delaying the loss of long-term engraftment potential. This is an important emerging
`field for the future impact on HSC gene therapy.
`While culture and transduction of HSC may be performed in standard tissue culture
`flasks, gas permeable flexible plastic culture bags are increasingly used to achieve
`more “closed system” handling. The earliest application of such systems in gene ther-
`apy clinical trials suggests better gas exchange, more consistent high viability and
`yield, and transduction efficiency.60,62
`
`Transduction Enhancers
`
`Maneuvers to achieve the highest efficiency transduction by gamma retroviral and len-
`tiviral vectors have been an important aspect of the history of gene therapy. Quite early
`on, it was shown that the addition of certain charged polymers to the transduction cul-
`ture, such as polybrene or protamine sulfate, would enhance transduction by gamma
`retrovirus vectors; of the 2, protamine sulfate continues to be used to enhance the
`transduction of lentivectors.63 Physical maneuvers such as centrifuging the culture
`plate or gas permeable bag have also been shown to enhance transduction,64 but
`the practicalities of application at clinical scale have limited the translation of this ma-
`neuver into the clinic. It has been presumed but not proven that ionic polymers and
`centrifugation worked in par

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket