Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Chapter 6

Genetic Manipulation Via Gene Transfer


Paul C. Edwards1 and Michael J. Passineau2
1
Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indianapolis, IN, USA
2
Division of Cardiovascular Medicine, Allegheny Health Network, Pittsburgh, PA, USA

Chapter Contents
6.1 Introduction to Basic Biology of Gene 6.3 Host Response to Gene Transfer 79
Transfer 75 6.3.1 Classical Anti-Viral Response to
6.2 Gene Transfer Methodologies 76 Viral-Mediated Gene Transfer 79
6.2.1 Specific Gene Delivery Approaches 77 6.3.2 Intracellular Response to Gene Transfer 80
6.2.1.1 Viral Vectors 77 6.3.3 Implications for Human Gene Therapy 80
6.2.1.2 Nanoparticles 77 6.4 Examples of Successful Genetic Manipulation
6.2.1.3 Naked DNA Entry Through Membrane Via In Vivo Gene Transfer 80
Disruption 77 6.4.1 Pre-Clinical Ex Vivo and In Vivo Gene Transfer
6.2.2 Gene Delivery Considerations 78 Strategies 80
6.2.2.1 Endosomal Escape Mediated by Viral 6.4.2 Future Potential Approaches to Gene Therapy
Vectors and Nanoparticles 78 Currently at Earlier Stages of Development 81
6.2.2.2 Cytoplasmic Trafficking of Genetic 6.4.3 Limitations of Pre-Clinical Studies 82
Payload 78 6.5 Pathway to Clinical Implementation of
6.2.2.3 Nuclear Import and Fate of Genetic Therapeutic Gene Transfer 82
Payload 78 6.6 Conclusion 83
6.2.2.4 Matching Vector and Target 79 References 83

6.1  INTRODUCTION TO BASIC BIOLOGY the cell and nuclear membranes. The gene drug, or “vector,”
serves as the template for synthesis of mRNA molecules
OF GENE TRANSFER
encoding the transgene sequence, and thus the transgene-
Gene transfer is a potentially powerful approach to stem cell encoding portion of the vector must survive the process and
biology and tissue engineering since it is the one method somehow arrive within the host cell nucleus intact. This
that allows manipulation of cells and tissues in their own transmembrane/­ transcytoplasmic voyage crosses multiple
language. By employing the subcellular machinery under- barriers and engages multiple subcellular mechanisms, and
lying the central dogma of molecular biology, gene transfer this diversity of challenges explains in part why gene therapy
engages and alters the software of cellular behavior. While has been so slow to demonstrate clinical fulfillment of its
drug or chemical-based therapeutics can certainly modify enormous potential.
cellular behavior, no method matches the potential of gene Transit of a gene transfer vector across the cell membrane
transfer for precise manipulation of cellular programming. has historically been regarded as the most challenging leg of
It should be appreciated that manipulation of the RNAi pro- the journey. The phospholipid bilayer of a cell is impervious
cess shares with gene transfer its mechanistic proximity to to charged molecules, and nucleic acid is strongly anionic.
the translational machinery of the cell, but is not considered Further, the highly regulated transmembrane transport em-
gene transfer per se, as RNAi strategies can only affect gene ployed by cells occurs by means of channels, pumps, and
expression by an inhibitory pathway and cannot introduce a other membrane complexes that regulate pores much too small
therapeutic transgene to a cell or tissue. for polynucleic acid molecules to transit. Thus, an intact cell
At its most basic level, gene transfer involves the trans- membrane presents an absolute barrier to gene transfer. If a
portation of nucleic acid (single or double-stranded) across cell membrane is impervious to the transit of polynucleic acid
Stem Cell Biology and Tissue Engineering in Dental Sciences. http://dx.doi.org/10.1016/B978-0-12-397157-9.00008-4
© 2015 Elsevier Inc. All rights reserved. 75
76  PART | II  In Vitro Regulation of Cell Behaviour and Tissue Development

FIGURE 6.1  Artist’s representation of the steps of viral-mediated gene transfer to a target cell.

vectors, gene transfer must rely upon one of three mechanisms: e­ lements (e.g., enhancers) in the vector engages native cellular
(1) receptor-mediated endocytosis (and to a much lesser ex- proteins that facilitate nuclear transport along microtubules.
tent pinocytosis); (2) disruption of the cell membrane; or (3) Similarly, nuclear entry has received more modest atten-
complexation of the polynucleic acid vector with an amphipa- tion, particularly when cellular targets are mitotic (e.g., stem
thic molecule that can mask its charge and allow it to diffuse cells), as nuclear breakdown during mitosis greatly facilitates
through the cell membrane. Gene transfer strategies have been nuclear entry. Assuming that most of the cell and tissue tar-
devised utilizing each of these membrane transit mechanisms, gets relevant to dental sciences are mitotic, this chapter will
and the advantages and challenges of each respective approach focus mainly upon strategies to transport a gene transfer vec-
will be discussed in more detail in the following section. tor from the exterior of the cell to the cytoplasm. Figure 6.1
Once the transgene-coding region of vector enters the summarizes the major steps in exogenous gene transfer.
cytoplasm, it engages the host cell’s microtubule network
to move toward the nucleus. This process has been studied
and appreciated, but is generally given far less attention than
6.2  GENE TRANSFER METHODOLOGIES
transit of the cell membrane. This is at least partially due As discussed above, gene transfer methodologies may
to the observation that inclusion of appropriate non-coding be classified according to the mechanism they employ to
Genetic Manipulation Via Gene Transfer Chapter | 6   77

FIGURE 6.2  Artist’s rendition of viral-mediated gene transfer (left), ultrasound-assisted gene transfer (center), and nanoparticle-mediated gene transfer
(right).

transit the cell membrane. Viral vectors utilize receptor- classical ligand(virus)/receptor(host cell) interactions, with
mediated cell uptake and subsequent endosomal escape. the ligand composed of a protein moiety embedded in the to-
Naked DNA methodologies, primarily electroporation pology of the viral capsid (e.g., adenovirus, ­adeno-associated
and sonoporation, rely upon physical disruption of the cell virus (AAV)) or comprising a domain of a glycoprotein
membrane, sometimes in combination with enhancement embedded in the viral membrane (e.g., Herpes simplex or
of endocytosis. So-called “nanoparticle” techniques em- lentivirus). Receptor binding stimulates endocytosis of the
body a continuum between the two, with such approaches receptor/virus complex by the host cell through the forma-
as simple transfection on one end and synthetic viruses on tion of an endosome budding from the cell membrane.
the other. Figure 6.2 provides representative illustrations of
these various methodologies. 6.2.1.2 Nanoparticles
In dental and craniofacial applications of gene therapy, it
Nanoparticles are a broadly applied term for any complex
is important to note that ease of access to target tissues defines
of naked DNA with an agent(s) acting as a carrier to en-
the paradigm according to which gene delivery strategies are
able the genetic payload to transit the cell membrane. It is
devised. Many target tissues are mucosal, and deeper struc-
important to note that while classical lipofection methods
tures can usually be accessed easily and precisely by injection.
work efficiently in vitro, the techniques are extremely lim-
This facile accessibility is not the norm in other applications of
ited in their in vivo efficacy. For this reason, most nanopar-
gene therapy, such as solid thoracic or visceral organs, where
ticle strategies involve a cellular entry process identical to
vascular transport of the vector must be utilized. As a result,
viral vectors, wherein there is a receptor/ligand interaction
some gene therapy innovations are relevant to dental and cra-
resulting in endocytosis and endosomal encapsulation. The
niofacial applications, and others (e.g., strategies to promote
DNA payload is compacted within a polymeric chemical
extravasation) are not. Overall, the target tissues contemplated
structure that often features multiple components. The va-
in this chapter bear greatest similarity to those target tissues
riety of nanoparticle formulations in the gene therapy lit-
where the greatest progress in clinical gene therapy has been
erature is myriad, but polyethylene glycol (PEG) is very
demonstrated (e.g., eye, muscle, skin), and these practical
considerations underscore the potential power of gene transfer commonly used as a component of nanoparticles due to its
apparent ability to extend the stability, and thus circulation
for dental and craniofacial therapeutics.
time, of nanoparticles in plasma.

6.2.1  Specific Gene Delivery Approaches 6.2.1.3  Naked DNA Entry Through
6.2.1.1  Viral Vectors Membrane Disruption
Viral vectors have evolved specific receptor affinities that In contrast to viral vectors and most nanoparticles, naked
govern their tropism for host tissues. These affinities mimic DNA methodologies rely on physical, rather than biological,
78  PART | II  In Vitro Regulation of Cell Behaviour and Tissue Development

entry to the cell. Pores are created in the cell membrane ei- understood, but it is clear that the mechanism of endosomal
ther through the application of a brief electrical pulse (“elec- escape is vector-specific and is accomplished by a wide va-
troporation”) or the violent destruction of microbubbles riety of viral capsid protein interactions. In at least one case,
in an applied acoustic field, resulting in inertial cavitation that of AAV, endosomal escape may not even occur until the
(“sonoporation”). Under optimal conditions, these pores are virus reaches the nucleus [2]. In the current state of the art,
large enough to permit the diffusion of naked DNA across protein-mediated endosomal disruption is not sufficiently un-
the cell membrane, while still small and transient enough to derstood to impart these properties to nanoparticles, and thus
avoid cellular toxicity from the reversal of transmembrane endosomal escape in nanoparticle strategies is mediated by
ionic gradients and uncontrolled influx of water. chemical structures that buffer the low pH environment of
Electroporation and sonoporation are rarely used for the endosome and lead to net water influx, leading to escape
in vitro gene transfer due to the increased fragility of cells through bursting of the endosome [3].
in culture and the availability of reliable chemical transfec-
tion techniques (e.g., lipofection). For in vivo gene transfer, 6.2.2.2  Cytoplasmic Trafficking
both techniques have been widely studied in animal models, of Genetic Payload
with a number of clinical trials now underway to evaluate
Cytoplasmic trafficking occurs via vector-specific pathways,
the safety and efficacy of gene electrotransfer in superfi-
but there is commonality in that the microtubule network
cial tissues such as prostate and skin [1]. Sonoporation has
is the physical substrate. For example, adenoviral infec-
not yet advanced to human clinical trials, although micro-
tion utilizes a predictable and rapid pathway involving viral
bubbles have found wide application in clinical practice as
uncoating, endosomal escape, and m ­ icrotubule-associated
ultrasound contrast agents and have been determined to be
transport involving dynein [4,5]. AAV, in contrast, ­appears to
safe, even at relatively large concentrations.
enter cells and traffic intracellularly via multiple ­pathways,
While electroporation and sonoporation are mechanisti-
and viral uncoating is delayed until nuclear entry [6].
cally similar, the instrumentation required to execute them
Nonviral vectors follow a pathways similar to a­ denovirus,
are fundamentally different, and these differences have im-
but are far less efficient at both endosomal escape and
portant implications for application in dental and craniofa-
­nuclear translocation [7,8].
cial tissues. Electroporation requires current to arc between
two electrodes, and thus the tissue target must be positioned
between the electrodes. In superficial structures, this is usu- 6.2.2.3  Nuclear Import and Fate
ally accomplished by needle-like electrodes inserted into of Genetic Payload
the target tissue. In contrast, the sonoporation relies upon an Nuclear import of exogenous DNA occurs efficiently in di-
externally applied ultrasound pulse that propagates through viding cells since the nuclear envelope disassembles dur-
soft tissue, requiring only that an uninterrupted pathway ing the M phase of cell division [9]. Mitotically active cells
through soft tissue exists between body surface and the comprise many, but not all, of the tissue targets relevant to
target tissue. For this reason, sonoporation is considered to this volume. For non-dividing cells, nuclear entry is more
be somewhat amenable to organ targeting through the en- complex, as the DNA molecules utilized in gene therapy
gineering of an emitter that creates an ultrasound “beam.” applications are too large to pass the nuclear pore com-
Supporting this concept is the feasibility of formulating plex and must enter the nucleus through facilitated trans-
antigen-targeted microbubbles intended to accumulate at a port. Characterization of so-called nuclear shuttle proteins,
disease target, such as a tumor. which bind exogenous DNA and subsequently interact with
nuclear transport mechanisms, has been an active area of
6.2.2  Gene Delivery Considerations research [10,11]. Fortunately, the practical upshot of this
complex topic is that viral vectors containing commonly
6.2.2.1  Endosomal Escape Mediated by utilized enhancer sequences, such as that from SV40, are
Viral Vectors and Nanoparticles sufficient to drive nuclear import and robust expression in
If cellular entry occurs via receptor-mediated uptake, the non-dividing cells.
mechanism utilized by viral vectors and nanoparticles, the Depending on the vector system used, genetic payloads
membrane budding that occurs results in the formation of an assume one of two distinctly different nuclear fates: epi-
endosomal compartment containing the vector. Once encased somal persistence or chromosomal integration. Episomal
within an endosome, a gene transfer vector must effect escape persistence is a key determinant of the duration of trans-
before the payload is degraded by nucleases present within gene expression, but is poorly understood, and may in-
the endosome. Several theories have been advanced as to the volve subcompartments of the nucleus, at least in the case
mechanism by which endosome escape is accomplished, via of AAV [6]. Vectors such as lentiviruses, HSV, and others
naturally occurring viruses or by chemical agents incorpo- rely upon the natural physiology of the parent virus to in-
rated into nanoparticles. The ­process remains incompletely tegrate the genetic payload into host cell chromosomes.
Genetic Manipulation Via Gene Transfer Chapter | 6   79

This ­integration has the appearance of being random, or at target. One can imagine that expression of a therapeutic
least unpredictable, and presents a risk of “genotoxicity,” a transgene in a periodontal ligament fibroblast might have
phenomenon in which the integration of the genetic payload a helpful effect, but expression of the same transgene in a
occurs proximal to an oncogene. Genotoxicity has been ex- dental pulp cell could create undesired effects. While cel-
tensively observed and studied in preclinical models, and lular specificity may be needed in some applications, this is
has been conclusively linked to oncogenesis in human pa- an area of research that has not advanced beyond proof-of-
tients treated with gene therapy [12–14]. Interestingly, the principle. In this respect, tropism modified viral vectors such
genotoxic risk associated with integration of a therapeu- as alternate AAV serotypes or chimeric adenoviruses have
tic expression cassette is not matched by the expected in- shown the most promise for cell-type-targeted gene delivery.
crease in duration of expression, as integrated transgenes
are silenced within approximately the same timeframe as 6.3  HOST RESPONSE TO GENE TRANSFER
transgenes persisting as episomes. In the aggregate, these
safety concerns explain the relative preference for the non- Delivery of foreign DNA to a mammalian cell does not oc-
integrating vectors adenovirus and AAV in dental and cra- cur naturally and the few scenarios in which this phenom-
niofacial applications of gene transfer. enon occurs, such as viral infection, are harmful to the host
organism. Accordingly, mammals have evolved multiple
6.2.2.4  Matching Vector and Target layers of defense against the exogenous delivery of foreign
It is now understood that ex vivo gene transfer to stem cells genes. The specific host response activated depends on the
and in vivo or in situ gene transfer to tissues or organs are mode of delivery, with two distinct pathways predominat-
fundamentally distinct processes and require different, often ing. In the case of viral-mediated gene transfer, an activation
non-overlapping, methodologies. The epithelial origin of of cell-mediated immunity can occur, through the classi-
most therapeutically relevant dental and craniofacial struc- cal anti-viral immune response initiated by MHC Class II
tures makes them natural targets of many viral vectors, and presentation of viral antigens. In the case of non-viral gene
indeed most prior literature reporting in vivo gene transfer transfer, and in addition to the anti-viral cellular response in
to this region has relied upon viral vector technologies. In the case of viral-mediated gene transfer, a more complex and
particular, adenovirus and adeno-associated virus account less understood intracellular immune response can occur.
for the majority of preclinical gene transfer studies in dental
science, and recently have advanced to clinical trials [15]. 6.3.1  Classical Anti-Viral Response
In contrast, the vectors of choice for gene transfer to
stem cells are usually retroviruses, specifically lentivirus
to Viral-Mediated Gene Transfer
(although newer non-viral methods such as transposons and Gene therapy appropriates a portion of the natural infection
zinc finger nucleases hold promise for permanent modifi- cycle of viruses and aborts the cycle at the replication stage.
cation of stem cells). One of the most important distinc- As a result, the cell binding, endocytosis, viral uncoat-
tions between in vivo gene transfer to tissues and in vitro ing, and endosomal escape processes proceed in a manner
gene transfer to stem cells explains much of the rationale identical to a wild-type infection. Viral capsid fragments
for lentiviruses. The therapeutic promise of stem cells often are retained in the endosome, which in turn fuses with the
assumes their expansion once delivered to the recipient’s Golgi apparatus and allows these antigens to be presented
body. Cell division dilutes the effect of gene transfer with on MHC Class II receptors. Anti-viral immune response is
non-integrating viruses like Ad and AAV, because the ge- thus generated in a manner identical to wild-type infection,
netic payload delivered by these viruses persists episomally although both humoral and cell-mediated responses to gene
and is usually not replicated during mitosis. As described therapy are usually more modest (depending on the vector
above, the defining feature of retroviruses is their ability to dose) due to the absence of the uncontrolled and sustained
integrate a genetic payload into the host cell chromosomes, viremia that normally accompanies wild-type infection.
usually resulting in the transgene being propagated across The human immune system has a remarkable capacity
subsequent generations. Thus, for therapeutic strategies for immunological memory, with the result that repeat infec-
predicated upon in situ expansion of stem cells, retroviruses tions with the same virus are met with an increasingly reso-
offer this critical feature. lute and rapid response. As an example, pre-existing human
Finally, the issue of vector affinity for a target tissue rela- immunity to AAV [16] has been shown to confound human
tive to non-target tissues must often be considered. For ex- gene therapy clinical trials [17]. While repeated administra-
ample, in a complex structure such as the root of a tooth, tion of viral gene therapy vectors has not been conclusively
many cell types with distinctly different characteristics are studied in human populations, it is assumed that repeated
present. Microsurgical injection of a gene transfer vector administration of the same viral vector within the time in-
to the root of a tooth will inevitably expose the vector to a terval needed to sustain therapeutic transgene ­expression
­variety of cell types, only one of which is likely to be the (at least every few years) will be clinically infeasible. It is
80  PART | II  In Vitro Regulation of Cell Behaviour and Tissue Development

not yet clear to what extent transient immunosuppression these challenges is how to match therapeutically ­meaningful
or viral sero-/pseudo-typing will circumvent this imposing duration of transgene expression to the progress that has
hurdle to lifelong gene therapy treatments. been made in magnitude of gene expression. In many, if
not most, applications of gene therapy, transgene expression
spanning years or even decades is needed, and this require-
6.3.2  Intracellular Response
ment exceeds the capability of any available clinical grade
to Gene Transfer vector. Inevitably, many gene therapy applications will re-
The intracellular response to foreign genetic material is a quire episodic re-administration of gene transfer.
very ancient, genetically encoded innate signaling system. Considerations of host response to gene transfer are am-
The relevance and role of this multi-component immune plified when considering a paradigm that involves periodic
cascade to gene therapy is not entirely clear, due both to its re-administration. In particular, repeated priming and am-
complexity and the practical difficulty of separately evalu- plification of cell-mediated immunity presents a potentially
ating anti-viral intracellular responses from those that are dire complication, as the end result is not simply mitigation
directed purely against foreign genetic material. With the of therapeutic effects, but destruction of the target tissue it-
increasing applicability of non-viral gene transfer technolo- self. As clinical application of gene therapy moves forward,
gies such as electroporation or sonoporation, this area is strategies aimed at suppression or evasion of cell-mediated
now somewhat simpler to study and is gaining in its rel- host immune response will have to be considered. Less clear
evance to clinical translation. is the impact of repeated administration on intracellular pro-
A number of recent reviews [18–21] have summarized gramming and the native structure and function of the target
current thinking about the multiple innate immune signal- tissue. As discussed above, the intracellular response to gene
ing pathways that likely mediate the intracellular response to transfer is only beginning to be investigated and it is rea-
gene transfer. Recently, a landmark study has used microarray sonable to hope that vector improvements made in consider-
technology to probe the transcriptional changes that occur in ation of intracellular response might minimize these effects.
muscle as a result of gene electrotransfer, using electropora- Overall, serial re-administration of gene transfer represents
tion without gene transfer as a control [22]. The relatively the next chapter in gene therapy technological development.
clean experimental design of this study allowed the authors to
parse out the intracellular response to a naked DNA plasmid 6.4  EXAMPLES OF SUCCESSFUL GENETIC
in isolation from the gene transfer methodology and with- MANIPULATION VIA IN VIVO GENE
out the confounding effects of viral proteins. It remains to TRANSFER
be determined whether intracellular immune responses can
be minimized or eliminated by purging all non-human se- While the early focus on potential applications for gene trans-
quences from the vector backbone and expression cassette. fer among researchers and clinicians with a primary interest
in the dental field and head and neck area has largely focused
6.3.3  Implications for Human on promoting repair of dentoalveolar bone defects (e.g., sec-
ondary to trauma, tumor resection, or congenital deformities)
Gene Therapy [26] and periodontal regeneration [27,28] as detailed in depth
As gene transfer vectors have steadily improved over the in later chapters in this book, the potential applicability of
past two decades, human gene therapy has become a clini- gene transfer is much broader than these two areas alone.
cal reality [23]. As the gene therapy field has progressed
from proof-of-principle into the serious consideration of 6.4.1 Pre-Clinical Ex Vivo and In Vivo
clinical practicality, host response has emerged as probably
the most important obstacle to wider impact. Notably, the
Gene Transfer Strategies
most successful and visible gene therapy clinical trials to Animal and cell culture-based studies have clearly dem-
date have involved intervention in immunologically privi- onstrated the feasibility of gene transfer, both ex vivo and
leged tissues, such as the retina [24], brain [25], and the in vivo. These studies have encompassed a wide range of
immune system itself [14]. Clinical trials involving gene potential applications, from increasing the success rate
transfer to immunologically active organs, such as the liver of osseointegration by stimulating the local environment
[17], have demonstrated the challenge that the host immune into which the implants are placed (e.g., bone marrow-
response continues to pose to this promising field. derived mesenchymal stem cells induced ex vivo with
This pivot to consideration of clinical practicality is hypoxia-inducible factor-1a-expressing lentiviral vector
cause for celebration as it brings gene therapy into a new [29] to promote bone growth around the implant surgical
chapter wherein research can focus on challenges, most site), to articular cartilage repair (e.g., recombinant adeno-­
­notably host response, that heretofore were postponed pend- associated virus delivery of SOX9 to a rabbit knee joint os-
ing demonstration of clinical efficacy. At the forefront of teochondral defect model to enhance the repair of cartilage
Genetic Manipulation Via Gene Transfer Chapter | 6   81

lesions [30]), wound ­repair (e.g., delivery of bone marrow- cystectomy), by shifting the balance from scar forma-
derived mesenchymal stem cells transfected ex vivo with tion towards axonal regeneration [36].
both human vascular endothelial growth factor 165 and l Development of dental caries encompasses both be-
human beta-defensin 3 to enhance wound repair in a rat havioral and microbial components. Gene therapy ap-
combined radiation-wound injury model [31]), pulp cap- proaches could be employed to increase the production
ping/dentin regeneration (e.g., primary cultured human of anti-cariogenic salivary constituents, potentially
dental pulp cells infected with recombinant human bone reducing caries activity in high-risk individuals. Jiang
morphogenetic protein-7-­expressing adenoviral vector with et al. [37] demonstrated reduced levels of streptococ-
the goal of promoting the differentiation of human pulp cus mutans in rats following the retrograde transfer of
cells into odontoblast-like cells in order to promote dentin the gene encoding the chemokine CCL28 to rat parotid
mineralization [32]), the prevention and management of acinar cells. On the behavioral side, methamphetamine
­radiation-induced salivary hypofunction (e.g., administration abuse, in addition to its devastating social consequences,
of an adenoviral vector overexpressing human keratinocyte is associated with extensive loss of tooth structure [38]
growth factor to murine submandibular glands by retrograde and an increased propensity to engage in high-risk be-
ductal instillation in order to reduce radiation damage to the haviors that predispose to contracting HIV infection and
salivary glands [33]) and the management of immune- other sexually transmitted diseases [39]. Chen and Chen
mediated conditions (e.g., administration of adeno-associated [40] propose using recombinant AAV-mediated gene
viral vectors expressing interleukin-27 to suppress disease transfer to achieve expression of anti-methamphetamine
severity in a murine model of Sjøgren’s syndrome [34]). antibodies, the goal being to reduce central nervous sys-
tem bioavailability of methamphetamine.
l The genodermatoses, many of which have both oral cav-
6.4.2  Future Potential Approaches to ity and extraoral head and neck manifestations, comprise
Gene Therapy Currently at Earlier a heterogeneous group of skin disorders, many resulting
Stages of Development from single gene mutations. Hence these represent po-
tentially ideal candidates for gene replacement therapy.
Numerous other prospective applications for in vivo gene
Of the more common genodermatoses affecting the head
transfer of interest to the clinician who focuses on pathologic
and neck area, a number are associated with a higher in-
processes involving the oral and maxillofacial region are at
cidence of developing benign or malignant neoplasms.
earlier stages of development. While by no means intended
These include Gardner syndrome, Peutz-Jeghers syn-
as an all-inclusive list, areas of active investigation include
drome, xeroderma pigmentosum, and nevoid basal cell
the treatment of less common oral and maxillofacial neo-
carcinoma syndrome (NBCCS). The latter results from
plasms, genetically modifying the plasticity of oral cavity-
mutations in the patched-1 gene, a down-regulator of
derived cells for regeneration of extra-oral tissues, interfering
the sonic hedgehog (Shh)-gli1 pathway, resulting in a
with unfavorable aspects of tissue repair (such as scar forma-
very high incidence of developmental cysts of the jaws
tion) with the goal of promoting regeneration, reducing caries
(specifically odontogenic keratocysts, also referred to
activity both by targeting pathogenic bacteria and behaviors
as keratocystic odontogenic tumors). NBCCS is also
associated with increased tooth decay, and treatment of vari-
associated with a predisposition to developing central
ous genodermatoses. These are discussed in sequence.
nervous system tumors (e.g., medulloblastoma) and
l As the molecular mechanisms underlying the less com- the development of hundreds to thousands of basal cell
mon primary neoplasms involving the oral cavity (e.g., carcinomas of the skin. The use of small molecular in-
malignant tumors of minor salivary glands such as ad- hibitors of the sonic hedgehog pathway has received at-
enoid cystic carcinoma) are better characterized, gene tention as a possible therapy for both late stage basal
therapy interventions to improve treatment of these neo- cell carcinomas and odontogenic keratocysts of the jaws
plasms are likely. [41]. The potential for reconstitution of patched-1 ac-
l Cells from the oral cavity, especially human periodontal tivity through gene replacement therapy may offer an
ligament-derived cells, appear to be excellent candidates additional therapeutic approach for patients with both
for ex vivo gene modification, with the goal of enhanc- NBCCS and late stage basal cell carcinomas. There are
ing expression of specific growth factors to stimulate the a number of other genodermatoses, such as the different
regeneration of non-oral tissues along specific lineages variants of inherited epidermolysis bullosa, which are
(e.g., promoting neuroregenerative and/or neurotrophic characterized by a varying propensity to form extensive
differentiation in the adult brain [35]). skin blisters, and consequently may be associated with
l Gene therapy also offers promise for neural repair of significant morbidity. The potential for gene therapy ap-
traumatic injuries to the sensory nerves (e.g., iatrogenic proaches in the management of these conditions is dis-
paresthesia following third molar surgery or mandibular cussed later in this chapter.
82  PART | II  In Vitro Regulation of Cell Behaviour and Tissue Development

6.4.3  Limitations of Pre-Clinical Studies evaluate safety; typically involving fewer than 100 subjects)
[44,45]. Until recently, successful stage 3 trials (larger size
As noted above, although there is a growing body of litera-
trials to confirm effectiveness, monitor for side effects,
ture investigating the potential benefits of gene transfer in
and to compare commonly used alternative treatments
these specific areas, to date these studies have largely been
before being made widely available to the general popu-
based on cell culture and/or animal models. There are clear,
lation) have been weighted towards the treatment of rare
and widely accepted, limitations to the translation of these
single gene Mendelian-inherited diseases. In China, there
findings to human in vivo studies. This has been further ham-
currently are two-stage four-gene therapy studies approved
pered by recent evidence that suggests that a significant mi-
for patients with advanced oral and maxillofacial squa-
nority of cell lines used in previously published studies were
mous cell carcinoma and advanced thyroid malignancies
not representative of the tissue from which they are claimed
involving adenoviral delivery of p53 protein (Gendicine,
to have been derived, as a result of both cross-­contamination
Shenzhen SiBiono GeneTech Co., Ltd., China). For a com-
between cell lines and misidentification [42]. A list of the
plete list of approved, ongoing or completed worldwide hu-
several hundred cross-contaminated or misidentified cell
man gene therapy clinical trials, the reader is directed to
lines [43] identified to date includes several purported head
http://www.abedia.com/wiley/index.html, or for trials reg-
and neck-derived cell lines (e.g., adenoid cystic carcinoma,
istered through the National Institutes of Health (NIH), the
oral squamous cell carcinoma, and papillary thyroid carci-
NIH Genetic Modification Clinical Research Information
noma cell lines). A current impetus by both funding agen-
System (GeMCRIS®) at http://www.gemcris.od.nih.gov/
cies and journals to require verification of the actual cell
Contents/GC_CT_RPT_MAKER.asp.
lines used in studies submitted for publication will obviate
Likewise, the preponderance of human clinical trials of
this concern moving forward, but some degree of prudence
in vivo gene transfer specifically involving the oral and max-
in interpreting previously published studies would seem
illofacial complex constitute stage 1 and stage 2 trials for the
warranted. These concerns notwithstanding, the use of cell
treatment of advanced oral and pharyngeal squamous cell
line and animal studies remain a prerequisite to the develop-
carcinoma and advanced thyroid carcinomas unresponsive to
ment of future in vivo human studies.
conventional surgical excision and/or ablation with radioac-
tive iodine. For example, a phase 2 multicenter trial [46] dem-
6.5  PATHWAY TO CLINICAL onstrated the technical feasibility of perioperative delivery of
IMPLEMENTATION OF THERAPEUTIC adenovirus-delivered p53 gene therapy for patients with head
GENE TRANSFER and neck squamous cell carcinoma. P53, a pro-apoptosis
tumor suppressor gene, is mutated in up to 50% of patients
Realistically, due to the large number of safety and regula- with head and neck squamous cell carcinoma. In this 12 site
tory issues involved in the use of in vivo, and perhaps to a multi-institutional study, spanning the period from March 1,
lesser extent ex vivo, gene transfer this approach is more 2003 to July 1, 2006, 13 individuals with newly diagnosed,
likely to find a role in the treatment of single gene defects previously untreated surgically resectable squamous cell car-
and multifactorial conditions associated with the greatest cinoma of the oral cavity, oropharynx, larynx, and hypophar-
potential for morbidity and mortality, i.e., malignant neo- ynx with nodal disease but without distant metastases, were
plasms such as oral squamous cell carcinoma, melanoma, given intraoperative and perioperative p53 gene therapy by
lymphoma, and adenoid cystic carcinoma. The treatment of injection into both the primary surgical site and the neck dis-
inherited conditions resulting from single gene defects in- section bed. This study highlighted some of the challenges
volves replacement of the single missing or defective gene involved in patient recruitment for gene therapy studies (the
by means of gene therapy, whereas interventions for the lat- goal of accruing 30 patients per year was not met by a fac-
ter conditions can range from approaches that interfere with tor of 10) and the complex regulatory steps involved (of 12
cell-cycle regulatory proteins involved in cancer cell pro- institutions initially recruited to participate, only five cen-
liferation, to inhibition of angiogenesis, to enhancing anti- ters were able to obtain both Institutional Review Board and
tumor cell immune activation by delivering genes coding Institutional Biosafety Committee approval, and only three
for co-stimulatory proteins. of these ultimately enrolled patients into the protocol, with
This is underscored by the available data on human gene one site taking 3 years from the time of initial regulatory
therapy. Of the 1800-plus ongoing, completed or approved ­application ­submission to receiving final approval). The au-
worldwide clinical trials involving gene therapy, 65% in- thors concluded that intraoperative p53 gene therapy, while
volve therapy for cancer [44]. Of these, the majority are at technically feasible, is “not logistically possibly when carried
stage 1 phase (60%; evaluation of safety, determination of out in a multi-institutional setting.”
a safe dosage range, and identification of side effects; typi- Other approved phase 1 and 2 trials for treating ad-
cally involving 15-30 subjects), and stage 2 (35%; larger vanced oral squamous cell carcinoma include gene therapy
study to determine potential effectiveness and to further with interferon-alpha cDNA, administration of combined
Genetic Manipulation Via Gene Transfer Chapter | 6   83

interferon-α/interleukin-12 cDNA, interleukin-2 cDNA, noted in six patients, five of whom also reported subjec-
interleukin-2-secreting semi-allogeneic human carcinoma tive improvement in their degree of oral dryness.
cell line transfected with autologous tumor DNA, HLA-B7/ l There have been scarce clinical trials involving gene
beta 2-microglobulin (Allovectin-7) cDNA, tumor necrosis therapy in the field of orthopedics, focusing primarily
factor cDNA, antisense epidermal growth factor receptor on therapies for rheumatoid arthritis, and osteoarthri-
DNA, granulocyte-macrophage colony stimulating fac- tis, and one targeting aseptic prosthetic joint loosening
tor cDNA, Herpes simplex virus thymidine kinase cDNA [49]. While there exists a substantial potential for this
administered with ganciclovir, recombinant Vesicular sto- approach in orthopedic tissue engineering and regenera-
matitis virus expressing interferon beta, HPV 16 E7 cDNA tive medicine, there has been surprisingly little in the
in HPV-16-associated head and neck cancer, attenuated way of human clinical studies in this area. This may be a
Vaccinia virus (GL-ONC1) with concurrent cisplatin and ra- reflection of the complex regulatory process required for
diotherapy, replication defective adenoviral vector express- study approval, the generally non-fatal nature of these
ing E. coli purine nucleoside phosphorylase in combination conditions, or the availability of mostly successful non-
with fludarabine monophosphate, and semi-allogeneic fi- gene based alternative therapies (e.g., osseointegrated
broblasts transfected with autologous tumor DNA. Phase 1 implants for tooth loss resulting from caries, periodontal
trials have also been approved for looking at the delivery of disease and trauma; bone morphogenetic protein; etc.).
sodium iodide symporter (NIS) using a modified measles
viral vector to treat advanced thyroid carcinomas. 6.6 CONCLUSION
While clearly in the majority, not all human clinical trials
involving in vivo gene transfer involve treatment of oral can- As gene delivery approaches are further refined, acceler-
cer. Other conditions of direct relevance to the head and neck ating the transition from the laboratory setting to direct
complex that have received regulatory approval include the clinical care, and as our knowledge of the underlying mech-
use of gene delivery for inherited connective tissue disorders, anisms involved in disease development continues to grow,
release of endogenous pain killers for intractable pain, and the potential for gene therapy to offer alternative and/or
therapy for radiation induced xerostomia, as detailed below. complementary approaches to the treatment and manage-
ment of disease processes involving the craniofacial struc-
l Specifically, this includes phase 1 and 2 trials for patients tures will continue to expand.
with the inherited blistering conditions recessive dystro-
phic epidermolysis bullosa and junctional epidermolysis REFERENCES
bullosa, conditions characterized by the development of [1] Murakami T, Sunada Y. Plasmid DNA gene therapy by electroporation:
debilitating oral lesions that cause significant pain, scar- principles and recent advances. Curr Gene Ther 2011;11(6):447–56.
ring, and, eventually, microstomia. These interventions [2] Xiao PJ, Samulski RJ. Cytoplasmic trafficking, endosomal escape, and
involve ex vivo therapy with autologous keratinocytes perinuclear accumulation of adeno-associated virus type 2 particles are
transfected with retroviral vectors expressing the human facilitated by microtubule network. J Virol 2012;86(19):10462–73.
collagen 7A1 and laminin 5 beta 3 genes, respectively. [3] Green JJ, Langer R, Anderson DG. A combinatorial polymer library
approach yields insight into nonviral gene delivery. Acc Chem Res
l A recently completed phase 1 trial utilizing intrader-
2008;41(6):749–59.
mal injection of a human preproenkephalin-expressing
[4] Scherer J, Vallee RB. Adenovirus recruits dynein by an evolution-
­replication-defective modified herpes simplex viral vec- ary novel mechanism involving direct binding to pH-primed hexon.
tor in 10 patients with moderate to severe intractable pain, Viruses 2011;3(8):1417–31.
despite treatment with more than 200 mg/day of morphine [5] Leopold PL, Crystal RG. Intracellular trafficking of adenovirus:
[47,48] demonstrated the safety of this approach. This ap- many means to many ends. Adv Drug Deliv Rev 2007;59(8):810–21.
proach, if validated in phase 3 trials, could potentially be [6] Nonnenmacher M, Weber T. Intracellular transport of recombinant
of benefit for patients with facial pain syndromes such as adeno-associated virus vectors. Gene Ther 2012;19(6):649–58.
trigeminal neuralgia and atypical facial pain, or head and [7] Pichon C, Billiet L, Midoux P. Chemical vectors for gene delivery: uptake
neck cancer patients with intractable pain. and intracellular trafficking. Curr Opin Biotechnol 2010;21(5):640–5.
l To date, the most widely documented gene therapy [8] Vaughan EE, DeGiulio JV, Dean DA. Intracellular trafficking of plas-
mids for gene therapy: mechanisms of cytoplasmic movement and
intervention in the head and neck complex involves a
nuclear import. Curr Gene Ther 2006;6(6):671–81.
phase 1 trial of an adenoviral vector encoding the hu-
[9] Tseng WC, Haselton FR, Giorgio TD. Mitosis enhances transgene
man aquaporin-1 gene delivered to 11 patients with se- expression of plasmid delivered by cationic liposomes. Biochim
vere xerostomia secondary to treatment for squamous ­Biophys Acta 1999;1445(1):53–64.
cell carcinomas involving the tongue base, tonsil, or [10] Munkonge FM, Amin V, Hyde SC, Green AM, Pringle IA, Gill
­hypopharynx [15]. In this proof-of-concept study, short- DR, et al. Identification and functional characterization of cyto-
term objective improvements in parotid gland salivary plasmic determinants of plasmid DNA nuclear import. J Biol Chem
flow rates between 60% and 540% over baseline were 2009;284(39):26978–87.
84  PART | II  In Vitro Regulation of Cell Behaviour and Tissue Development

[11] Lam AP, Dean DA. Progress and prospects: nuclear import of nonvi- [31] Xia Z, Zhang C, Zeng Y, Wang T, Ai G. Transplantation of BMSCs
ral vectors. Gene Ther 2010;17(4):439–47. expressing hVEGF165/hBD3 promotes wound healing in rats with
[12] Dropulic B. Lentiviral vectors: their molecular design, safety, and use in combined radiation-wound injury. Int Wound J 2014;293–303.
laboratory and preclinical research. Hum Gene Ther 2011;22(6):649–57. [32] Lin ZM, Qin W, Zhang NH, Xiao L, Ling JQ. Adenovirus-mediated re-
[13] Shaw KL, Kohn DB. A tale of two SCIDs. Sci Transl Med combinant human bone morphogenetic protein-7 expression promotes
2011;3(97):97ps36. differentiation of human dental pulp cells. J Endod 2007;33(8):930–5.
[14] Hacein-Bey-Abina S, Hauer J, Lim A, Picard C, Wang GP, Berry [33] Zheng C, Cotrim AP, Rowzee A, Swaim W, Sowers A, Mitchell JB,
CC, et al. Efficacy of gene therapy for X-linked severe combined et al. Prevention of radiation-induced salivary hypofunction f­ollowing
immunodeficiency. N Engl J Med 2010;363(4):355–64. hKGF gene delivery to murine submandibular glands. Clin C ­ ancer Res
[15] Baum BJ, Alevizos I, Zheng C, Cotrim AP, Liu S, McCullagh L, et al. 2011;1717(9):2842–51.
Early responses to adenoviral-mediated transfer of the ­aquaporin-1 [34] Lee BH, Carcamo WC, Chiorini JA, Peck AB, Nguyen CQ. Gene
cDNA for radiation-induced salivary hypofunction. Proc Natl Acad therapy using IL-27 ameliorates Sjögren’s syndrome-like autoim-
Sci U S A 2012;109(47):19403–7. mune exocrinopathy. Arthritis Res Ther 2012;14:R172.
[16] Boutin S, Monteilhet V, Veron P, Leborgne C, Benveniste O, Montus [35] Bueno C, Ramirez C, Rodríguez-Lozano FJ, Tabarés-Seisdedos R,
MF, et al. Prevalence of serum IgG and neutralizing factors against Rodenas M, Moraleda JM, et al. Human adult periodontal ligament-
adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy derived cells integrate and differentiate after implantation into the
population: implications for gene therapy using AAV vectors. Hum adult mammalian brain. Cell Transplant 2013;22(11):2017–28.
Gene Ther 2010;21(6):704–12. [36] Verhaagen J, Van Kesteren RE, Bossers KA, Macgillavry HD, Mason
[17] Manno CS, Pierce GF, Arruda VA, Glader B, Ragni M, Rasko JJ, MR, Smit AB. Molecular target discovery for neural repair in the
et al. Successful transduction of liver in hemophilia by AAV-Factor functional genomics era. Handb Clin Neurol 2012;109:595–616.
IX and limitations imposed by the host immune response. Nat Med [37] Jiang P, Lan J, Hu Y, Li D, Jiang G. Enhancing CCL28 expression
2006;12(3):342–7. through the gene transfer to salivary glands for controlling cariogenic
[18] Sharma S, Fitzgerald KA. Innate immune sensing of DNA. PLoS microbe. Cytokine 2012;59(1):94–9.
Pathog 2011;7(4):e1001310. [38] Saini T, Edwards PC, Kimmes N, Carroll L, Shaner J, Dowd F.
[19] Barbalat R, Ewald SE, Mouchess ML, Barton GM. Nucleic acid recogni- Etiology of xerostomia and dental caries among methamphetamine
tion by the innate immune system. Annu Rev Immunol 2011;29:185–214. abusers. Oral Health Prev Dent 2005;3(3):189–95.
[20] Barber GN. Cytoplasmic DNA, innate immune pathways. Immunol [39] Shaner JW, Kimmes N, Saini T, Edwards PC. Meth mouth: ram-
Rev 2011;243(1):99–108. pant caries in methamphetamine abusers. AIDS Patient Care STDS
[21] Rathinam VA, Fitzgerald KA. Innate immune sensing of DNA vi- 2006;20:4–8.
ruses. Virology 2011;411(2):153–62. [40] Chen YH, Chen CH. The development of antibody-based immunother-
[22] Mann CJ, Anguela XM, Montané J, Obach M, Roca C, Ruzo A, apy for methamphetamine abuse: immunization, and ­virus-mediated
et al. Molecular signature of the immune and tissue response to gene transfer approaches. Curr Gene Ther 2013;13(1):39–50.
­non-coding plasmid DNA in skeletal muscle after electrotransfer. [41] Goldberg LH, Landau JM, Moody MN, Kazakevich N, Holzer
Gene Ther 2012;19(12):1177–86. AM, Myers A. Resolution of odontogenic keratocysts of the jaw
[23] Breakthrough of the year. The runners-up. Science 2009; in basal cell nevus syndrome with GDC-0449. Arch Dermatol
326(5960):1600–7. 2011;147(7):839–41.
[24] Maguire AM, Simonelli F, Pierce EA, Pugh Jr EN, Mingozzi F, [42] Phuchareon J, Ohta Y, Woo JM, Eisele DW, Tetsu O. Genetic profil-
Bennicelli J, et al. Safety and efficacy of gene transfer for Leber’s ing reveals cross-contamination and misidentification of 6 adenoid
congenital amaurosis. N Engl J Med 2008;358(21):2240–8. cystic carcinoma cell lines: ACC2, ACC3, ACCM, ACCNS, ACCS
[25] Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt and CAC2. PLoS One 2009 June 25;4(6):e6040.
M, Kutschera I, et al. Hematopoietic stem cell gene therapy with [43] Available online at: http://standards.atcc.org/kwspub/home/the_
a lentiviral vector in X-linked adrenoleukodystrophy. Science international_cell_line_authentication_committee_iclac_/Database_
2009;326(5954):818–23. of_Cross_Contaminated_or_Misidentified_Cell_Lines.pdf; [accessed
[26] Edwards PC, Ruggiero S, Fantasia JE, Burakoff R, Moorji SE, Raz- 17.02.13].
zano P, et al. Sonic hedgehog gene enhanced tissue engineering for [44] Available online at: http://www.abedia.com/wiley/indications.php;
bone regeneration. Nature Gene Therapy 2005;12(1):75–86. [accessed 13.02.13].
[27] Edwards PC, Mason JM. Gene-enhanced tissue engineering for den- [45] Lee B, Davidson BL. Gene therapy grows into young adulthood: spe-
tal hard tissue regeneration: (2) Dentin-pulp and periodontal regen- cial review issue. Hum Mol Genet 2011;20(R1):R1.
eration. Head Face Med 2006;2:16. [46] Yoo GH, Moon J, Leblanc M, Lonardo F, Urba S, Kim H, et al. A
[28] Edwards PC, Mason JM. Dental hard tissue engineering. In: Meyer phase 2 trial of surgery with perioperative INGN 201 (Ad5CMV-
U, Meyer T, Handschel J, Wiesmann H, editors. Fundamentals of p53) gene therapy followed by chemoradiotherapy for advanced,
tissue engineering and regenerative medicine. Heidelberg, Germany: resectable squamous cell carcinoma of the oral cavity, oropharynx,
Springer; 2009. p. 345–67. hypopharynx, and larynx: report of the Southwest Oncology Group.
[29] Zou D, He J, Zhang K, Dai J, Zhang W, Wang S, et al. The Arch Otolaryngol Head Neck Surg 2009;135(9):869–74.
bone-forming effects of HIF-1α-transduced BMSCs promote
­ [47] Wolfe D, Mata M, Fink DJ. A human trial of HSV mediated gene trans-
­osseointegration with dental implant in canine mandible. PLoS One fer for the treatment of chronic pain. Gene Ther 2009;16(4):455–60.
2012;7(3):e32355. [48] Wolfe D, Marina Mata M, Fink DJ. Targeted drug delivery to the periph-
[30] Cucchiarini M, Orth P, Madry H. Direct rAAV SOX9 administration eral nervous system using gene therapy. Neurosci Lett 2012;527(2):85–9.
for durable articular cartilage repair with delayed terminal differen- [49] Evans CH, Ghivizzani SC, Robbins PD. Orthopaedic gene therapy –
tiation and hypertrophy in vivo. J Mol Med 2013;91(5):625–36. lost in translation? J Cell Physiol 2012;227(2):416–20.

You might also like