INFOGENETICSANAMI
EMBRYONIC STEM CELLS MIGHT PLAY A ROLE IN GENE THERAPY RESEARCH
Home
EMBRYONIC STEM CELLS MIGHT PLAY A ROLE IN GENE THERAPY RESEARCH
THE EMBRYONIC STEM CELLS
AUTOIMMUNE DISEASES AND THEPROMISE OF STEM CELL-BASED THERAPIES
CERVICAL CANCER
AIDS
STEM CELLS
ADULT STEM CELLS
HIV

With one notable exception, no therapeutic effects

have been achieved in gene therapy trials to date.

The first successful gene therapy occurred in a recent

French study in which a therapeutic transgene for

correcting X-linked severe combined immune

deficiency was introduced into the bone marrow

cells of children, resulting in improved function of their

immune systems and correction of the disease [5].

This encouraging success aside, the generally disappointing

results are due, in part, to the inherent limitations

of adult and cord blood stem cells. In principle

at least, the use of human embryonic stem cells

might overcome some of these limitations, but further

research will be needed to determine whether

embryonic stem cells are better suited to meet the

needs of gene therapy applications than are adult

stem cells.

One important feature of the optimal cell for delivering

a therapeutic transgene would be its ability to

retain the therapeutic transgene even as it proliferates

or differentiates into specialized cells. Most of the

cell-based gene therapies attempted so far have

used viral vehicles to introduce the transgene into the

hematopoietic stem cell. One way to accomplish this

is to insert the therapeutic transgene into the one of

the chromosomes of the stem cell. Retroviruses are

able to do this, and for this reason, they are often

used as the vehicle for infecting the stem cell and

introducing the therapeutic transgene into the

chromosomal DNA. However, mouse retroviruses are

only efficient at infecting cells that are actively dividing.

Unfortunately, hematopoietic stem cells are quiescent

and seldom divide. The percentage of stem

cells that actually receive the therapeutic transgene

has usually been too low to attain a therapeutic

effect. Because of this problem, investigators have

been exploring the use of viral vehicles that can

infect nondividing cells, such as lentiviruses (e.g., HIV)

or adeno-associated viruses. This approach has not

been entirely successful, however, because of

problems relating to the fact that the cells themselves

are not in an active state [13, 19].

One approach to improving the introduction of transgenes

into hematopoietic stem cells has been to

stimulate the cells to divide so that the viral vehicles

can infect them and insert the therapeutic transgene.

Inder Verma of the Salk Institute has noted, however,

that this manipulation can change other important

properties of the hematopoietic stem cells, such as

plasticity, self-renewal, and the ability to survive and

grow when introduced into the patient [23]. This possibility

might be overcome with the use of embryonic

stem cells if they require less manipulation. And in

fact, some preliminary data suggest that retroviral

vectors may work more efficiently with embryonic

stem cells than with the more mature adult stem

cells. For example, researchers have noted that retroviral

vectors introduce transgenes into human fetal

cord blood stem cells more efficiently than into cord

blood stem cells from newborns, and that the fetal

cord blood stem cells also had a higher proliferative

capacity (i.e., they underwent more subsequent cell

divisions). This suggests that fetal cord blood stem cells

might be useful in cell-based in utero gene therapy

to correct hematopoietic disorders before birth [15, 21].

In some casessuch as a treatment of a chronic

diseaseachieving continued production of the therapeutic

transgene over the life of the patient will be

very important. Generally, however, gene therapies

using hematopoietic stem cells have encountered a

phenomenon known as gene silencing,where, over

time, the therapeutic transgene gets turned offdue

to cellular mechanisms that alter the structure of the

area of the chromosome where the therapeutic

gene has been inserted [6, 7, 11, 22, 24]. Whether

the use of embryonic stem cells in gene therapy

could overcome this problem is unknown, although

preliminary evidence suggests that this phenomenon

may occur in these cells as well [8, 18].

Persistence of the cell containing the therapeutic

transgene is equally important for ensuring continued

availability of the therapeutic agent. Verma noted

that the optimal cells for cell-mediated gene transfer

would be cells that will persist for the rest of the

patients life; they can proliferate and they would

make the missing protein constantly and forever[23].

Persistence, or longevity, of the cells can come about

in two ways: a long life span for an individual cell, or

a self-renewal process whereby a short-lived cell

undergoes successive cell divisions while maintaining

the therapeutic transgene. Ideally, then, the genetically

modified cell for use in cell-based gene therapy

should be able to self-renew (in a controlled manner

so tumors are not formed) so that the therapeutic

agent is available on a long-term basis. This is one of

the reasons why stem cells are used, but adult stem

cells seem to be much more limited in the number

of times they can divide compared with embryonic

stem cells. The difference between the ability of adult

and embryonic stem cells to self-renew has been

documented in the mouse, where embryonic stems

cells were shown to have a much higher proliferative

capacity than do adult hematopoietic stem cells [25].

Researchers are beginning to understand the biological

basis of the difference in proliferative capacity

between adult and embryonic stem cells. Persistence

of cells and the ability to undergo successive cell

divisions are in part, at least, a function of the length

of structures at the tips of chromosomes called

telomeres. Telomere length is, in turn, maintained by

an enzyme known as telomerase. Low levels of

telomerase activity result in short telomeres and, thus,

fewer rounds of cell divisionin other words, shorter

longevity. Higher levels of telomerase activity result in

longer telomeres, more possible cell divisions, and

overall longer persistence. Mouse embryonic stem

cells have been found to have longer telomeres and

higher levels of telomerase activity compared with

adult stem cells and other more specialized cells in

the body. As mouse embryonic stem cells give rise to

hematopoietic stem cells, telomerase activity levels

drop, suggesting a decrease in the self-renewing

potential of the hematopoietic stem cells [3, 4]. (For

more detailed information regarding telomeres and

telomerase, see Figure C.2. Telomeres and

Telomerase.)

Human embryonic stem cells have also been shown

to maintain pluripotency (the ability to give rise to

other, more specialized cell types) and the ability to

proliferate for long periods in cell culture in the laboratory

[2]. Adult stem cells appear capable of only a

limited number of cell divisions, which would prevent

long-term expression of the therapeutic gene needed

to correct chronic diseases. Embryonic stem cells

can be maintained in culture, whereas that is nearly

impossible with cord blood stem cells,says Robert

Hawley of the American Red Cross Jerome H. Holland

Laboratory for Biomedical Sciences, who is developing

gene therapy vectors for insertion into human

hematopoietic cells [12]. So with embryonic stem

cells, you have the possibility of long-term maintenance

and expansion of cell lines, which has not

been possible with hematopoietic stem cells.

The patients immune system response can be

another significant challenge in gene therapy. Most

cells have specific proteins on their surface that allow

the immune system to recognize them as either self

or nonself.These proteins are known as major histocompatibility

proteins, or MHC proteins. If adult stem

cells for use in gene therapy cannot be isolated from

the patient, donor cells can be used. But because of

the differences in MHC proteins among individuals,

the donor stem cells may be recognized as nonself

by the patients immune system and be rejected.

John Gearhart of Johns Hopkins University and Peter

Rathjen at the University of Adelaide speculate that

embryonic stem cells may be useful for avoiding

such immune reactions [10, 20]. For instance, it may

be possible to establish an extensive bankof

embryonic stem cell lines, each with a different set

of MHC genes. Then, an embryonic stem cell that is

immunologically compatible for a patient could be

selected, genetically modified, and triggered to

develop into the appropriate type of adult stem cell

that could be administered to the patient. By genetically

modifying the MHC genes of an embryonic

stem cell, it may also be possible to create a universal

cell that would be compatible with all patients.

Another approach might be to customizeembryonic

stem cells such that cells derived from them

have a patients specific MHC proteins on their

surface and then to genetically modify them for use

in gene therapy. Such approaches are hypothetical

at this point, however, and research is needed to

assess their feasibility.

Ironically, the very qualities that make embryonic

stem cells potential candidates for gene therapy (i.e.,

pluripotency and unlimited proliferative capacity) also

raise safety concerns. In particular, undifferentiated

embryonic stem cells can give rise to teratomas,

tumors composed of a number of different tissue

types (see Chapter 10. Assessing Human Stem Cell

Safety). It may thus be preferable to use a differentiated

derivative of genetically modified embryonic

stem cells that can still give rise to a limited number

of cell types (akin to an adult stem cell). Cautions

Esmail Zanjani of the University of Nevada, We could

differentiate embryonic stem cells into, say, liver cells,

and then use them, but I dont see how we can take

embryonic stem cells per se and put genes into

them to use therapeutically[26].

Further research is needed to determine whether the

differentiated stem cells retain the advantages, such

as longer life span, of the embryonic stem cells from

which they were derived. Because of the difficulty in

isolating and purifying many of the types of adult

stem cells, embryonic stem cells may still be better

targets for gene transfer. The versatile embryonic

stem cell could be genetically modified, and then, in

theory, it could be induced to give rise to all varieties

of adult stem cells. Also, since the genetically modified

stem cells can be easily expanded, large, pure

populations of the differentiated cells could be produced

and saved. Even if the differentiated cells

were not as long-lived as the embryonic stem cells,

there would still be sufficient genetically modified

cells to give to the patient whenever the need

arises again.

Achieving clinical success with cell-based gene

therapy will require new knowledge and advances in

several key areas, including the design of viral and

nonviral vehicles for introducing transgenes into cells,

the ability to direct where in a cell the transgene is

introduced, the ability to direct the genetically modified

stem cells or the secreted therapeutic agent to

diseased tissues, optimization and regulation of the

production of the therapeutic agent within the stem

cell, and management of immune reactions to the

gene therapy process. The ability of embryonic stem

cells to generate a wide variety of specialized cell

types and being able to maintain them in the laboratory

would make embryonic stem cells a promising

model for exploring critical questions in many of

these areas.

There are possibilities of long-term maintenance and

expansion of embryonic stem cells and of differentiation

along specific lineages that have not been

possible with hematopoietic stem cells,Zanjani says.

And if they [embryonic stem cells] could be used [in

the laboratory] as a model for differentiation, you

could evaluate vectors for gene delivery and get

an idea of how genes are translated in patients.

Cynthia Dunbar, a gene therapy researcher at the

National Institutes of Health, similarly notes that

embryonic stem cells could be useful not only in

screening new viral and nonviral vectors designed to

introduce therapeutic transgenes into cells, but especially

for testing levels of production of the therapeutic

agent after the embryonic stem cells differentiate

in culture [9]. Explains Dunbar, These behaviors are

hard to predict for human cells based on animal

studies so this would be a very useful laboratory

tool.Indeed, the major contribution of embryonic

stem cells to gene therapy may be to advance the

general scientific knowledge needed to overcome

many of the current technical hurdles to successful

therapeutic gene transfer.

REFERENCES

1. Aboody, K.S., Brown, A., Rainov, N.G., Bower, K.A., Liu, S.,

Yang, W., Small, J.E., Herrlinger, U., Ourednik, V., Black, P.M.,

Breakefield, X.O., and Snyder, E.Y. (2000). Neural stem cells

display extensive tropism for pathology in adult brain:

evidence from intracranial gliomas. Proc. Natl. Acad. Sci.

U. S. A. 97, 12846-12851.