Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Current Molecular Medicine 2005, 5, 187-196 187

Telomere Dynamics in Response to Chemotherapy


N. Beeharry and D. Broccoli*

Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA
19111, USA
Abstract: The use of chemotherapy provides an essential arm in the treatment of a number of
cancers. The biological feature common to all cancerous cells that sensitizes them to
chemotherapeutic agents is their elevated division rate. Rapidly dividing cells, such as tumor cells,
are more sensitive to chemotherapeutic agents that act to initiate pathways leading to cell death, a
process enhanced in cells with compromised DNA damage responses. The toxicity accompanying
chemotherapy is due to side-effects induced in normal regenerative tissues which also have relatively
high replication rates, such as hair follicles, the hematopoietic system, the gastrointestinal system,
the germline and skin cells. While the side-effects of chemotherapy may be tolerated by the patient,
the long term impact of the cytotoxic effects of chemotherapy on healthy tissues is only now
becoming apparent. Chemotherapy-induced cytotoxicity in regenerative tissues requires multiple cell
divisions in order to reconstitute the affected tissues. At least in part as a consequence of these
extra divisions, telomeres in individuals treated with chemotherapy are shorter than age-matched
control individuals who have never been exposed to these drugs. Given the essential role of telomeres
in regulating cellular aging and chromosomal stability, it is possible that the prematurely shortened
telomeres that arise following chemotherapy may impact the long-term replicative potential of these
tissues. This review is focused on how telomeres may be modulated, directly or indirectly, by
anticancer drugs and the potential long-term consequences of accelerated telomere shortening in
healthy tissue as a result of current cancer treatment protocols.

INTRODUCTION individual. While the current data indicate that


telomere attrition is at least in part a side-effect of
Chemotherapy is successfully used to treat a
chemotherapy-induced replicative stress, an
wide range of neoplasms, including hematological
alternative possibility is that erosion of telomeric
malignancies and a variety of carcinomas. Most
sequences is a result of mechanisms acting directly
anticancer drugs work by inducing apoptosis, either
on telomere structure or telomere maintenance
through perturbation of cellular signaling pathways
pathways. For example, there is evidence
or via induction of an excessive amount of DNA
suggesting that under certain conditions telomeric
damage. Chemotherapy and radiation therapy
DNA may be a more favorable target of DNA
target tumor cells primarily on the basis that
damaging chemotherapy than bulk genomic DNA
cancerous cells have a higher mitotic index than
[4]. In this review, we summarize putative
normal cells. Since chemotherapy is damaging to
mechanisms by which chemotherapy may
cells with a high replicative rate, it is reasonable to
influence telomere dynamics in normal tissues and
expect regenerative tissues to be inadvertently
suggest potential long-term consequences for
affected by anticancer drugs. While the bone
patients requiring chemotherapeutic treatment.
marrow and gastrointestinal tract are the two tissues
that generally exhibit the greatest sensitivity to most
chemotherapeutic drugs [1], there is also data TELOMERE ATTRITION IN RESPONSE TO
reporting molecular mechanisms underlying toxic CHEMOTHERAPY
side effects of chemotherapy on hair follicles It is now evident that an unanticipated side
(reviewed by [2]) and germline cells (reviewed by effect of cytotoxic chemotherapeutic protocols is
[3]). The cell death induced in healthy tissues as a telomere attrition in healthy replicative cells. To
result of drug toxicity is countered by regenerative date, there is a paucity of studies that have
cell divisions. A growing body of evidence supports investigated the effect of chemotherapy on
the hypothesis that the tissue reconstitution telomere dynamics in regenerative tissues such as
following each round of chemotherapy is hair follicles, germline, and gastrointestinal cells. In
accompanied by telomere attrition, thereby contrast, hematopoietic cells provide a tractable
producing cells with prematurely shortened model system whereby the impact of chemotherapy
telomeres relative to the actual age of the on telomere length can be investigated because of
the relative ease and minimally invasive procedure
*Address correspondence to this author at the Department of Medical through which samples may be collected. Thus,
Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, telomere length in peripheral blood mononuclear
Philadelphia, PA 19111, USA; Tel: (215) 728-7134; Fax: (215) 728-4333; cells (PBMCs) of patients who have been exposed
E-mail: K_Broccoli@fccc.edu to myelotoxic chemotherapeutics is shorter than

1566-5240/05 $50.00+.00 © 2005 Bentham Science Publishers Ltd.


188 Current Molecular Medicine, 2005, Vol. 5, No. 2 Beeharry and Broccoli

that observed in age-matched control individuals establish the effects of myelotoxic chemotherapy
who have never been exposed to such treatments and forced regeneration upon the healthy
(see below). Complicating issues that one must be hematopoietic cells within the population.
sensitive to when analyzing telomeres of Accelerated telomere shortening of normal cells
hematopoietic cells are lineage-specific as a result of exposure to chemotherapy was first
differences in telomere dynamics and telomerase noted by Engelhardt and co-workers [10]. Telomere
activation during differentiation and clonal lengths of mononuclear cells (MNCs) and
expansion (reviewed in [5]). These differences in granulocytes from pediatric patients with acute
telomere length between lineages and through lymphocytic leukemia (ALL), acute myeloid
differentiation are paralleled by cell-type specific leukemia (AML) or solid tumors were determined
differences in telomerase regulation [6, 7]. prior to and following a course of chemotherapy.
Furthermore, the rate of telomere attrition varies Telomeres were shortened in normal hematopoietic
among the different hematopoietic lineages, with cells in all patients following chemotherapy,
greater attrition per year in T cells than in B cells regardless of tumor type. However, the extent of
[7, 8]. This makes it difficult to directly extrapolate telomere shortening after repeated cycles of
telomere attrition in a specific lineage of chemotherapy was different between the cell types.
hematopoietic cells to other cell types, much less Telomere loss in granulocytes exceeded that
other tissues. However, while tissue-specific observed in MNCs for both leukemias and solid
differences in both the sensitivity to chemotherapy tumors, with a mean sequence loss of
and telomere metabolism accompanying cellular approximately 1 kb for MNCs and 1.8 kb for
divisions are probable, it is anticipated that the data granulocytes. The type of chemotherapy that these
obtained from the hematopoietic system will be patients underwent for the treatment of their cancer
generally relevant to other regenerative tissues. was myelosuppressive, with repeated exposure
The effect of chemotherapy on telomere being followed each time by proliferation. Since
dynamics has been measured by determining the the effect of chemotherapeutic treatment on
mean telomeric restriction fragment length in telomere loss was more pronounced in granulocytes
hematopoietic cells of patients before, during and compared with MNCs, the authors concluded that
after treatment. This methodology has the benefit of the chemotherapy treatment had a greater effect on
being able to reliably correlate the induction of the myeloid than the lymphoid lineage. The
chemotherapy with any changes in telomeric variation of telomere attrition between different
restriction fragment values within individual patients hematopoietic lineages is not surprising given the
over a period of time. It is important to stress that variation in telomere dynamics among
those studies performed using bulk PBMCs will hematopoietic lineages [5]. Alternatively, cells may
contain cells of a number of lineages [7, 8] and vary in their sensitivity to other mechanisms of
thus the resulting telomere length analysis will be chemotherapy-induced telomere loss, such as that
complicated by inter-lineage variability in average induced by oxidative stress (reviewed in [11]),
telomere length. Additionally, inter-lineage discussed in more detail below.
variability in sensitivity to drug and/or regenerative In a follow-up study by the same group, the
divisions will be masked when analyzing bulk medium-term outcome of chemotherapy on
PBMCs. A final caveat for using such approaches is telomere length in pediatric patients was
that while treatment with chemotherapy may be investigated [12]. In MNCs from patients with ALL
correlated with an alteration in telomere length, telomere length was 9.8±0.9 kb at the time of
little if any, information is gained regarding diagnosis and had decreased, on average, to
underlying mechanisms. 9.0±1.4 kb at a latest follow up period of 20 months,
The studies investigating telomere dynamics in similar to the average 1 kb loss observed by
the hematopoietic system in response to Engelhardt et al. [10]. In granulocytes from the
chemotherapy can be broadly divided into those same individuals, average telomere length at
investigating effects in the context of treatment for diagnosis was 9.3±1.6 kb and decreased to 8.7±1.8
hematological malignancies and those in the kb at follow up [12], less than that observed
context of treatment for solid tumors. Analysis of previously [10]. The erosion of telomeric sequences
telomere length changes before and after exposure in these cells corresponds to a loss rate of 30-40
to chemotherapy in hematological malignancies is bp/month (360-480 bp/year), significantly higher
complicated by the mixture of tumor and normal than the range of 20-60 bp/year of erosion that
cells in the samples being tested. The mixed accompanies natural human aging in the
population of cells results in a biphasic distribution hematopoietic system [7, 10, 13]. An even greater
of telomere lengths, with the shorter population of rate of telomere loss, approximately 100 bp/month
telomeres representing those present in the tumor (1200 bp/year), was observed in the MNCs of
cells [9]. Following successful treatment, the children being treated for solid tumors Standard risk
population with shorter telomeres is reduced or ALL treatment is generally a low intensity non-
absent resulting in an apparent increase in average myleoablative regimen compared to the high
telomere length. By comparing length changes intensity chemotherapy that is used to treat solid
within the longer population of telomeres, one can tumors. The authors reasoned that since a more
Telomere Dynamics in Response to Chemotherapy Current Molecular Medicine, 2005, Vol. 5, No. 2 189

intense treatment protocol was used for patients receiving high dose therapy also received stem cell
with solid tumors as compared to treating patients support, it is possible that fewer divisions were
with ALL, the greater toxicity associated with the needed to achieve hematopoietic reconstitution.
high dose treatment underpinned the differences in These data indicate that multiple cycles of high
the effect of chemotherapy between ALL patients dose chemotherapy with peripheral blood stem cell
and solid tumor patients. re-infusion is likely to be different than repeated
doses of standard dose therapy in terms of
The relationship between chemotherapeutic
hematopoietic regeneration. Standard doses are
dosage and effect on telomere loss has been
myelosuppressive rather than myeloablative, such
directly studied by Schroder et al.[14]. In this
that regenerative requirement following each
analysis, individual patients were followed through
exposure is less than that in patients receiving high
a course of standard dose or high dose
dose regimens. Under such conditions, the extent of
combinatorial chemotherapy for treatment of breast
telomere attrition for individuals receiving standard
cancer. Leukocyte telomere length was measured
dose chemotherapy would be additive over the
by obtaining blood samples from patients directly
cycles. Myeloablative treatment requires greater
prior to the start of chemotherapy, and 5 months
proliferation in order to repopulate the tissue
after completion of therapy [14]. Similar to the two
following each cycle. Since re-infusion of
studies discussed above [10, 12], although there is a
peripheral blood stem cells containing intact
trend towards telomere shortening in individuals
telomeres occurs after each cycle, although
exposed to chemotherapy, the actual changes
potentially more severe, telomere loss may not be
measured were highly variable between individuals.
additive with additional cycles. A final possibility is
In the standard dose group of 17 patients, 9 patients
that it may be less harmful to completely ablate the
exhibited a decrease in telomere length, while 4
hematopoietic system than to induce DNA damage
patients had an increased telomere length. The
within telomeres, which may result in rapid large
range of telomere length changes following
losses of DNA due to repair reactions (see below).
standard dose chemotherapy was +0.4 to -2.2 kb.
Similarly, in the high dose group of 16 patients, 9
patients had decreased telomere length, while 5 MECHANISMS OF CHEMOTHERAPY
patients exhibited an increase in telomere length. INDUCED TELOMERE LOSS
The range of telomere length changes in patients
who received high dose chemotherapy was +0.8 to 1. Proliferation Associated Telomere Loss
-1.1 kb. By following individual patients, the raw Although there are limited studies providing
data demonstrated that some patients are more mechanistic insight as to how chemotherapeutic
affected than others by chemotherapy treatment. drugs might induce loss of telomeric DNA, there is
The information from this 'susceptible' group of evidence that telomere attrition may be a
patients is 'masked' when the total data are consequence of a number of distinct mechanisms.
amalgamated, since telomere length had only The simplest explanation is based on the fact that
decreased by on average 0.2 kb in this study. It may many anticancer drugs are toxic to the
be argued that because 52% of patients who hematopoietic system of the patient. Stem and
received standard dose treatment and 56% of progenitor hematopoietic cells will under-go a
patients who received high dose therapy showed a number of cell divisions during reconstitution of the
decrease in telomere length of leukocytes, hematopoietic system. These additional cell
resources to alleviate the potential deleterious divisions may lead to an apparent enhancement in
outcomes associated with 'artificially accelerated' telomere attrition during the time period that
telomere loss in normal tissues should be patients are undergoing treatment and recovery,
concentrated on this subset of individuals. However, relative to similarly aged individuals whose
since the mechanisms contributing to the hematopoietic systems have never been
susceptibility to telomere loss in healthy tissues challenged in this way (Fig. 1). According to this
remain unknown, targeted therapy aimed at model telomere erosion occurs at the usual rate per
subpopulations is currently not feasible. Future cell division, but this rate appears elevated per unit
studies comparing susceptible versus resistant time due to the significantly greater number of cell
patients may provide information regarding divisions that are required to repopulate the tissue
underlying pathways and allow identification of during recovery. This hypothesis is supported by
those individuals most likely to benefit from numerous studies determining telomere dynamics
approaches to modulate chemotherapy associated following bone marrow transplantation (BMT) [15-
telomere attrition. 21]). Under the conditions of BMT, proliferation is
Surprisingly, less telomere loss accompanied increased in order to achieve reconstitution of the
myeloablative treatment than standard dose hematopoietic system and there is a concomitant
treatment [14]. For example, the maximum reduction of telomere length in peripheral blood
telomere loss among patients receiving standard mononuclear cells [16, 21], T cells [17, 19],
dose chemotherapy was 2.2 kb, while in the high neutrophils [17, 20] and granulocytes [15, 18] in
dose group the maximum telomere loss measured recipients following recovery as compared to the
was only 1.1 kb. However, since the patients telomere length in the respective donors. Consistent
190 Current Molecular Medicine, 2005, Vol. 5, No. 2 Beeharry and Broccoli

with the model that the telomere loss is due to the telomeric DNA beyond that which accompanies
increased proliferation necessary to regenerate the cell division. One such possibility is by inhibition of
hematopoietic system, the accelerated loss of the telomere maintenance enzyme, telomerase. If
telomeric DNA was present only for the first year telomerase acts to attenuate sequence loss as stem
following transplant [18-20]. Furthermore, the cells differentiate to reconstitute tissues, then direct
amount of telomere reduction correlated negatively inhibition of telomerase would lead to an increased
with the number of cells infused [15, 16], again amount of sequence loss per cell division, thereby
suggesting that the amount of sequence loss was a contributing to the shorter telomeres induced in
result of the total number of cellular divisions healthy regenerative tissues exposed to
required to achieve reconstitution of the chemotherapy. While there is evidence from in vitro
hematopoietic system. Based on these data from data suggesting that certain anticancer drugs in
BMT studies, the accelerated rate of telomere clinical use may be associated with telomerase
attrition that accompanies chemotherapy would be inhibition [22, 23], this does not always hold true
predicted to be limited to the time period of during [24, 25]. In the breast adenocarcinoma cell line
which cells are exposed to drug, with the rate of (MCF-7), telomerase activity was inhibited by
attrition after removal of replicative stress returning adriamycin and 5-fluorouracil [22]. In addition,
to one that is similar to that observed during normal using the human hepatoblastoma cell line HepG2,
aging (Fig. 1). it was shown that cell survival was deceased when
treated with tamoxifen as compared to untreated
controls. At higher doses of tamoxifen, inhibition of
2. Telomerase Inhibition telomerase preceded apoptotic cell death [23]. In
There are a number of possibilities by which contrast, the anticancer drugs 5-fluorouracil, methyl
chemotherapeutic drugs may stimulate the loss of methanesulfonate, etoposide, cisplatin and

Fig. (1). Chemotherapy-associated telomere loss as a result of regenerative cell divisions. Individuals exhibit telomere loss
in many somatic cells each time a cell divides such that telomeres shorten with age (blue line). In circumstances of
extreme proliferative stress, such as that induced by the cytotoxic effects of chemotherapy, rapid telomere shortening is
observed in healthy regenerative cells of patients (red line). While the amount of telomere loss per cell division remains
similar to that accompanying normal aging, the increase in proliferation per unit time causes an apparent increase in the
rate of telomere attrition. Once chemotherapeutic treatment has ended and regeneration is compete, the increased
proliferative burden ceases, paralleled by the telomere loss rate per unit time returning to a similar level to that observed in
untreated individuals, albeit with the cells having less telomeric DNA.
Telomere Dynamics in Response to Chemotherapy Current Molecular Medicine, 2005, Vol. 5, No. 2 191

daunorubicin all failed to inhibit telomerase activity telomere would block replication through this
in nasopharyngeal carcinoma cells [24]. Further, region, leading to the attendant loss of terminal
while cisplatin caused a dose-dependent reduction DNA sequences [30].
of telomerase activity in human testicular cancer Another possibility, and one that would be
cells, other anticancer agents such as doxorubicin, consistent with this study, is that platinum
bleomycin, methotreate and melphalan were containing anti-cancer drugs, like cisplatin, may
without effect [25]. These studies are complicated cause DNA base modification [29], which could
by the use of different combinations of drugs in perturb telomere structure or function. Since a
different cell lines harboring a complex and distinct number of proteins localize to the telomere and are
series of genetic abnormalities. Further, it is unclear required to maintain telomere homeostasis, it is
how much of the observed decrease in telomerase possible to envisage that chemotherapeutic agents
activity is an indirect consequence of cell death. may cause DNA adducts in telomeric DNA,
It is possible that even partial inhibition of preventing essential telomere/protein interactions
telomerase activity may contribute to telomere and thereby resulting in perturbed telomere
shortening induced by chemotherapy. For example, maintenance. This could lead to an increased rate
during B cell differentiation, activation of of telomere degradation per cell division or
telomerase results in lengthening of telomeres in alternatively block the ability of telomerase to gain
early stages of maturation, which helps to offset access to chromosome termini. Either mechanism
overall loss incurred during the cell divisions might contribute to shortened telomeres through
accompanying the differentiation program [26, 27]. increased loss of telomeric sequences per cell
In addition, mice that are deleted for one copy of division. In addition, cisplatin-induced DNA
the catalytic subunit of telomerase, hTERT, are adducts, which may be enriched at telomeres due
unable to maintain telomere arrays at wildtype to the G-rich content [31] are preferentially
lengths indicating that the level of telomerase is recognized by DNA mismatch repair (MMR) proteins
limiting [28]. Thus, reduction of telomerase activity [32]. Repair of tracts of platinum adducts via the
induced as an unanticipated effect of chemothera- MMR pathway may result in net loss of telomeric
peutic drugs might contribute to the rate of DNA. It remains to be determined whether any of
telomere attrition observed in healthy regenerative these mechanisms contribute to the telomere
tissues during recovery from cytotoxic treatment. attrition observed in patients in vivo following
Targeting telomerase as part of a chemothera- chemotherapeutic treatment.
peutic protocol would also be predicted to
exacerbate the telomere attrition that accompanies
4. Generation of Reactive Oxygen Species (ROS)
regeneration in healthy tissues, although this has
yet to be demonstrated in the clinic. If deleterious Many classes of anticancer drugs, such as
long-term consequences accompany the chemo- anthracyclines, alkylating agents, platinum
therapy-induced, prematurely aged telomeres in containing complexes, epipodophyllotoxins and
regenerative tissues (see below), it may affect the camptothecin, generate high levels of ROS
use of telomerase inhibition as a therapeutic (reviewed in [33]). These may be produced through
strategy (Harley this issue). direct perturbation of the electron transport system
or as a consequence of activation of the apoptotic
program. Intriguingly, a number of studies support
3. Modification of Telomere Structure the hypothesis that oxidative stress plays an active
Certain drugs, like cisplatin, may directly modify role in stimulating telomere loss (reviewed in [11],
telomeric DNA thereby leading to the rapid loss of see von Zglinicki and Martin-Ruiz in this issue).
terminal sequences, through a variety of Thus, culturing cells under hyperoxic conditions is
mechanisms. The effect of cisplatin on telomeres, associated with accelerated telomere shortening
which is known to cause DNA adducts [29], was and a concomitant reduction in proliferative
examined using HeLa cells [30]. Cells cultured for potential [34, 35]. Furthermore, exposure of
one cycle in the presence of low doses of cisplatin endothelial cells to homocysteine, a pro-oxidant,
led to a loss of telomeric DNA without affecting the significantly increased the rate of telomere
replication of bulk DNA. Loss of telomeric DNA was sequence loss per cell division as compared to
followed 24 hours later by cell death, leading to the untreated cells, a process blocked by inclusion of
suggestion that the shortened telomeres might act catalase, an antioxidant that abolishes ROS [36].
as the apoptosis activating signal. The model Oxidative damage can lead to a variety of base
proposed to account for the rapid loss of terminal modifications, one of which is from guanine to 7,8-
DNA was based upon incomplete replication of dihydro-8-oxoguanine (8-OG). A DNA fragment
telomeres. Due to the absence of transcribed containing telomeric repeats was five times more
regions directly adjacent within telomeres, likely to be modified than one not containing the
transcription-coupled nucelotide excision repair GGG motif present in human telomeres, suggesting
was presumed not to be operational in repairing that 8-OG damage may be enriched within
cisplatin-induced damage to telomeric DNA. The telomeric repeats [4]. The kinetics of telomere loss
presence of unrepaired platinum adducts within the when cells are cultured under mild oxidative stress
192 Current Molecular Medicine, 2005, Vol. 5, No. 2 Beeharry and Broccoli

is consistent with unrepaired ROS-induced damage The studies investigating the effect of
within telomeric DNA inhibiting complete chemotherapy on telomere dynamics, as discussed
replication of this region, analogous to the above, did not distinguish between 'artificially
mechanism proposed above for cisplatin-induced accelerated' telomere loss as a result of
telomere shortening. Telomeres containing 8-OG hematopoietic reconstitution versus that induced
are poor substrates for telomerase in vitro [37], via modification of telomeric structure or
suggesting that oxidative damage induced at maintenance pathways by anticancer drugs. It is
telomeres may impede the ability of telomerase- likely that both processes play, in part, a causal role
dependent maintenance, thereby contributing to in the loss of telomeric DNA in normal cells after
the telomere shortening seen in patients who have treatment with chemotherapy. However, depending
undergone antineoplastic treatment. In contrast, on which pathway is favored, there may be different
more dramatic and rapid loss of telomeric DNA may outcomes. If we consider that chemotherapy-
be associated with ROS generated early during induced telomere shortening occurs as a result of
DNA-damage-induced apoptosis [38]. This telomere increased replication of regenerative cells to
shortening may play a role in amplifying the repopulate the tissue, then it is reasonable to
apoptotic signal but is unlikely to contribute directly expect telomere shortening at chromosome ends
to the production of the shorter average telomere within a given cell to occur at a similar rate in all
length seen in cancer patients who have received chromosomes. Alternatively, if we consider that
chemotherapy. chemotherapy-induced damage causes loss of
telomeric DNA through modification of telomere
In vivo studies suggest that chemotherapeutic
metabolism or structure, this process would
treatment can cause oxidative stress in
presumably act at random telomeres, potentially
hematopoietic cells. Free radicals, such as
resulting in sequence loss at only one or a few
superoxide and hydrogen peroxide were generated
chromosome ends. Although more limited in scope,
in human polymorphonuclear leukocytes from
the second pathway might be immediately
patients having undergone chemotherapy [39].
detrimental to cell survival, depending on the
Using 8-OG as a measure of DNA damage induced
extent of sequence loss. In addition, any telomere-
by oxidative stress, an increase in oxidative related
free chromosomes might be expected to
damage was found in mononuclear cells of patients
compromise genome stability and would be
with ALL following aggressive chemotherapy [40].
available to interact with other chromosomes to
Likewise, in patients treated with doxorubicin, there
instigate inappropriate fusions, possibly giving rise
was an increase in 9 different oxidation-dependent
to oncogenic mutations. In contrast, the gradual
DNA base modifications [41]. Interestingly, telomere
loss associated with proliferation may be tolerated
loss following chemotherapy is greater in patients
for a number of years before any detrimental effects
who have a polymorphism of the gene encoding
are manifested. The distinction as to which
NADPH-quinone oxidoreductase [42]. The NQO1-
mechanism acts is pertinent to the potential of
187Ser polymorphism decreases the ability of the
modulating telomere attrition (see below).
protein to protect against oxidative and free-radical
induced damage [43]. This finding is consistent
with oxidative stress contributing to the telomere FUNCTIONAL CONSEQUENCES OF 'ARTIFI-
shortening observed in hematopoietic cells after CIALLY ACCELERATED'TELOMEREATTRI-
chemotherapy. Furthermore, these data are the first TION
that directly demonstrate genetic factors that may Having thus far limited ourselves to evaluating
affect the susceptibility of a patient to suffer data regarding the effect of chemotherapy on
detectable telomere attrition in normal regenerative human telomeres, there are observations in murine
tissues as a consequence of chemotherapy. models that may shed light on potential functional
consequences of chemotherapy-induced telomere
attrition in humans (reviewed in [44]; Fig. 2). During
WHAT IS A SAFE AMOUNT OF TELOMERE serial transplantation, murine hematopoietic stem
LOSS? cells progressively lose telomeric DNA despite
The amount of telomere attrition caused in having robust telomerase activity in the stem cell
patients due to anticancer drugs is dependent on a compartment (see Zimmermann and Martens in this
number of variables. Chemotherapeutic dose [14], issue). Telomere attrition was correlated with a
cell type [10] and the presence of other contributory reduction of proliferative potential in secondary
genetic factors [42] may each play a role in recipients [45]. Similarly, hematopoietic progenitor
determining overall telomere attrition induced by cells from late generation mice deficient for the
chemotherapy. Having established a link between RNA template component of telomerase (mTERC -/-
chemotherapy treatment and telomere shortening ), had a reduced capacity to differentiate into all
in hematopoietic cells, the functional relevance of lineages following cytokine stimulation, although
telomere shortening is of particular importance immune function was not compromised in these
raising the question: “what is a safe amount of animals as measured by intravenous challenge with
telomere shortening?” Listeria [46]. While bone marrow-derived stem cells
from late generation mTERC-/- mice could still
Telomere Dynamics in Response to Chemotherapy Current Molecular Medicine, 2005, Vol. 5, No. 2 193

reconstitute the hematopoietic system of shortening in hematopoietic cells [10, 12, 14, 42,
myeloablated animals, the contribution of the 49, 50], the long-term functional outcome of
infused stem cells to the regenerated tissue was accelerated telomere loss induced by exposure to
about half that seen in cells with wildtype telomere chemotherapy remains unclear. We propose that
lengths [47]. These findings provide evidence that the telomere attrition observed in hematopoietic
although the ability of the hematopoietic system to cells of BMT recipients is analogous to that
regenerate vastly exceeds the requirements accompanying chemotherapy, allowing extrapola-
associated with normal lifespan, excessive telomere tion from one system to the other. Lewis et al. [21]
attrition may compromise the degree of concurrently measured both telomere lengths and
regeneration and the ability of some mature readouts of immune function in BMT recipients and
lymphocytes to proliferate. Similarly, liver compared these to the functional measurements
regeneration in the mTERC-/- mouse was obtained from the respective donors. For all donor-
compromised following partial hepatectomy [48], recipient pairs, there was complete engraftment
suggesting that shortened telomeres affect the and reconstitution of the hematopoietic system as
regenerative potential of tissues in addition to the measured using a short tandem repeat locus. It was
hematopoietic system. shown that T-cell proliferation following phytohem-
agglutinin stimulation was severely impaired in
While in vivo studies provide evidence that
transplant recipients as compared to their
chemotherapeutic treatment may lead to telomere

Fig. (2). Chemotherapy-induced telomere attrition in regenerative tissues may have long-term consequences. Following
chemotherapy treatment, regenerative tissues undergo multiple cell divisions in order to repopulate the system, which
causes appreciable telomere loss. Excessively short telomeres may induce cellular senescence or genome instability.
Genome instability can precipitate a number of different outcomes depending on cell type and the status of genome
monitoring and DNA damage response pathways. Induction of cellular senescence and/or activation of apoptosis may lead
to compromised immune function, while telomere dysfunction induced mutations may contribute to the initiation of
secondary malignancies.
194 Current Molecular Medicine, 2005, Vol. 5, No. 2 Beeharry and Broccoli

respective donors. Similarly, the ability of myeloid 12, 42, 49]. Depending on the cell type, this
progenitor cells to differentiate in response to amount of telomere loss corresponds to an
cytokine stimulation was attenuated [21]. These additional aging of 20-40 years. Shortened
results are similar to observations made in murine telomeres in elderly individuals may contribute to
models whereby no obvious dysfunction was attenuation of the ability to mount an immune
apparent in the mature hematopoietic cell response (reviewed in [58], see von Zglinicki and
compartment but where impaired proliferative Martin-Ruiz in this issue) which may manifest
ability could be observed when the cells were significantly earlier in individuals whose immune
challenged in functional assays [46]. systems are prematurely aged following
chemotherapy. This scenario may not affect adults
Aplastic anaemia (AA) is a condition that results
having undergone chemotherapy since the
from a compromised number of hematopoietic stem
reduction in replicative potential of hematopoietic
cells (reviewed by [51]). This places extra
cells may not be manifested during the lifespan of
replicative demand on the remaining viable stem
the individual. However, rare cases of shortened
cells that, in turn, results in distinctly shorter
telomeres associated with graft failure suggest that
telomeres in PBMCs from individuals with AA as
telomere length in human hematopoietic stem cells
compared to age-matched controls [52]. Patients
may limit regenerative potential. In addition,
with AA are at greater risk to develop
genetic influences put some patients at greater risk
myleodysplastic syndrome (MDS), paroxysmal
to chemotherapy-induced telomere loss [42, 59],
nocturnal haemoglobinuria [53] or acute leukemia
making it important to establish the parameters that
(reviewed in [54]). It has been suggested that the
define sensitivity to chemotherapy-induced
replicative stress and resulting shorter telomeres
telomere attrition.
present in AA directly contribute to the
development of these clonal disorders [55],
although this has yet to be proven. STRATEGIES TO ATTENUATE CHEMO-
THERAPY-INDUCED TELOMERE LOSS
We suggest that the replicative stress induced by
chemotherapeutic treatment in patients as a result The studies discussed above provide in vivo
of tissue reconstitution, may place some patients at evidence that myelosuppressive or myeloablative
risk of developing secondary complications, akin to cancer treatment protocols impact on the telomere
those observed in patients with AA. This idea is length of cells constituting the hematopoietic
supported by observations demonstrating that in system. This concept predicts that the bystander
some patients following chemotherapy there is an effect of chemotherapy on hematopoietic cells
increased risk in developing secondary would be diminished if telomere attrition could be
malignancies (reviewed by [56]). The most reported attenuated. In support of this model is a study that
secondary cancer following chemotherapy is evaluated the effects of granulocyte colony-
leukemia and the associated MDS. What remains to stimulating factor (G-CSF) on lymphoma patients
be clarified is whether telomere integrity is related receiving chemotherapy [50]. G-CSF was shown to
to the occurrence of secondary malignancies. It is increase telomerase activity in hematopoietic
likely that telomere dysfunction may be a risk factor progenitor cells expressing the CD34+ antigen that
to developing a secondary cancer, given the were isolated from bone marrow and peripheral
essential role of telomeres in maintaining blood. The key findings were made in patients
chromosomal stability (reviewed in [57] and see having undergone chemotherapy. MNC telomere
Greenberg in this issue). Although forced telomere- length was decreased significantly in patients after
based crisis in murine models can contribute to the two cycles of treatment. The range of telomere loss
development of cancer in a permissive genetic measured among 5 patients was 200bp-1500bp in
background, studies are still needed to explore accordance with other studies [10, 12, 42, 49].
whether telomeres are abrogated to such an extent However, in the 5 patients that received G-CSF after
during chemotherapy treatment to treat an initial chemotherapy treatment, MNC telomere length was
cancer that the patient is then predisposed to extended in all patients, increasing by 300-3100bp.
developing secondary malignancies. The simplest explanation of these data is that the
G-CSF-induced increase in telomerase activity
The potential long-term outcome of patients resulted in telomere length preservation, an
having received chemotherapy depends on a unanticipated beneficial effect of G-CSF. However,
number of theoretical predictions regarding rates of it is also possible that G-CSF specifically stimulates
telomere loss. It is likely that the accelerated loss of proliferation of a subpopulation of stem or
telomeric sequences is only evident during progenitor cells with longer telomeres leading to an
reconstitution and once the tissue has repopulated, apparent increase in telomere length in MNCs.
the loss rates are normalized as has been While the use of G-CSF abolishes telomere loss in
documented in BMT studies [18-20]. Quantitative hematopoietic cells by chemotherapy, the long-
analysis of the amount of telomere loss in term consequences of G-CSF use has not been
hematopoietic cells ranges from 20-60bp/year [7, evaluated [50]. Nevertheless, this study provides
10, 13]. The measured loss of telomeric DNA in proof of principle that modulating telomerase or
patients after chemotherapy is around 1-2 kb [10, pathways governing telomere length maintenance
Telomere Dynamics in Response to Chemotherapy Current Molecular Medicine, 2005, Vol. 5, No. 2 195

may circumvent chemotherapy induced telomere LIST OF ABBREVIATIONS


loss in healthy tissues. AA = Aplastic anemia
Based on the discussion above, it seems likely ALL = Acute lymphocytic leukemia
that the generation of free radicals contributes at
least in part to chemotherapy-induced telomere loss AML = Acute myeloid leukemia
(reviewed in [60]). Since several studies show a BMT = Bone marrow transplantation
decrease in the levels of plasma antioxidants
following chemotherapy treatment [61, 62], it is DAHK = Asp-Ala-His-Lys (the four N terminal
plausible that free radical-induced telomere loss amino acids of albumin)
may be further exacerbated due to a decreased G-CSF = Granulocyte-colony stimulating factor
ability to detoxify the free radicals. This proposition
has prompted a number of investigators to examine MDS = Myleodysplastic syndrome
the potential beneficial effect of co-administering MMR = Mismatch repair
antioxidants along with chemotherapy (reviewed in
[33]) despite the potential complication that MNCs = Mononuclear cells
antioxidants may interfere with anticancer drugs mTERC-/-= Mice deficient for the RNA template
and reduce their effectiveness (reviewed in [63]). component of telomerase
Whether or not the beneficial effect of antioxidants
PBMCs = Peripheral blood mononuclear cells
will also protect cells of regenerative tissues against
chemotherapy-induced telomere loss remains to be ROS = Reactive oxygen species
addressed. TRF = Terminal restriction fragment
8-OG = 7,8-Dihydro-8-oxoguanine
SUMMARY
While the goal of chemotherapy is to maximize REFERENCES
tumor killing potential, while minimizing damage to
healthy tissues of the patient, it is evident that [1] Chu, E. and DeVita, V.T. (2001) Principles of cancer
management: chemotherapy, in Cancer: principles & practice of
cytotoxic anticancer treatments cause the oncology, V.T. DeVita, S. Hellman, and S.A. Rosenberg,Editors.
inadvertent loss of telomeric DNA in cells of normal Lippencott, Williams & Wilkins: Philadelphia, PA. pp. 289-306.
tissues. This is in part due to increased divisions of [2] Botchkarev, V.A. (2003) J. Investig. Dermatol. Symp. Proc., 8,
regenerative cells in order to repopulate the tissue 72-75.
[3] Colpi, G.M., Contalbi, G.F., Nerva, F., Sagone, P. and Piediferro,
due to the chemotherapy cytotoxicity. However, it is G. (2004) Eur. J. Obstet. Gynecol. Reprod. Biol., 113, S2-S6.
possible that there also exist a number of other [4] Kawanishi, S. and Oikawa, S. (2004) Ann. N. Y. Acad. Sci., 1019,
mechanisms through which chemotherapy 278-84.
treatment may cause telomere attrition. These are [5] Hodes, R.J., Hathcock, K.S. and Weng, N.P. (2002) Nat. Rev.
suggested to be anticancer drugs inhibiting Immunol., 2, 699-706.
[6] Weng, N.P., Granger, L. and Hodes, R.J. (1997) Proc. Natl.
telomerase, directly modifying telomeric Acad. Sci. USA, 94, 10827-10832.
sequences, or modifying factors that are responsible [7] Son, N.H., Murray, S., Yanovski, J., Hodes, R.J. and Weng, N.P.
for maintaining telomere homeostasis. In addition, (2000) J. Immunol., 165, 1191-1196.
there may be other factors, such genetic [8] Li, Y.H., Ma, S.K., Wan, T.S., Au, W.Y., Fung, L.F., Leung, A.Y.,
Lie, A.K. and Chan, L.C. (2002) Bone Marrow Transplant, 30,
background, which predispose some patients to be 475-7.
more sensitive to the unintended, yet deleterious, [9] Adamson, D.J.A., King, D.J. and Haites, N.E. (1992) Cancer
side-effects of chemotherapy. The consequences of Genet. Cytogenet., 61, 204-206.
artificially accelerated telomere loss due to [10] Engelhardt, M., Ozkaynak, M.F., Drullinsky, P., Sandoval, C.,
chemotherapy may not necessarily manifest Tugal, O., Jayabose, S. and Moore, M.A.S. (1998) Leukemia, 12,
13-24.
immediately, but may become pronounced after [11] von Zglinicki, T. (2002) Trends Biochem. Sci., 27, 339-344.
multiple chemotherapeutic regimens and/or later in [12] Franco, S., Ozkaynak, M.F., Sandoval, C., Tugal, O., Jayabose,
life. An understanding of the underlying S., Engelhardt, M. and Moore, M.A.S. (2003) Leukemia, 17, 401-
mechanisms involved in chemotherapy-induced 410.
[13] Hastie, N.D. and Allshire, R.C. (1989) Trends Genet., 5, 326-
telomere loss may help to identify patients who are 330.
especially susceptible. It is hoped that in the future [14] Schroder, C.P., Wisman, G.B.A., de Jong, S., van der Graaf,
telomere loss in the healthy regenerative of tissues W.T.A., Ruiters, M.H.J., Mulder, N.H., de Leij, L., van der Zee,
these patients may be minimized, which in turn A.G.J. and de Vries, E.G.E. (2001) Br. J. Cancer, 84, 1348-1353.
would help to circumvent deleterious long-term [15] Notaro, R., Cimmino, A., Tabarini, D., Rotoli, B. and Luzzatto, L.
(1997) Proc. Natl. Acad. Sci. USA, 94, 13782-13785.
consequences. [16] Lee, J.J., Kook, H., Chung, I.J., Kim, H.J., Park, M.R., Kim, C.J.,
Nah, J.A. and Hwang, T.J. (1999) Bone Marrow Transplant, 24,
411-415.
ACKNOWLEDGEMENTS [17] Wynn, R., Thornley, I., Freedman, M. and Saunders, E.F. (1999)
Br. J. Haematol., 105, 997-1001.
We thank the members of the laboratory for [18] Brummendorf, T.H., Rufer, N., Baerlocher, G.M., Roosnek, E.
helpful comments. The research in the Broccoli and Lansdorp, P.M. (2001) Ann. N. Y. Acad. Sci. , 938, 1-7;
laboratory is supported by CA098087-01 (NIH, DB) discussion 7-8.
and CA006927 (NIH, F.C.C.C.).
196 Current Molecular Medicine, 2005, Vol. 5, No. 2 Beeharry and Broccoli

[19] Rufer, N., Brummendorf, T.H., Chapuis, B., Helg, C., Lansdorp, [42] Fern, L., Pallis, M., Carter, G.I., Seedhouse, C., Russell, N. and
P.M. and Roosnek, E. (2001) Blood, 97, 575-577. Byrne, J. (2004) Br. J. Haematol., 126, 63-71.
[20] Robertson, J.D., Testa, N.G., Russell, N.H., Jackson, G., [43] Traver, R.D., Siegel, D., Beall, H.D., Phillips, R.M., Gibson,
Parker, A.N., Milligan, D.W., Stainer, C., Chakrabarti, S., N.W., Franklin, W.A. and Ross, D. (1997) Br. J. Cancer, 75, 69-
Dougal, M. and Chopra, R. (2001) Bone Marrow Transplant, 27, 75.
1283-6. [44] Harrington, L. (2004) Oncogene, 23, 7283-9.
[21] Lewis, N.L., Mullaney, M., Mangan, K.F., Klumpp, T., Rogatko, [45] Allsopp, R.C., Cheshier, S. and Weissman, I.L. (2001) J. Exp.
A. and Broccoli, D. (2004) Bone Marrow Transplant, 33, 71-78. Med., 193, 917-924.
[22] Ishikawa, T., Kamiyama, M., Hisatomi, H., Ichikawa, Y., [46] Lee, H.-W., Blasco, M.A., Gottlieb, G.J., Horner , J.W.N.,
Momiyama, N., Hamaguchi, Y., Hasegawa, S., Narita, T. and Greider, C.W. and DePinho, R.A. (1998) Nature, 392, 569-574.
Shimada, H. (1999) Cancer Lett., 141, 187-194. [47] Samper, E., Fernandez, P., Eguia, R., Martin-Rivera, L.,Bernad,
[23] Brandt, S., Heller, H., Schuster, K.D. and Grote, J. (2004) Liver A., Blasco, M.A. and Aracil, M. (2002) Blood, 99, 2767-75.
Int., 24, 46-54. [48] Satyanarayana, A., Wiemann, S.U., Buer, J., Lauber, J., Dittmar,
[24] Ku, W.C., Cheng, A.J. and Wang, T.C. (1997) Biochem. K.E., Wustefeld, T., Blasco, M.A., Manns, M.P. and Rudolph,
Biophys. Res. Commun., 241, 730-736. K.L. (2003) EMBO J., 22, 4003-13.
[25] Burger, A.M., Double, J.A. and Newell, D.R. (1997) Eur. J. [49] Lee, J.J., Nam, C.E., Cho, S.H., Park, K.S., Chung, I.J. and Kim,
Cancer, 33, 638-644. H.J. (2003) Ann. Hematol., 82, 492-5.
[26] Weng, N.P., Granger, L. and Hodes, R.J. (1997) Proc. Natl. [50] Szyper-Kravitz, M., Uziel, O., Shapiro, H., Radnay, J., Katz, T.,
Acad. Sci. USA, 94, 10827-32. Rowe, J.M., Lishner, M. and Lahav, M. (2003) Br. J. Haematol.,
[27] Martens, U.M., Brass, V., Sedlacek, L., Pantic, M., Exner, C., 120, 329-36.
Guo, Y., Engelhardt, M., Lansdorp, P.M., Waller, C.F. and Lange, [51] Polychronopoulou, S. and Koutroumba, P. (2004) Acta
W. (2002) Br. J. Haematol., 119, 810-8. Haematol., 111, 125-31.
[28] Erdmann, N., Liu, Y. and Harrington, L. (2004) Proc. Natl. Acad. [52] Lee, J.J., Kook, H., Chung, I.J., Na, J.A., Park, M.R., Hwang,
Sci. USA, 101, 6080-5. T.J., Kwak, J.Y., Sohn, S.K. and Kim, H.J. (2001) Br. J.
[29] Lepre, C.A., Chassot, L., Costello, C.E. and Lippard, S.J. (1990) Haematol., 112, 1025-30.
Biochemistry, 29, 811-23. [53] Tichelli, A., Gratwohl, A., Nissen, C. and Speck, B. (1994) Leuk.
[30] Ishibashi, T. and Lippard, S.J. (1998) Proc. Natl. Acad. Sci. USA, Lymphoma, 12, 167-75.
95, 4219-4223. [54] Socie, G., Rosenfeld, S., Frickhofen, N., Gluckman, E. and
[31] Oikawa, S., Tada-Oikawa, S. and Kawanishi, S. (2001) Tichelli, A. (2000) Semin Hematol., 37, 91-101.
Biochemistry, 40, 4763-4768. [55] Ball, S.E., Gibson, F.M., Rizzo, S., Tooze, J.A., Marsh, J.C. and
[32] Chaney, S.G., Campbell, S.L., Temple, B., Bassett, E., Wu, Y.B. Gordon-Smith, E.C. (1998) Blood, 91, 3582-92.
and Faldu, M. (2004) J. Inorg. Biochem., 98, 1551-1559. [56] Boice Jr. J.D. (2001) Second malignancies after chemotherapy,
[33] Conklin, K.A. (2004) Integr. Cancer Ther., 3, 294-300. in The Chemotherapy Source Book, M.C. Perry, Editor.
[34] von Zglinicki, T., Saretzki, G., Docke, W. and Lotze, C. (1995) Lippincott Williams & Wilkins: Philadelphia, PA. pp. 526-537.
Exp. Cell Res., 220, 186-193. [57] Artandi, S.E. and DePinho, R.A. (2000) Nat. Med., 6, 852-855.
[35] Lorenz, M., Saretzki, G., Sitte, N., Metzkow, S. and von [58] Akbar, A.N., Beverley, P.C. and Salmon, M. (2004) Nat. Rev.
Zglinicki, T. (2001) Free Radic. Biol. Med., 31, 824-31. Immunol., 4, 737-43.
[36] Xu, D., Neville, R. and Finkel, T. (2000) FEBS Lett., 470, 20-24. [59] Solder, B., Weiss, M., Jager, A. and Belohradsky, B.H. (1998)
[37] Szalai, V.A., Singer, M.J. and Thorp, H.H. (2002) J. Am. Chem. Clin. Pediatr. (Phila)., 37, 521-30.
Soc., 124, 1625-31. [60] Das, U. (2002) Med. Sci. Monit., 8, RA79-92.
[38] Ramirez, R., Carracedo, J., Jimenez, R., Canela, A., Herrera, [61] Clemens, M.R., Ladner, C., Schmidt, H., Ehninger, G., Einsele,
E., Aljama, P. and Blasco, M.A. (2003) J. Biol. Chem., 278, 836- H., Buhler, E., Waller, H.D. and Gey, K.F. (1989) Free Radic.
842. Res. Commun., 7, 227-32.
[39] Sangeetha, P., Das, U.N., Koratkar, R. and Suryaprabha, P. [62] Weijl, N.I., Hopman, G.D., Wipkink-Bakker, A., Lentjes, E.,
(1990) Free Radic. Biol. Med., 8, 15-9. Berger, H.M., Cleton, F.J. and Osanto, S. (1998) Ann. Oncol., 9,
[40] Kennedy, D.D., Tucker, K.L., Ladas, E.D., Rheingold, S.R., 1331-1337.
Blumberg, J. and Kelly, K.M. (2004) Am. J. Clin. Nutr., 79, 1029- [63] Lamson, D.W. and Brignall, M.S. (1999) Altern. Med. Rev., 4,
1036. 304-29.
[41] Doroshow, J.H., Synold, T.W., Somlo, G., Akman, S.A. and
Gajewski, E. (2001) Blood, 97, 2839-2845.

You might also like