You are on page 1of 6

PERSPECTIVES

17. Moyer, A. Psychosocial outcomes of breast-conserving 29. Feeny, D. et al. Multi-attribute health-status classification distal primer. In addition to this, the single-
surgery versus mastectomy: a meta-analytic review. systems: Health Utilities Index. Pharmacoeconomics 7,
Health Psychol. 16, 284–298 (1997). 490–502 (1995). stranded GT-rich 3′ overhang at each telom-
18. Fallowfield, L. J. et al. Psychological outcomes of 30. Rabin, R. & de Charro, F. EQ-5D: a measure of health ere involves the action of a putative 5′–3′
different treatment policies in women with early breast status from the EuroQol Group. Ann. Med. 33, 337–343
cancer outside a clinical trial. BMJ 301, 575–580 (1990). (2001). exonuclease that degrades the 5′ end of the
19. Meyer, T. J. & Mark, M. M. Effects of psychosocial 31. Carr-Hill, R. Current practice in obtaining the ‘Q’ in
interventions with adult cancer patient: a meta analysis of QALYs: a cautionary note. BMJ 303, 699–701 (1991).
underhanging CA-rich strand4,5. Both of
randomised experiments. Health Psychol. 14, 101–108 32. Gelber, R. D., Goldhirsch, A. & Cole, B. F. Evaluation of these processes lead to shortening of the
(1995). effectiveness: Q-TWiST. The International Breast Cancer
20. Sheard, T. & Maguire, P. The effect of psychological Study Group. Cancer Treat. Rev. 19 (Suppl. A), 73–84
telomeric DNA template that is available for
interventions on anxiety and depression in cancer (1993). semiconservative replication in the next
patients: results of two meta-analyses. Br. J. Cancer 80, 33. Griffin, A. M. et al. On the receiving end. V: Patient
1770–1780 (1999). perceptions of the side effects of cancer chemotherapy in
round of DNA synthesis. In most normal
21. Fallowfield, L. et al. Positive epoetin alfa effect on quality 1993. Ann. Oncol. 7, 189–195 (1996). somatic cells, therefore, telomeres shorten
of life in anemic cancer patients receiving chemotherapy: 34. Lachaine, J. et al. Cost-effectiveness and quality of life
results from a randomized placebo–controlled trial. Eur. J. evaluation of ondansetron and metoclopramide for with every cell division11. Conversely, for rea-
Cancer 35, 1461 (1999). moderately emetogenic chemotherapy regimens in sons described below, prevention of telomere
22. Coates, A. & Gebski, V. On the receiving end. VI. Which breast cancer. Crit. Rev. Oncol. Hematol. 32, 105–112
dimensions of quality-of-life scores carry prognostic (1999). shortening is crucially important for the
information? Cancer Treat. Rev. 22 (Suppl. A), 63–67 (1996). 35. Velikova, G. et al. Automated collection of quality-of-life development of most human cancers.
23. Earlam, S. et al. Relation between tumor size, quality of data: a comparison of paper and computer touch-
life, and survival in patients with colorectal liver screen questionnaires. J. Clin. Oncol. 17, 998–1007 Cancers might use more than one mecha-
metastases. J. Clin. Oncol. 14, 171–175 (1996). (1999). nism to prevent telomere shortening. What
24. Maisey, N. R. et al. Baseline quality of life predicts survival 36. Fayers, P. M. & Machin, D. in Quality of Life: Assessment,
in patients with advanced colorectal cancer. Eur. J. Analysis and Interpretation 404 (John Wiley & Sons, are these mechanisms, and how does their
Cancer 38, 1351–1357 (2002). Chichester, UK, 2000). elucidation impact on our understanding of
25. Fraser, S. C. A. et al. A daily diary for quality of life
measurement in advanced breast cancer trials. Br. J. cancer biology and our ability to treat cancer?
Cancer 67, 341–346 (1993). Online links
26. Brady, M. J. et al. Reliability and validity of the Functional
Assessment of Cancer Therapy-Breast quality-of-life DATABASES Cellular immortalization
instrument. J. Clin. Oncol. 15, 974–986 (1997). Cancer.gov: http://www.cancer.gov/cancer_information/
27. Coster, S. & Fallowfield, L. The impact of endocrine breast cancer | colorectal cancer | lung cancer | melanoma
Many cancers contain cells that have an
therapy on patients with breast cancer: a review of the LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/ apparently unlimited capacity to proliferate,
literature. Breast 11, 1–12 (2002). erythropoietin-α
28. Klastersky, J. & Paesmans, M. Response to
and acquisition of this property is referred to
chemotherapy, quality of life benefits and survival in FURTHER INFORMATION as immortalization. This contrasts with the
advanced non-small cell lung cancer: review of QOLID.com:
literature results. Lung Cancer 34 (Suppl. 4), S95–S101 http://www.QOLID.com
finite proliferative capacity of normal somatic
(2001). Access to this interactive links box is free online. cells12. The early evidence that immortaliza-
tion has a key role in the cancer phenotype
came from two main lines of investigation.
The first was in vitro/in vivo studies with
OPINION hamster cells that were treated with chemical
carcinogens, in which it was found that
immortality was necessary but not sufficient
Telomere maintenance and cancer — for malignant transformation13. The second
was from studies of human cells that had
look, no telomerase been transduced with the simian virus 40
(SV40) early-region genes, which sometimes
results in immortalization. It was found that
Axel A. Neumann and Roger R. Reddel these cells could undergo malignant transfor-
mation by an activated RAS oncogene, but
Activation of a telomere maintenance Telomeric DNA and telomere-specific bind- only if they were immortalized14–16 (FIG. 2).
mechanism seems to be indispensable for ing proteins (reviewed in REF. 3), together, At that time, little was known about the
the immortalization of human cells. Most have an essential role in stabilizing chromo- molecular events in immortalization. Shortly
cancers and cancer cell lines maintain their some ends by forming a cap structure that after the discovery that normal human cells
telomeres via telomerase. In some cancers, protects chromosome ends from degradation become senescent after a limited number of
however, telomeres are maintained in the and terminal fusions. Human telomeres are cell divisions12, it was shown that cells
absence of telomerase activity by one or 5–15 kb long and are predominantly double infected with the SV40 virus could tem-
more mechanisms that are known as stranded; however, they end in a 30–200 porarily evade senescence, but that the cul-
alternative lengthening of telomeres (ALT). nucleotide single-stranded GT-rich 3′ over- ture soon entered a state that is referred to as
Successful telomere-targeted anticancer hang 4–6. This 3′ overhang has been shown to ‘crisis’ in which the population stopped
therapy might therefore require a combination form a lariat structure — referred to as a expanding17. It was subsequently shown that
of telomerase and ALT inhibitors, emphasizing telomere (T)-loop — by invading the dou- this effect of the SV40 virus could be repro-
the importance of understanding the ble-stranded region of the telomeric DNA7 duced by transfecting cells with a plasmid
molecular details of telomere maintenance (see FIG. 1). that contains the viral early region18 and that
mechanisms in immortal cells and their It was recognized in the early 1970s that encodes at least two oncoproteins, which
repression in normal cells. the known DNA polymerases would be include the SV40 large T antigen that binds
unable to replicate the very end of linear DNA to the protein products of the TP53 and RB
Telomeres are the ends of linear chromo- molecules8,9 — a phenomenon known as the genes (reviewed in REF. 19). Sometimes, a cell
somes, and in most eukaryotes they contain ‘end replication problem’10. RNA-primed expressing the SV40 early-region genes can
tandem arrays of a GT-rich nucleotide repeat DNA synthesis of the lagging strand results in bypass or escape from crisis, but this occurs
sequence — 5′TTAGGG3′ in vertebrates1,2. a terminal gap after degradation of the most in only 1 in 107 cells20,21. Although this is a

NATURE REVIEWS | C ANCER VOLUME 2 | NOVEMBER 2002 | 8 7 9


© 2002 Nature Publishing Group
PERSPECTIVES

ative cell lines was much greater than in their


TRF2 TRF2 pre-crisis counterparts and was greater than
TRF1 TRF1
TRF1
KU RAP1 KU TANK1 in normal cells (FIG. 3), indicating that the
TRF1 TANK2 telomeres had undergone extensive lengthen-
TIN2
TRF1 ing32. The increased telomere length was
maintained over hundreds of population
T-loop doublings in the persistent absence of
TRF2 TRF1
NBS1 RAD50 detectable telomerase activity 32.
MRE11 After technical reasons for this apparent
TRF1 lack of telomerase activity were excluded, it
TRF2
A1 A2-B1
was deduced that there must be at least one
D E
hnRNPs non-telomerase mechanism of telomere
5′
D-loop length maintenance 32; these mechanisms
3′ were dubbed alternative lengthening of
TRF2 TRF2 3′ telomeres (ALT)33. Definitive evidence that
5′
telomerase was not involved in ALT was pro-
Figure 1 | Telomeric DNA and protein assembly. Schematic representation of putative telomere T-loop vided by a study showing that some ALT cells
structure with telomere-specific binding proteins. The single-stranded DNA at the end of the telomere is lack expression of a subunit of telomerase
able to invade and anneal with part of the duplex DNA (thereby forming a displacement (D)-loop) in the (the RNA template, TERC), which is essential
same telomere, with the overall result being a telomere (T)-loop7. Several proteins bind specifically to for telomerase activity34. Mouse cells that are
telomeric DNA, and these recruit other proteins to the chromosome end. null for the telomerase RNA gene, Terc, are
also able to undergo immortalization35.
Although ALT was defined as any telomerase-
rare event, it is much less rare than sponta- cells proliferate11,24,25. The second was the dis- independent mechanism of telomere length
neous immortalization, which is estimated covery of the telomerase enzyme, which adds maintenance, so far there is no clear evidence
to occur in less than 1 in 1012 human cells22. telomeric DNA onto the chromosomes of for the existence of more than one ALT mech-
It therefore seems clear that expression of protozoa26. The telomeres of the SV40-trans- anism in human cells.
the SV40 genes results in increased probabil- formed cells at crisis were even shorter than at
ity of a change that is responsible for senescence, but their length stabilized post- ALT and cancer
immortalization. crisis, after telomerase activity commenced23. The original evidence for ALT came from
It was subsequently shown that transducing studies of cell lines that were immortalized
Telomerase and immortalization SV40-transformed cells with the catalytic sub- in vitro by SV40 and other means. Its rele-
The nature of the change that occurs at unit of telomerase (TERT) — which switches vance to cancer was indicated by the obser-
immortalization was elucidated in a study on telomerase activity — allows them to vation that ALT cell lines that have been
showing that SV40-transformed cells bypass crisis completely27,28 and to be trans- transduced with an activated RAS oncogene
expressed the enzyme telomerase on emer- formed by an activated RAS oncogene29. These are tumorigenic36,37. The presence of ALT in
gence from crisis, but not beforehand23. This observations showed that expression of telom- human cancer samples showed most clearly
key observation resulted from the convergence erase is a key event in the immortalization of that it is not an artefact of cell culture38. Of
of two lines of investigation. The first was the SV40-transformed cells. 57 human tumours, four were telomerase
development of the telomere hypothesis of negative and had a significantly increased
senescence — namely, that senescence is trig- Alternative lengthening of telomeres mean telomere length; the telomere length
gered when telomeres become short, and the However, some SV40-immortalized cells have was also extremely heterogeneous, which is
experimental verification of the prediction been found to be telomerase negative 30–32. characteristic of ALT cells. Similarly, 4 out of
that telomeres shorten progressively as normal The mean telomere length of telomerase-neg- 56 tumour cell lines had ALT. So, in this lim-
ited survey, 8 out of 113 (7%) tumours and
tumour cell lines had ALT 38, compared with
Activated RAS Malignant 35% of cell lines that were immortalized
oncogene transformation
in vitro 39. The reason for this discrepancy
Loss of tumour might be that most of the in vitro immortal-
suppressors ized cell lines that were examined were of
fibroblast (that is, mesenchymal) origin,
whereas most human cancers are carcino-
Activation
of TMM Unlimited
mas (that is, of epithelial origin; many nor-
Normal Cell Senescence Crisis proliferation mal epithelia have low levels of telomerase
activity). In support of this explanation,
Figure 2 | Senescence and immortalization. Normal somatic cells permanently exit from the cell cycle preliminary data indicate that sarcomas
(that is, become senescent) after a limited number of cell divisions. Cells might escape temporarily from (which are of mesenchymal origin) are
the senescence barrier if they lose the function of key tumour-suppressor genes, especially TP53 and/or
RB, but most will eventually die, at which stage the cell population is described as being in ‘crisis’. Cells
more likely to use ALT than are carcino-
might bypass crisis and become immortalized (capable of unlimited proliferation) if a telomere mas38, perhaps because carcinomas are
maintenance mechanism — telomerase or ALT — is activated. Immortalized cells, but not their mortal derived from cells that have fewer barriers to
predecessors, might be susceptible to malignant transformation by an activated oncogene such as RAS. upregulation of telomerase activity.

880 | NOVEMBER 2002 | VOLUME 2 www.nature.com/reviews/cancer


© 2002 Nature Publishing Group
PERSPECTIVES

An extensive survey of the published litera- were treated with small-molecule inhibitors of a
ture on telomerase activity in human cancer telomerase or dominant-negative TERT ALT Telomerase
concluded that 85% of all cancers are telom- mutants47–50, indicating that it is not a high
erase positive40. It cannot be assumed, how- frequency event. This might be a problem,
ever, that the remaining 15% must use an ALT however, in clinically significant tumours con-
mechanism: as argued in more detail else- taining as many as 1012 cells. It might, there-
where41, some malignancies (solid tumours fore, be necessary to develop ALT inhibitors.
and leukaemias) might not need any telomere For tumours that use both telomere mainte-
maintenance mechanism. It is possible that nance mechanisms, treatment might need to
solid tumours and leukaemias that have a rela- be initiated with a combination of telomerase
b
tively small number of crucial genetic changes, and ALT inhibitors. Both telomerase and ALT
Population

106
113
123
142
70
72
75
76
77
78
80
and/or a low cell turnover, can become clini- must access the telomere, but how this might doublings
cally significant without immortalization41. It be achieved is, at present, unknown. It would
is important to determine how many of the however, be surprising, if the sets of proteins
telomerase-negative tumours use ALT. that are involved do not at least partly overlap,
To complicate the situation further, how- so it might be possible to identify molecular
ever, it seems that some tumours use both targets for simultaneous inhibition of both
kb
telomere maintenance mechanisms38. It is not telomere maintenance mechanisms.
yet known whether ALT and telomerase are It is expected that telomerase inhibitors 48
sometimes co-expressed spontaneously within will be developed that have far fewer side
individual tumour cells, although cell-culture effects than many of the cancer chemothera-
10
experiments in which telomerase is artificially peutic agents that are available at present. The 8
switched on in ALT cells have shown, in princi- inherited syndrome dyskeratosis congenita
ple, that this is possible42–44 and that telomerase (DKC) is caused by a mutation in one of the
activity can sometimes repress or mask the components of telomerase, so individuals
ALT phenotype45. One ALT cell line was more with DKC are deficient for telomerase activity
likely to become tumorigenic if transduced (reviewed in REF. 51). The features of DKC
with expression constructs for both activated include abnormalities of the skin and nails,
RAS and TERT than if transduced with RAS and eventual failure of proliferation in the
alone46. However, it is not yet known whether bone marrow, which indicates that telomerase
ALT and telomerase are sometimes co- is required for normal proliferative capacity in Figure 3 | Telomere-length phenotype of ALT
expressed spontaneously within tumour cells. these somatic tissues. Despite this telomerase cells. a | Telomere-specific fluorescence in situ
hybridization (FISH) on metaphase chromosomes
Another possible explanation for the deficiency, onset of pancytopaenia in these
of ALT and telomerase-positive cells, illustrating
presence of both telomere maintenance individuals does not occur until a median age the highly heterogeneous telomere lengths within
mechanisms within a tumour is intratumoral of 10 years52, which indicates that it might be individual ALT cells (yellow, telomere-specific
heterogeneity. In SV40-transformed fibrob- relatively safe to administer telomerase probe; blue, DAPI-stained metaphase
lasts, the probability that telomerase or ALT inhibitors continuously for several years. chromosomes). b | Terminal restriction fragment
will become activated to allow escape from The potential toxicity of ALT inhibitors (TRF) length analysis of an ALT cell line before
crisis (at population doubling (PD) 76) and after
crisis is quite similar (ALT is activated in at cannot be predicted without knowing the role
crisis (at PD 77), showing the temporal correlation
least one-third of SV40-immortalized fibrob- of ALT in normal cells. It is important to between immortalization and occurrence of the
last lines)32. If this is the case for some types of develop an assay that can determine whether ALT characteristic telomere-length pattern.
tumour cell, then it would not be surprising if an ALT-like mechanism might be active during Reproduced with permission from REF. 37 ©
ALT and telomerase are sometimes turned on meiosis, or might modulate telomere length in American Association of Cancer Research.
in separate areas of the same tumour. somatic cells in one or more tissue compart-
ments. Interestingly, there is preliminary evi-
Implications for cancer treatment dence indicating that an ALT-like activity telomerase null by targeting the telomerase
When telomerase activity was downregulated might be responsible for lengthening telomeres RNA gene, it was found that telomeric short-
in telomerase-positive cancer cells by expres- of lymphocytes in telomerase-null mice53. ening occurred and the cells eventually died.
sion of a dominant-negative TERT mutant, Although this is an entirely speculative idea at Survivors were found to maintain their
the cells became apoptotic47,48. This showed, in present, inherited defects in the putative nor- telomeres by a mechanism that was depen-
principle, that telomerase inhibitors could be mal counterpart of ALT might also contribute dent on RAD52, which encodes a protein
very useful for treating cancer. Telomerase to features of premature ageing, which is anal- that is involved in recombination54. In the
inhibitors will not be useful, however, for the ogous to telomerase deficiency in DKC. period before activation of a survivor path-
minority of tumours that use ALT. In addi- way, proliferation was enhanced if the DNA
tion, in telomerase-positive tumours it would Lessons from yeast mismatch-repair pathway was defective55.
be predicted that effective telomerase A rational approach to inhibiting ALT A genome-wide survey of genes with altered
inhibitors will exert a very strong selection requires an understanding of the mechanism. expression after deletion of telomerase activ-
pressure for the emergence of resistant cells As is often the case in contemporary life sci- ity identified several genes that remain
that use the ALT mechanism. Activation of ences research, studies of yeast genetics have upregulated in survivors, including the puta-
ALT was not observed in cell-culture experi- pointed the way. When budding yeast — tive meiotic recombination/DNA-repair
ments in which telomerase-positive cell lines Saccharomyces cerevisiae — was made gene MSC1 (REF. 56).

NATURE REVIEWS | C ANCER VOLUME 2 | NOVEMBER 2002 | 8 8 1


© 2002 Nature Publishing Group
PERSPECTIVES

a polymerase-chain-reaction-based method
Subtelomeric Y′ was used to sequence the subtelomeric region
repeat element
of specific chromosome ends before and after
Telomere repeat shortening Telomeric G-rich immortalization of ALT cells69. Immediately
repeat
proximal to the telomere, there is a region of
the chromosome that contains a few
TTAGGG repeats that are admixed with vari-
ant repeats such as TGAGGG, TTGGGG and
Type I survivors Type II survivors
TCAGGG. According to the model, if severe
shortening of the chromosome end occurs
( )n
before crisis, then all of the telomere and part
of the subtelomeric region might be lost; after
Figure 4 | Telomerase-null Saccharomyces cerevisiae type I and type II survivors. Schematic
representation of the two telomere phenotypes in telomerase-deficient yeast (S. cerevisiae) survivors.
ALT is activated, annealing of a residual sub-
Shortening of telomeric G-rich repeats results in cell death. Rare survivors either show amplification and telomeric TTAGGG repeat with the comple-
dispersal of the subtelomeric Y′ repeat elements (blue rectangles; type I survivors) or show elongation of the mentary sequence of another telomere that
terminal G-rich sequence (red triangles; type II survivors) as a result of recombination. Adapted from REF. 83. still retains some telomeric sequence will
allow the synthesis of new telomeric
sequence. The net result would be replace-
Further analyses identified two classes of synthesized sequence can then be converted ment of variant repeats with (TTAGGG)n
telomerase-null survivors57. Type I survivors to double-stranded DNA, resulting in a net sequence; this predicted outcome was
undergo amplification of a subtelomeric increase of telomeric DNA. This is obviously observed69.
sequence named Y′, and type II survivors very different from reciprocal recombina-
have telomeres with lengths that are very het- tion, which results in exchange between a Inter-telomeric
erogeneous, but are increased overall (FIG. 4). chromosomes, but no net increase in total
Telomeres of the type II cells therefore resem- genetic material.
ble those of human ALT cells. At present, it is Further indirect evidence that the ALT
unknown whether any human cancers use an mechanism might involve recombination
ALT mechanism that is analogous to that of came from the finding that some PML bodies
type I yeast survivors. However, the chromo- in the nuclei of ALT cells are unusual37,66. PML b Intra-telomeric
some ends of telomerase-null mouse embry- bodies are found in the nuclei of most normal
onic stem cells that underwent spontaneous cells, and are domains that contain accumula-
immortalization in culture contained an tions of proteins that are involved in a wide
amplified non-telomeric sequence58. variety of functions (reviewed in REF. 67). In
The type I and type II mechanisms are every ALT cell line that has been examined so
both dependent on RAD52, but other gene far, a proportion of the cells have PML bodies
requirements vary. Type I requires RAD51, that contain telomeric DNA (at least some of
RAD54 and RAD57, whereas type II requires which is extrachromosomal), telomeric bind- c Extrachromosomal (circle)
RAD50, RAD59 and SGS1 (REFS 59–64). The ing proteins, and proteins that are involved in
protein products of all of these RAD genes are DNA replication and recombination (FIG. 6).
involved in recombination. Sgs1 is a helicase PML bodies with these contents have not been
of the RecQ class and has five human homo- found in telomerase-positive or mortal cells,
logues, three of which — WRN, BLM and so they are referred to as ALT-associated PML
RECQL4 — are known to be mutated in bodies (APBs)37. Although the function of
familial syndromes with features of prema- APBs is unknown at present, the presence of
ture ageing and increased cancer incidence recombination proteins within them — d Extrachromosomal (linear)
(Werner, Bloom, and Rothmund–Thomson including RAD52, RAD51 and RAD50 —
syndromes, respectively). confirmed the importance of testing the
recombinational model of ALT.
Recombination To test the proposed model, a DNA tag
Adding to the genetic evidence implicating was inserted into telomeres of ALT and
Figure 5 | Recombination-mediated telomere
recombination in the survival of telomerase- telomerase-positive cells68. According to the
lengthening in ALT cells. It is proposed that a
null yeast cells, Murnane and colleagues ALT model, when a tagged telomere is used by terminal telomeric DNA strand within an ALT cell
found striking increases and decreases in another telomere as a copy template and can invade other telomeric DNA and anneal to the
telomere length in a telomerase-negative when the point of strand invasion is proximal complementary strand. Synthesis of new
human cell line that they attributed to (centromeric) to the tag site, the tag will be telomeric DNA sequence can be primed on the
telomeric recombination events30. A recom- copied to the other telomere. In clonal popu- template to which the invading strand is annealed,
bination-based model for ALT was lations of ALT, but not telomerase-positive, and this can be converted to double-stranded
DNA (not shown). Potential templates include
proposed65 in which one DNA strand of a cells it was found that the number of tagged another telomere (a), the proximal (that is the
short telomere anneals to the complemen- telomeres increased during cellular prolifera- centromeric) region of the same telomere via
tary strand of another telomere and acts as a tion68. This proposed mechanism was also T-looping (b), and extrachromosomal telomeric
primer for DNA synthesis (FIG. 5a). The newly supported by the findings of a study in which DNA that is either circular (c) or linear (d).

882 | NOVEMBER 2002 | VOLUME 2 www.nature.com/reviews/cancer


© 2002 Nature Publishing Group
PERSPECTIVES

ALT cells Telomerase and ALT: interchangeable?


TRF2 PML DAPI Merge There is a sense in which the telomere
maintenance mechanisms are equivalent:
immortalization can be associated with the
activation of either telomerase or ALT, and
fully malignant tumours can use either
mechanism. It seems inadvisable, however, to
assume that they are completely equivalent,
especially if telomerase does have other func-
Telomerase-positive cells
tions in the cancer cell. Also, telomeres that
TRF2 PML DAPI Merge are maintained by telomerase or ALT might
not be functionally equivalent. Despite their
long mean telomere length, most ALT cells
contain some very short telomeres; these
might be more prone to end-to-end fusion
events than telomerase-maintained telom-
eres and so might be a source of genetic
Figure 6 | ALT-associated PML bodies. ALT cell lines and tumours contain PML bodies (multiprotein
instability 82. One of the questions that needs
complexes that are found in the nuclei of most normal cells) that have some unusual contents: telomeric to be addressed is whether tumours of the
DNA and specific telomere-binding proteins, including TRF2. PML protein is found in all PML bodies. ALT- same stage and grade that use either ALT or
associated PML bodies (APBs; yellow merge) are visualized here by co-localization of PML (red telomerase have the same prognosis. In addi-
immunostaining) and TRF2 (green). APBs are seen in ALT cells, but not in normal or telomerase-positive tion, the response of such tumours to exist-
cells, and are therefore a useful marker for ALT. With appropriate validation, it is possible that this ing therapies needs to be examined: are there
technique can be used to detect ALT in tumour specimens that have been stored as paraffin blocks,
existing cancer chemotherapeutic agents to
which is how most tumour samples are archived by pathologists.
which tumours using one or other telomere
maintenance mechanism are particularly
sensitive or resistant? If yes, there might be
Although these results strongly support cells with ALT cells and showing that the some tumour types for which it will be nec-
a recombinational mechanism for ALT, they ALT mechanism was repressed in the essary to stratify clinical trials according to
do not exclude the possibility that the hybrids74. Some telomerase-positive cells telomere maintenance mechanism. It will
telomeres might sometimes use copy tem- also contain repressors of ALT 44,74,75. An also be important to determine whether an
plates other than another telomere. It is not understanding of how this repression is individual patient’s tumour has telomerase
known whether telomeres in ALT cells form achieved might also identify useful molecu- and/or ALT activity — or neither — when
the T-loops that are observed in telom- lar targets for cancer treatment. telomere maintenance inhibitors become
erase-positive cells (FIG. 1), but if they do it available for cancer therapy.
seems possible that a telomere could use Why is telomerase needed at all? Axel A. Neumann and Roger R. Reddel are at the
itself as a copy template (FIG. 5b). They could The recombinational and DNA synthesis Cancer Research Unit, Children’s Medical Research
also use extrachromosomal telomeric DNA machinery is much more ancient than Institute, 214 Hawkesbury Road, Westmead,
— circular or linear — as a copy template telomerase. Telomerase is unique to eukary- Sydney, New South Wales 2145, Australia.
Correspondence to R.R.R.
(FIG. 5c,d). A rolling circle can template an otes, but conserved recombination proteins
e-mail: rreddel@cmri.usyd.edu.au
essentially unlimited amount of lengthen- are found in bacteria and more primitive doi: 10.1038/nrc929
ing, and there is evidence that telomerase- organisms. For example, human RAD51 has
negative yeast can use such a mechanism of structural and functional homology to the 1. Moyzis, R. K. et al. A highly conserved repetitive DNA
sequence, (TTAGGG)n, present at the telomeres of
telomere elongation70. Human ALT cells are bacterial RecA protein76. Another obvious human chromosomes. Proc. Natl Acad. Sci. USA 85,
known to contain extrachromosomal question that therefore arises is why telom- 6622–6626 (1988).
2. Blackburn, E. H. Structure and function of telomeres.
telomeric DNA 37,71,72, as are telomerase- erase is needed at all. There are a few Nature 350, 569–573 (1991).
negative insect cells73. eukaryotes that do not have telomerase, and 3. de Lange, T. Protection of mammalian telomeres.
Oncogene 21, 532–540 (2002).
maintain their telomeres, instead, by recom- 4. Wellinger, R. J., Ethier, K., Labrecque, P. & Zakian, V. A.
Repression of ALT in normal cells bination (such as mosquitoes and Evidence for a new step in telomere maintenance. Cell
85, 423–433 (1996).
ALT-mediated lengthening of telomeres midges77,78) or retrotransposition (such as 5. Makarov, V. L., Hirose, Y. & Langmore, J. P. Long G tails
at both ends of human chromosomes suggest a C strand
depends on the availability of copy tem- Drosophila and related Dipterans 79,80), but degradation mechanism for telomere shortening. Cell 88,
plates (TTAGGG repeats, which are present the overwhelming majority of eukaryotes 657–666 (1997).
6. Wright, W. E., Tesmer, V. M., Huffman, K. E., Levene, S. D.
in every telomere) and proteins that are do have telomerase. Maybe telomerase is a & Shay, J. W. Normal human chromosomes have long G-
involved in recombination and DNA syn- more readily controlled mechanism for rich telomeric overhangs at one end. Genes Dev. 11,
2801–2809 (1997).
thesis. All of these are present in normal telomere maintenance than ALT. A recent 7. Griffith, J. D. et al. Mammalian telomeres end in a large
cells, so an obvious question is why the Opinion article81 reviewed accumulating duplex loop. Cell 97, 503–514 (1999).
8. Olovnikov, A. M. [Principle of marginotomy in template
telomeres of normal somatic cells undergo data from Terc-null mice and suggested that synthesis of polynucleotides]. Doklady Akademii. Nauk.
shortening. Presumably, normal cells have a telomerase might have functions in addition SSR 201, 1496–1499 (1971).
9. Watson, J. D. Origin of concatemeric T7 DNA. Nature
mechanism for repressing telomere length to telomere maintenance that could provide New Biol. 239, 197–201 (1972).
10. Levy, M. Z., Allsopp, R. C., Futcher, A. B., Greider, C. W.
maintenance by ALT. The existence of such a a selective advantage to organisms (and & Harley, C. B. Telomere end-replication problem and cell
mechanism was shown by fusing normal cancers) that express this enzyme. aging. J. Mol. Biol. 225, 951–960 (1992).

NATURE REVIEWS | C ANCER VOLUME 2 | NOVEMBER 2002 | 8 8 3


© 2002 Nature Publishing Group
PERSPECTIVES

11. Harley, C. B., Futcher, A. B. & Greider, C. W. Telomeres 38. Bryan, T. M., Englezou, A., Dalla-Pozza, L., Dunham, M. A. 65. Reddel, R. R., Bryan, T. M. & Murnane, J. P.
shorten during ageing of human fibroblasts. Nature 345, & Reddel, R. R. Evidence for an alternative mechanism Immortalized cells with no detectable telomerase activity.
458–460 (1990). for maintaining telomere length in human tumors and A review. Biochemistry 62, 1254–1262 (1997).
12. Hayflick, L. & Moorhead, P. S. The serial cultivation of tumor-derived cell lines. Nature Med. 3, 1271–1274 66. Grobelny, J. V., Godwin, A. K. & Broccoli, D. ALT-
human diploid cell strains. Exp. Cell Res. 25, 585–621 (1997). associated PML bodies are present in viable cells and
(1961). 39. Henson, J. D., Neumann, A. A., Yeager, T. R. & Reddel, R. R. are enriched in cells in the G2/M phase of the cell cycle.
13. Newbold, R. F., Overell, R. W. & Connell, J. R. Induction Alternative lengthening of telomeres in mammalian cells. J. Cell Sci. 113, 4577–4585 (2000).
of immortality is an early prerequisite event in malignant Oncogene 21, 598–610 (2002). 67. Negorev, D. & Maul, G. G. Cellular proteins localized at
transformation of mammalian cells by carcinogens. 40. Shay, J. W. & Bacchetti, S. A survey of telomerase activity and interacting within ND10/PML nuclear bodies/PODs
Nature 299, 633–635 (1982). in human cancer. Eur. J. Cancer 33, 787–791 (1997). suggest functions of a nuclear depot. Oncogene 20,
14. O’Brien, W., Stenman, G. & Sager, R. Suppression of 41. Reddel, R. R. The role of senescence and immortalization 7234–7242 (2001).
tumor growth by senescence in virally transformed in carcinogenesis. Carcinogenesis 21, 477–484 (2000). 68. Dunham, M. A., Neumann, A. A., Fasching, C. L. &
human fibroblasts. Proc. Natl Acad. Sci. USA 83, 42. Cerone, M. A., Londono-Vallejo, J. A. & Bacchetti, S. Reddel, R. R. Telomere maintenance by recombination
8659–8663 (1986). Telomere maintenance by telomerase and by in human cells. Nature Genet. 26, 447–450 (2000).
15. Rhim, J. S. et al. Neoplastic transformation of human recombination can coexist in human cells. Hum. Mol. 69. Varley, H., Pickett, H. A., Foxon, J. L., Reddel, R. R. &
epidermal keratinocytes by AD12-SV40 and Kirsten Genet. 10, 1945–1952 (2001). Royle, N. J. Molecular characterization of inter-telomere
sarcoma viruses. Science 227, 1250–1252 (1985). 43. Grobelny, J. V., Kulp-McEliece, M. & Broccoli, D. Effects and intra-telomere mutations in human ALT cells. Nature
16. Reddel, R. R. et al. Human bronchial epithelial cells of reconstitution of telomerase activity on telomere Genet. 30, 301–305 (2002).
neoplastically transformed by v-Ki-ras: altered response maintenance by the alternative lengthening of telomeres 70. Natarajan, S. & McEachern, M. J. Recombinational
to inducers of terminal squamous differentiation. (ALT) pathway. Hum. Mol. Genet. 10, 1953–1961 (2001). telomere elongation promoted by DNA circles. Mol. Cell.
Oncogene Res. 3, 401–408 (1988). 44. Perrem, K., Colgin, L. M., Neumann, A. A., Yeager, T. R. Biol. 22, 4512–4521 (2002).
17. Girardi, A. J., Jensen, F. C. & Koprowski, H. SV40- & Reddel, R. R. Coexistence of alternative lengthening of 71. Tokutake, Y. et al. Extra-chromosome telomere repeat
induced transformation of human diploid cells: crisis and telomeres and telomerase in hTERT-transfected GM847 DNA in telomerase-negative immortalized cell lines.
recovery. J. Cell. Comp. Physiol. 65, 69–84 (1965). cells. Mol. Cell. Biol. 21, 3862–3875 (2001). Biochem. Biophys. Res. Commun. 247, 765–772
18. Murnane, J. P., Fuller, L. F. & Painter, R. B. Establishment 45. Ford, L. P. et al. Telomerase can inhibit the (1998).
and characterization of a permanent pSV ori-transformed recombination-based pathway of telomere maintenance 72. Ogino, H. et al. Release of telomeric DNA from
ataxia-telangiectasia cell line. Exp. Cell Res. 158, in human cells. J. Biol. Chem. 276, 32198–32203 (2001). chromosomes in immortal human cells lacking
119–126 (1985). 46. Stewart, S. A. et al. Telomerase contributes to telomerase activity. Biochem. Biophys. Res. Commun.
19. Bryan, T. M. & Reddel, R. R. SV40-induced tumorigenesis by a telomere length-independent 248, 223–227 (1998).
immortalization of human cells. Crit. Rev. Oncogenesis 5, mechanism. Proc. Natl Acad. Sci. USA 99, 73. Rosen, M., Kamnert, I. & Edstrom, J. E.
331–357 (1994). 12606–12611 (2002). Extrachromosomal RNA-DNA complex containing long
20. Huschtscha, L. I. & Holliday, R. Limited and unlimited 47. Zhang, X., Mar, V., Zhou, W., Harrington, L. & Robinson, M. O. telomeric repeats in chironomids. Insect Mol. Biol. 11,
growth of SV40-transformed cells from human diploid Telomere shortening and apoptosis in telomerase- 167–174 (2002).
MRC-5 fibroblasts. J. Cell Sci. 63, 77–99 (1983). inhibited human tumor cells. Genes Dev. 13, 2388–2399 74. Perrem, K. et al. Repression of an alternative mechanism
21. Shay, J. W. & Wright, W. E. Quantitation of the frequency (1999). for lengthening of telomeres in somatic cell hybrids.
of immortalization of normal human diploid fibroblasts 48. Hahn, W. C. et al. Inhibition of telomerase limits the Oncogene 18, 3383–3390 (1999).
by SV40 large T-antigen. Exp. Cell Res. 184, 109–118 growth of human cancer cells. Nature Med. 5, 75. Ishii, Y., Tsuyama, N., Maeda, S., Tahara, H. & Ide, T.
(1989). 1164–1170 (1999). Telomerase activity in hybrids between telomerase-
22. McCormick, A. & Campisi, J. Cellular aging and 49. Herbert, B.-S. et al. Inhibition of human telomerase in negative and telomerase-positive immortal human cells
senescence. Curr. Opin. Cell Biol. 3, 230–234 (1991). immortal human cells leads to progressive telomere is repressed in the different complementation groups but
23. Counter, C. M. et al. Telomere shortening associated with shortening and cell death. Proc. Natl Acad. Sci. USA 96, not in the same complementation group of immortality.
chromosome instability is arrested in immortal cells which 14276–14281 (1999). Mech. Ageing Dev. 110, 175–193 (1999).
express telomerase activity. EMBO J. 11, 1921–1929 50. Damm, K. et al. A highly selective telomerase inhibitor 76. Yu, X., Jacobs, S. A., West, S. C., Ogawa, T. &
(1992). limiting human cancer cell proliferation. EMBO J. 20, Egelman, E. H. Domain structure and dynamics in the
24. Olovnikov, A. M. A theory of marginotomy. The 6958–6968 (2001). helical filaments formed by RecA and Rad51 on DNA.
incomplete copying of template margin in enzymic 51. Collins, K. & Mitchell, J. R. Telomerase in the human Proc. Natl Acad. Sci. USA 98, 8419–8424 (2001).
synthesis of polynucleotides and biological significance of organism. Oncogene 21, 564–579 (2002). 77. Roth, C. W., Kobeski, F., Walter, M. F. & Biessmann, H.
the phenomenon. J. Theor. Biol. 41, 181–190 (1973). 52. Dokal, I. Dyskeratosis congenita in all its forms. Br. J. Chromosome end elongation by recombination in the
25. Harley, C. B., Vaziri, H., Counter, C. M. & Allsopp, R. C. Haematol. 110, 768–779 (2000). mosquito Anopheles gambiae. Mol. Cell. Biol. 17,
The telomere hypothesis of cellular aging. Exp. Gerontol. 53. Herrera, E., Martínez, C. & Blasco, M. A. Impaired 5176–5183 (1997).
27, 375–382 (1992). germinal center reaction in mice with short telomeres. 78. López, C. C., Nielsen, L. & Edström, J.-E. Terminal long
26. Greider, C. W. & Blackburn, E. H. Identification of a EMBO J. 19, 472–481 (2000). tandem repeats in chromosomes form Chironomus
specific telomere terminal transferase activity in 54. Lundblad, V. & Blackburn, E. H. An alternative pathway pallidivittatus. Mol. Cell. Biol. 16, 3285–3290 (1996).
Tetrahymena extracts. Cell 43, 405–413 (1985). for yeast telomere maintenance rescues est1- 79. Biessmann, H. et al. Addition of telomere-associated
27. Halvorsen, T. L., Leibowitz, G. & Levine, F. Telomerase senescence. Cell 73, 347–360 (1993). HeT DNA sequences ‘heals’ broken chromosome ends
activity is sufficient to allow transformed cells to escape 55. Rizki, A. & Lundblad, V. Defects in mismatch repair in Drosophila. Cell 61, 663–673 (1990).
from crisis. Mol. Cell. Biol. 19, 1864–1870 (1999). promote telomerase-independent proliferation. Nature 80. Mason, J. M. & Biessmann, H. The unusual telomeres of
28. Counter, C. M. et al. Dissociation among in vitro 411, 713–716 (2001). Drosophila. Trends Genet. 11, 58–62 (1995).
telomerase activity, telomere maintenance, and cellular 56. Nautiyal, S., DeRisi, J. L. & Blackburn, E. H. The 81. Blasco, M. A. Telomerase beyond telomeres. Nature
immortalization. Proc. Natl Acad. Sci. USA 95, genome-wide expression response to telomerase Rev. Cancer 2, 627–633 (2002).
14723–14728 (1998). deletion in Saccharomyces cerevisiae. Proc. Natl Acad. 82. Scheel, C. et al. Alternative lengthening of telomeres is
29. Hahn, W. C. et al. Creation of human tumour cells with Sci. USA 99, 9316–9321 (2002). associated with chromosomal instability in
defined genetic elements. Nature 400, 464–468 (1999). 57. Teng, S.-C. & Zakian, V. A. Telomere-telomere osteosarcomas. Oncogene 20, 3835–3844 (2001).
30. Murnane, J. P., Sabatier, L., Marder, B. A. & Morgan, W. F. recombination is an efficient bypass pathway for telomere 83. Lundblad, V. Telomere maintenance without telomerase.
Telomere dynamics in an immortal human cell line. EMBO maintenance in Saccharomyces cerevisiae. Mol. Cell. Oncogene 21, 522–531 (2002).
J. 13, 4953–4962 (1994). Biol. 19, 8083–8093 (1999).
31. Kim, N. W. et al. Specific association of human 58. Niida, H. et al. Telomere maintenance in telomerase- Acknowledgements
telomerase activity with immortal cells and cancer. deficient mouse embryonic stem cells: characterization of Research in the authors’ laboratory was supported by the
Science 266, 2011–2015 (1994). an amplified telomeric DNA. Mol. Cell. Biol. 20, Carcinogenesis Fellowship of the Cancer Council New South
32. Bryan, T. M., Englezou, A., Gupta, J., Bacchetti, S. & 4115–4127 (2000). Wales and project grants from the National Health and Medical
Reddel, R. R. Telomere elongation in immortal human 59. Teng, S.-C., Chang, J., McCowan, B. & Zakian, V. A. Research Council of Australia.
cells without detectable telomerase activity. EMBO J. 14, Telomerase-independent lengthening of yeast telomeres
4240–4248 (1995). occurs by an abrupt Rad50p-dependent, Rif-inhibited
33. Bryan, T. M. & Reddel, R. R. Telomere dynamics and recombinational process. Mol. Cell 6, 947–952 (2000). Online links
telomerase activity in in vitro immortalised human cells. 60. Le, S., Moore, J. K., Haber, J. E. & Greider, C. W. RAD50
Eur. J. Cancer 33, 767–773 (1997). and RAD51 define two pathways that collaborate to DATABASES
34. Bryan, T. M., Marusic, L., Bacchetti, S., Namba, M. & maintain telomeres in the absence of telomerase. The following terms in this article are linked online to:
Reddel, R. R. The telomere lengthening mechanism in Genetics 152, 143–152 (1999). GenBank: http://www.ncbi.nih.gov/Genbank/
telomerase-negative immortal human cells does not 61. Chen, Q., Ijpma, A. & Greider, C. W. Two survivor SV40 large T antigen
involve the telomerase RNA subunit. Hum. Mol. Genet. 6, pathways that allow growth in the absence of telomerase LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/
921–926 (1997). are generated by distinct telomere recombination events. RAS | RB | Terc | TERC | TERT | TP53
35. Hande, M. P., Samper, E., Lansdorp, P. & Blasco, M. A. Mol. Cell. Biol. 21, 1819–1827 (2001). OMIM: http://www.ncbi.nlm.nih.gov/Omim/
Telomere length dynamics and chromosomal instability in 62. Huang, P.-H. et al. SGS1 is required for telomere Bloom syndrome | dyskeratosis congenita |
cells derived from telomerase null mice. J. Cell Biol. 144, elongation in the absence of telomerase. Curr. Biol. 11, Rothmund–Thomson syndrome | Werner syndrome
589–601 (1999). 125–129 (2001). Saccharomyces Genome Database:
36. Gollahon, L. S. et al. Telomerase activity during 63. Cohen, H. & Sinclair, D. A. Recombination-mediated http://genome-www.stanford.edu/Saccharomyces/
spontaneous immortalization of Li–Fraumeni syndrome lengthening of terminal telomeric repeats requires the MSC1 | RAD50 | RAD51 | RAD52 | RAD54 | RAD57 | RAD59 |
skin fibroblasts. Oncogene 17, 709–717 (1998). Sgs1 DNA helicase. Proc. Natl Acad. Sci. USA 98, SGS1
37. Yeager, T. R. et al. Telomerase-negative immortalized 3174–3179 (2001).
human cells contain a novel type of promyelocytic 64. Johnson, F. B. et al. The Saccharomyces cerevisiae WRN FURTHER INFORMATION
leukemia (PML) body. Cancer Res. 59, 4175–4179 homolog Sgs1p participates in telomere maintenance in Genecards: http://bioinfo.weizmann.ac.il/cards/index.html
(1999). cells lacking telomerase. EMBO J. 20, 905–913 (2001). Access to this interactive links box is free online.

884 | NOVEMBER 2002 | VOLUME 2 www.nature.com/reviews/cancer


© 2002 Nature Publishing Group

You might also like