Transcript Document

W I LT
an ambitious but truly un-escapable
anti-cancer therapy
Aubrey D.N.J. de Grey
Department of Genetics, University of Cambridge
Structure of this talk
1) Why a big cancer session in an aging meeting?
2) Outline of a really, I mean really, crazy idea
3) Analysis (i): Is it crazy enough to work?
4) Analysis (ii): Is it too crazy to work?
Cancer: the hardest aspect of aging
to combat???
Reason why it might be: every cancer is a moving target
the smarter we get, the smarter it gets
Treating an “inert” type of damage can be done periodically,
and the same treatment works just as well every time :-)
Treating a neoplastic type of damage selects for mutants that
resist the treatment. 6Gb of DNA is a lot to play with :-(
Is such pessimism really justified?
My take -- yes, shown by:
1) the huge variety of innovative approaches to treating
cancer that show promise but only modest efficacy
2) the inherent subtlety of the cell-biological differences
between cancer and non-cancer cells, which limits any
(?) treatment’s therapeutic index
Must we stick with the “cocktail” approach, or is
there a “clean” (even if very ambitious) solution?
Tackling genomic instability head-on
Selection can only do so much: some events are just too
unlikely, so no cells in a cancer will experience them
Problem:
Gene expression changes are not unlikely enough
What about gene existence changes?
Better acronym, anyone?
Whole-body
Interdiction of
Lengthening of
Telomeres
The proposed therapy, in a nutshell
1) Engineer (in vitro) a patient’s cells to:
a) be stem cells of each rapidly-renewing tissue
b) be deleted for telomerase and ALT genes
c) have (initially) natural-length telomeres
d) have genetic resistance to some chemotherapies
2) Introduce these cells prior to chemotherapy
3) Repeat (2) every decade or so, forever
4) Delete telomerase/ALT genes in situ in satellite cells etc.
The four options
1) Without any cancer treatment:
2) With current or foreseeable cancer treatment:
3) With uncompensated telomere maintenance deficiency:
4) With compensated telomere maintenance deficiency (WILT):
Crazy enough? Too crazy?
SENS III, December 2nd 2002, Cambridge, UK
1) Would lack of telomerase and ALT be totally protective?
Steve Artandi, Nicola Royle
2) Can cells be genetically engineered to be chemoresistant?
Leslie Fairbairn
3) Can all relevant stem cell types be transplanted?
Gerry Graham, Colin Jahoda, Charles Campbell
4) Can quiescent precursors (e.g. satellite cells) have their
telomerase/ALT genes deleted efficiently by gene therapy?
Andrew Porter
5) Can we remain healthy for a decade with no telomerase?
Inderjeet Dokal
Analysis (i):
is WILT crazy enough to work?
Cancer protection by lack of
telomerase/ALT: current status
Fallacy #1: there aren’t enough cell divisions. Log2(1012)
is indeed too few, but (a) huge rate of cell death, (b)
multi-event nature of cancer development mean that at
least a few hundred divisions precede clinical relevance.
Fallacy #2: telomere shortening is dangerous because it
causes genomic instability (which promotes cancer). It
indeed promotes cancer initiation, but it totally prevents
cancer progression once the telomeres are gone.
ALT: as clear-cut as telomerase?
The bad news:
1) it works by hijacking recombination and possibly
constitutive DNA repair systems
2) there may be many such systems that it can use
The good news:
1) some DNA repair and recombination systems are
dispensable (e.g., those involved in meiosis)
2) all ALT cancers/lines studied thus far have many
phenotypic characteristics in common
Engineering stem cell
chemoresistance: current status
Already being pursued as an anti-cancer strategy:
- Alkylating agents: Fairbairn LJ (various), etc
- Methotrexate: e.g. Patel et al., Blood 95:2356
- Cisplatin: e.g. Pradat et al., Human Gene Therapy 12:2237
- 5-FU: e.g. Yoshisue et al., Canc Chemo Pharm 46:51
Analysis (ii):
is WILT too crazy to work?
Transplantation of engineered stem
cells: current status
Blood: bone marrow transplant is routine.
Immune system: see next slide
Gut: seems very easy by surgery in mice (Tait 1994); may
be doable using endoscopy technology in humans.
Lung: being actively explored as a cystic fibrosis therapy.
Skin: the epidermis renews rapidly, but the (negligiblydividing) dermis directs its behaviour. Burns research
has exploited this (including in tissue engineering).
Immune senescence:
WILT’s Achilles heel?
Doubt #1: who needs memory cells anyway?
Elderly people (with few/sluggish naïve cells)!
Doubt #2: Maybe memory cells are sufficiently
oligoclonal that we can use the same method to
relengthen their telomeres in vitro?
Doubt #3: Maybe CMV (etc)-induced senescence
can also be avoided this way?
Mesenchymal cancers, gene targeting:
current status
Needed because we can’t dilute away the progenitor cells
when they only divide very rarely (on demand)
A promising approach: changing a gene by triggering the
cells’ homologous recombination machinery
A big plus, compared to viral (etc.) gene therapy, is that
multiple hits to the same cell are harmless (if targeted!)
Single-bp changes: many approaches (ssDNA, RNA/DNA
hybrids, triplex-forming oligonucleotides)
Big changes, e.g. deletions: target flanking sequences
Harmful effects of telomere
shortening: current status
In mice: none at all, unless engineered to have telomeres at
birth much shorter than they normally are at death
In humans: dyskeratosis congenita (DC) -- age of onset 7-8
years on average (big variance). Symptoms: as you
might guess (bone marrow failure, skin disorders,
malignancy. Mostly caused by mutations in TERC or
dyskerin (a key telomere-maintenance protein)
Stem cell therapy (bone marrow transplantation) has long
been used against DC -- despite immune problems, which
would not be relevant for WILT
Promoting stem cell longevity:
current status
Key idea:
inhibited stem cell differentiation
 increased stem cell number
 slower necessary stem cell division rate
 extended time before stem cell telomeres run out
Key regulatory genes are being discovered:
Blood: MIP-1 (Graham GJ, others)
Skin: 14-3-3 (Dellambra et al., J Cell Biol 149:1117)
What about the gut?????
The gut paradox
Literature consensus: human blood and epidermal stem
cells divide only every few months, but gut stem cells
divide once a week. Thus, other tissues might survive a
decade without telomerase but surely the gut would not.
However.... if so, why don’t DC sufferers or TERT-/mice get gut problems far sooner than anything else?
A curiosity: crypts are usually monoclonal. Why?
A possible explanation: a third form
of stem cell population dynamics
Option 1: all stem cells divide all the time (but slowly)
Option 2: “clonal selection”: one stem cell does all the
work until it fails, then another takes over. Much data
contradicts this
Option 3: most stem cells divide all the time, but a few
“ultra-stems” divide only when the “stemness” of their
neighbours falls (e.g. a stem neighbour dies), and then
usually produce an ultra-stem and a normal stem cell
few
cell
abundance
many
slow
stem
stem (50%)
progenitor (50%)
stem (rarely)
cell
div.
rate
progenitor
fast
committed
nil
differentiated
progenitor (rarely)
committed (usually)
differentiated (all)
very
few
very
slow
few
slow
abundance
many
ultrastem
stem
ultrastem (50%)
stem (50%)
stem (50%)
progenitor (50%)
stem (rarely)
cell
div.
rate
progenitor
fast
committed
nil
differentiated
progenitor (rarely)
committed (usually)
differentiated (all)
Conclusion: crazy or not?
Is it crazy enough?
Probably; the only big question is the genetics of ALT
Is it too crazy?
Maybe not: many daunting problems to solve, but all
are already at a promising stage
Is it worth pursuing now?
The more successful the other work discussed at this
meeting is, the more people will die of cancer in the
future if cancer therapy doesn’t keep up. You decide....