SlideShare a Scribd company logo
1 of 14
Download to read offline
See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/306401849
Does the mobilization of circulating tumour cells during cancer therapy cause
metastasis?
Article in Nature Reviews Clinical Oncology · August 2016
DOI: 10.1038/nrclinonc.2016.128
CITATIONS
144
READS
1,508
4 authors:
Olga A Martin
University of Wollongong
143 PUBLICATIONS 13,568 CITATIONS
SEE PROFILE
Robin L Anderson
Olivia Newton-John Cancer Research Institute
234 PUBLICATIONS 9,019 CITATIONS
SEE PROFILE
Kailash Narayan
Peter MacCallum Cancer Centre
124 PUBLICATIONS 2,517 CITATIONS
SEE PROFILE
Michael P Macmanus
University of Melbourne
333 PUBLICATIONS 9,527 CITATIONS
SEE PROFILE
All content following this page was uploaded by Olga A Martin on 26 June 2018.
The user has requested enhancement of the downloaded file.
Patients with the most common carcinomas, such as
those affecting the lung, prostate, bowel or breast, fre-
quently die from distant metastasis, even after curative-­
intent surgery, radiotherapy, or chemoradiotherapy
for apparently locoregionally confined disease. Distant
metastases that become apparent after successful treat-
ment to achieve locoregional control are considered to
derive from micrometastases that were present before
treatment commenced1–3
. In patients for whom attempts
to eliminate locoregionally confined disease have failed,
distant metastasis might also derive from residual local
disease4
. Another unwelcome possibility is that metastasis
can sometimes be caused or promoted by the antitumour
treatment itself. Events surrounding treatment with sur-
gery, radiotherapy, and systemic antitumour therapies can
perturb tumours in complex ways that might influence
either positively or negatively the risk of distant metas-
tasis. Evidence indicates that all three modalities might,
under some ­
circumstances, mobilize tumour cells into
the bloodstream.
Circulating tumour cells (CTCs) can be detected
in patients with all of the major carcinomas and are
necessary for distant metastasis to occur5
. In general,
although patients with high numbers of CTCs have a
poor prognosis6
, the mobilization of CTCs induced by a
therapeutic or diagnostic intervention is not necessarily
associated with an adverse outcome. Metastasis is some-
times regarded as a linear process, whereby tumour cells
acquire enhanced migratory and invasive capacities, enter
lymphatic and blood vessels, attain both the capacity to
survive the harsh environment of the circulation and the
ability to extravasate and proliferate to form a metastatic
lesion, in a new and potentially unfavourable microenvi-
ronment7
. Luzzi et al.8
reported that most CTCs can sur-
vivetheshearforcesthattheyencounterinthecirculation,
enabling them to lodge in capillary beds and often extra­
vasate into tissues. The most common outcome in such
situations is cell death, but a small proportion of tumour
cells (approximately 0.02%) survive and, after a vari­
able
period of latency, expand into clinically detectable lesions.
In a modelling study, Coumans et al.9
assessed data from a
large cohort of patients with breast cancer and estimated
thatthemetastaticrateismuchlowerthanthatreportedin
thestudydiscussed4
:onlyonemetastasisformsfromevery
60 million cells that disseminate from the primary site.
The time to first metastasis varies widely among
different malignancies, owing to their diverse natural
histories. In many patients with apparently localised
1
Division of Radiation
Oncology and Cancer
Imaging, Peter MacCallum
Cancer Centre.
2
Molecular Radiation Biology
Laboratory, Peter MacCallum
Cancer Centre.
3
Metastasis Research
Laboratory, Peter MacCallum
Cancer Centre, 305 Grattan
Street, Melbourne, Victoria
3000, Australia.
4
The Sir Peter MacCallum
Department of Oncology,
University of Melbourne.
5
Department of Obstetrics
and Gynaecology, University
of Melbourne, Grattan street,
Melbourne, Victoria 3000,
Australia.
Correspondence to M.P.M.
Michael.macmanus@
petermac.org
doi:10.1038/nrclinonc.2016.128
Published online 23 Aug 2016
Does the mobilization of circulating
tumour cells during cancer therapy
cause metastasis?
Olga A. Martin1,2,4
, Robin L. Anderson3,4
, Kailash Narayan1,4,5
and Michael P. MacManus1,4
Abstract|Despiteprogressiveimprovementsinthemanagementofpatientswithlocoregionally
confined,advanced-stagesolidtumours,distantmetastasisremainsaverycommon—andusually
fatal—modeoffailureafterattemptedcurativetreatment.Surgeryandradiotherapyarethe
primarycurativemodalitiesforthesepatients,oftencombinedwitheachotherand/orwith
chemotherapy.Distantmetastasisoccurringaftertreatmentcanarisefrompreviouslyundetected
micrometastasesor,alternatively,frompersistentlocoregionaldisease.Anotherpossibilityisthat
treatmentitselfmightsometimescauseorpromotemetastasis.Surgicalinterventionsinpatients
withcancer,includingbiopsies,arecommonlyassociatedwithincreasedconcentrationsof
circulatingtumourcells(CTCs).HighCTCnumbersareassociatedwithanunfavourableprognosis
inmanycancers.RadiotherapyandsystemicantitumourtherapiesmightalsomobilizeCTCs.We
reviewthepreclinicalandclinicaldataconcerningcancertreatments,CTCmobilizationandother
factorsthatmightpromotemetastasis.Contemporarytreatmentregimensrepresentthebest
availablecurativeoptionsforpatientswhomightotherwisediefromlocallyconfined,
advanced-stagecancers;however,ifsuchtreatmentscanpromotemetastasis,thisprocessmust
beunderstoodandaddressedtherapeuticallytoimprovepatientsurvival.
REVIEWS
32 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
cancers, metastasis occurs early, even before diagnosis3
.
This discovery has invoked an alternative parallel pro-
gression model of metastasis, whereby tumour cells that
disseminate early evolve independently, but in parallel,
from the primary tumour10,11
. Disseminated tumour
cells might lie dormant in bone marrow and/or other
tissues for many years. These occult cells can enter a
G0-like state and remain as quiescent single cells, or
they can expand to form a cluster that is unable to
expand further, possibly owing to effective immune
surveillance, or the inability to trigger angiogenesis12
.
In either case, these micrometastases are presumed to be
the source of the CTCs that are detected after primary
tumour control in many patients. Genomic analyses13,14
have reported ‘early’ and ‘late’ metastases, an observa-
tion confounded by the fact that a metastasis can seed
­secondary metastases.
Curing the most-common epithelial and mesen-
chymal malignancies is generally impossible without
loco­
regional control provided by surgery and/or radio-
therapy; however, if such potentially curative therapies
are also able to mobilize tumour cells into the circulation,
or to promote the growth of dormant micro­
metastases,
a detailed understanding of these events is essential.
This knowledge could lead to the design of new thera­
peutic approaches capable of increasing the cure rates
for ­
cancers for which metastasis is a frequent cause of
treatment failure. In this Review, we examine the pre-
clinical and clinical effects of the major modalities used
in curative treatment of epithelial cancers on the mobil­
ization of tumour cells into the circulation, and consider
other treatment-related factors that could potentially
modulate the risk of metastasis. When evaluating these
data, we have considered in vitro and animal studies sep-
arately from clinical studies, with the latter constituting a
higher level of evidence because of their direct relevance
to patients.
Circulating tumour cells
Both CTCs and cell-free tumour-derived DNA are com-
monly found in the blood of patients with carcinoma,
including those who will never develop overt metastatic
disease. This observation reflects the extremely inefficient
natureofmetastasis8
.Despitethisinefficiency,thenumber
of CTCs and cell-free tumour DNA found in the circula-
tion has both prognostic and predictive value in several
different cancer types15–18
.
CTCs escape from the primary tumour environment
through an active process, in which they acquire expres-
sion of genes involved in migration and invasion, and of
genes that switch their morphology to a mesenchymal-­
like motile phenotype. Epithelial-to‑mesenchymal tran-
sition (EMT) has been investigated in many settings and
evidence for its role in driving the release of CTCs is
growing19,20
. However, EMT is not essential for tumour
cell escape; an elevated intratumour interstitial pressure,
an incompetent or damaged tumour vasculature, and
migration of actively motile cells can collectively result in
the entry of single CTCs or clusters of cells into the termi-
nal lymphatics or bloodstream21–23
. Both in clinical sam-
ples and in preclinical tumour models, tumour cells have
been shown to be associated with macrophages during
migration towards blood vessels, where they interact with
endothelial cells before intravasation24,25
. The presence of
CTCs with both epithelial and mesenchymal properties
has been reported in blood samples from patients with
breast20,26
, prostate26
and lung27
­
cancer; cell clusters, com-
posed in part of tumour cells, have also been detected
in the circulation of these patients, and their presence
is associated with an earlier onset of metastatic disease
­
compared with single CTCs28–31
.
The ability of CTCs to lodge in capillaries and expand
to form macrometastases has been demonstrated in sev-
eral studies, including a report using tumour xenografts
generated from blood samples of patients with metastatic
prostate or colon cancer32
. In subsequent studies, CTCs
from patients with breast and small-cell lung ­
cancer
(SCLC) injected into immunologically ­
compromised
mice resulted in tumour formation33–35
.
Researchers have reported that breast and prostate
CTCs can be propagated in vitro for prolonged ­
periods
of time (>6 months in some cases) as spheroids or
­organoids32,36
— a hallmark of cancer stem cells (CSCs)
that are known to have a high capacity for self-renewal,
a radioresistance and chemoresistance phenotype, and a
high metastatic potential37,38
. The cell lines and tumours
derived from these CTCs share histological and immuno­
chemical features and a mutational landscape similar
to the primary tumour, indicating that their biologi-
cal properties remain after long-term culture. In one
study35
, multiple tumour cell lines could be established
from six out of 36 patients with breast cancer from whom
CTCs had been obtained during chemotherapy, but not
from CTCs harvested before treatment, indicating that
the CTCs isolated during treatment were more numer-
ous and/or more autonomous in their growth require-
ments than those obtained before treatment. Taken
together, these data demonstrate that, in addition to
being prognostic or predictive markers, CTCs can also
be ­metastasis-initiating cells.
While, theoretically, CTCs can access all tissues in the
body, secondary tumours tend to arise in selected tissues,
depending on the type of cancer. This observation is the
basis of the famous ‘seed and soil’ hypothesis proposed
over 125 years ago by Stephen Paget39
. Studies conducted
Key points
• Distant metastasis remains a frequent cause of death, even after locoregional
disease control is achieved using surgery, radiotherapy, and/or systemic therapy
• Antitumour therapies can, under some circumstances, mobilize tumour cells into the
peripheral circulation that might influence the risk of distant metastasis
• Irradiation can enable tumour cells to acquire properties that facilitate their
dissemination and the subsequent generation of metastases
• Severalmechanismsmightexplainametastasis-promotingeffectderivedfromsurgical
procedures,suchastissuedisruptionandleakageofbloodcontainingtumourcells
• A potential link between systemic therapies and metastasis has not been established,
but the results of isolated studies indicate that this question needs to be addressed
• The risk of distant failure from antitumour therapy can potentially be reduced if
treatment-related factors capable of promoting metastasis are identified, and
targeted therapeutically
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 33
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
over the past decade have indicated that CTCs ‘home’
to sites within tissues in which tumour-promoting stro-
mal cells — including cells of the myeloid, fibroblast and
endothelial lineages — have already been recruited
and provide a supportive niche for the tumour cell40,41
.
The proposed mechanism by which the primary tumour
initiates the formation of the premetastatic niche is by
secreting factors, either as freely circulating molecules or
encapsulated within exosomes, that direct stromal cells to
tissue sites (or modulate the phenotypes of cells resident
at the site) that later attract CTCs42,43
. Whether the for-
mation of a niche is essential for metastases to develop,
and whether the early arrival of CTCs might stimulate the
initiation of the niche remains to be determined.
Radiotherapy
Preclinical studies
In the 1940s, the suspicion that an increased rate of
metastasis might follow radiotherapy led to testing of this
hypothesis in animal tumour models. The initial investi­
gations, summarized by von Essen44
, involved a range
of tumour-bearing animals irradiated at vari­
ous doses
in single or multiple fractions. In general, these ­
studies
indicated that irradiation of primary tumours with lower
doses (insufficient for local ­
control) was associated
with the highest risk of metastases. Kaplan and Murphy45
observed an unexpectedly high number of early distant
metastases in patients with epidermoid carcinoma of the
lower lip and buccal mucosa treated with radiotherapy.
In 1949, they conducted a series of animal experiments
using subcutaneously transplanted murine mammary
carcinomas that were locally irradiated with 4–10Gy
(non-curative) in single X‑ray doses. Within 8 weeks,
pulmonary metastases were found in 43.5% of irradiated
mice and 9.6% of controls. Those metastases were exclu-
sively pulmonary and grew intravascularly in arteries
and arterioles. The irradiated tumours recurred locally
within 2 weeks. In 1976, Sheldon and Fowler46
irradiated
mammary carcinomas implanted on the anterior chest
wall of syngeneic mice using either a single dose of 5Gy
or two fractions of 3.5Gy; excision of the tumours was
performed while the mice were under anaesthesia for the
second (real or sham) irradiation. Pulmonary metastasis
occurred 10 weeks later in 20% of non-­
irradiated mice,
25% of mice that received 5Gy and 30% of those that
received two doses of 3.5Gy, indicating that the metas-
tasis rate was enhanced by ­
two-fraction irradiation
­
compared with surgical removal alone.
In the past 15 years47–51
, investigators have sought to
explain this phenomenon. For example, Camphausen
and colleagues47
injected Lewis lung carcinoma cells
into the hindlimbs of mice and irradiated the resulting
tumours. Local disease control was achieved in 71%
of the irradiated mice, but these mice had a higher
number of lung metastases when compared with non-­
irradiated mice. In further experiments involving mice
that had been injected with fibrosarcoma-derived cells,
irradiation markedly increased the numbers of pul-
monary metastases compared with those detected in
non-­
irradiated tumour-bearing mice. The researchers
suggested that previously dormant metastases had been
activated by radiation47
, in agreement with von Essen
and colleagues44
, who showed that both tumour
cells and stroma are modified by radiation, promoting
the selection of ­
more-aggressive and invasive cells with
­metastatic potential.
Studies of tumour cell biology indicate that radio­
therapy can alter tumour cells, making them more
aggressive than non-irradiated cells. The two phases of
a typical course of conventionally fractionated high-dose
radiotherapy (~2Gy per fraction) can have different bio-
logical effects (FIG. 1). In the later phase of radiotherapy
(for example, after 30 daily fractions of 2Gy), the high
cumulative radiation doses will have caused permanent
loss of reproductive capacity in all clonogenic tumour
cells, if the treatment is successful. Tumour cell death
occurs primarily through the induction of irreparable
DNA damage and cell cycle arrest52
. Irradiated cells usu-
ally undergo one or two error-prone cell cycles before
succumbing to mitotic catastrophe, apoptosis, necrosis, or
autophagy53–55
, depending on the dose. In the early stages
of fractionated radiotherapy (that is, after delivery of only
sublethal cumulative radiation doses, such as 2–6 Gy in
1–3 fractions), a significant proportion of the irradiated
tumour cells can repair the DNA damage; tumour cells
are much more likely to survive if they escape into the
circulation at this stage than the more-heavily irradiated
cells that enter the circulation later in treatment.
Cellular radioresistance and radiosensitivity depend
on many factors, including cell type and origin, cell-­cycle
phase, tissue oxygenation, and genetic background. CSCs
are more radioresistant than non-stem cells56,57
and can
selectively survive irradiation. If they are not elimi­
nated
or do not undergo permanent cell-cycle arrest (senes-
cence or dormancy), CSCs have the capacity to cause
disease recurrence. Furthermore, non-small-cell lung
carcinoma (NSCLC)-derived cells have been reported to
survive 5Gy irradiation by acquiring new CSC charac­
teristics, including the capacity to form 3D spheroids
in vitro, to self-renew and to generate differentiated
progeny. These cells also acquire the ­more-mesenchymal
phenotype typical of EMT38
.
Failure to repair DNA double-strand breaks caused
by radiotherapy is a major threat to genomic integrity58,59
.
Irradiated tumour cells have increased genome instability
and plasticity compared with unirradiated cells, and thus
can become more radioresistant. Rofstad et al.48
reported
that irradiation of mouse melanoma xenografts induced
tumour hypoxia that, in turn, promoted metastasis by
upregulating the expression of urokinase plasminogen
activator surface receptor (uPAR). Injections of anti-
uPAR antibodies almost completely blocked metastasis.
Changes in invasiveness and biomechanical properties
might also occur in irradiated cells. For example, the
expression levels of the proto-oncogene MET (encoding
HGFR, a known driver cell invasiveness), were fivefold
higher in tumour cells irradiated at 10Gy than in non-­
irradiated tumour cells60
; silencing of MET expression
prevented radiation-induced proliferation and invasive-
ness,andpromotedapoptosis.Zheng et al.61
,reportedthat
low-doseirradiation(1–4Gy)ofcellsderivedfromtongue
squamous-cellcarcinomaincreasedtheirinvasivenessina
REVIEWS
34 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
dose-dependent manner and changed some of their bio-
mechanical properties, including the ­organization of their
actin cytoskeleton.
In 2015, a study revealed that the migratory capacity of
tumour cells induced by low doses of radiation was medi-
ated by G‑CSF secreted by the tumour cells62
. Human
NSCLC cells expressing G‑CSF receptors were stimulated
with radiation to release G‑CSF, which subsequently pro-
moted EMT through an autocrine mechanism involving
JAK/STAT3 signalling62
. Radiotherapy can also stimu-
late primary tumour self-seeding from CTCs released
either before or after radiation exposure. Vilalta and
co-­workers63
used preclinical models to demonstrate the
release of GM‑CSF from irradiated tumour cells stimu­
lated the migration of tumour cells into the irradiated site,
thus triggering local tumour recurrence.
Abundant evidence indicates that radiation can indi-
rectly affect metastasis by modulating angio­
genesis,
thereby affecting the biological behaviour of tumour
cells. For example, radiation stimulates hypoxia-­inducible
factor 1 (HIF‑1) and, subsequently, the expression
of VEGF64,65
. In a clinical trial in which patients with
hepato­
cellular carcinoma received either chemotherapy
alone or chemotherapy plus conformal radiotherapy, an
associ­ationwasobservedbetweencombinedchemoradio­
therapy and rapid tumour progression outside the irradi­
ated field66
. In cell-culture experiments performed in
hypoxia-mimicking conditions, Sofia Vala et al.67
demon-
strated that low doses of radiation (<0.8 Gy) led to the
rapid phosphorylation of VEGFR‑2 and induced VEGF
production — thereby, enhancing endothelial-cell migra-
tion. Moreover, Shen et al.68
reported that irradiation of
cultured cancer cells induces secretion of protein-lysine
6‑oxidase (LOX), a key player in hypoxia-dependent
tumour-celldisseminationandmetastasis.IncreasedLOX
secretion was also detected in mouse serum after tumour
xenografts were exposed to localized radiation63
.
Clinical studies of radiotherapy
In some clinical situations, locoregional radiotherapy
can reduce the risk of distant metastasis. For example, in
patients with stage I–III breast cancer, adjuvant regional
radiotherapy to the draining lymph nodes is associated
with a substantial improvement in distant metastasis-­
free survival (hazard ratio (HR) 0.82), leading to a sig-
nificant improvement of overall survival (HR 0.85, 95%
CI 0.75–0.96; P=0.011) at 10 years69
. Nevertheless, our
group reported the first clinical evidence that localized
radiotherapy, when delivered to an intact tumour, can
mobilize viable tumour cells into the circulation31,70
,
a phenomenon that could potentially increase the risk
of distant metastasis. We detected increased numbers of
CTCs, both as single entities or in clusters, in the blood
of patients with NSCLC early in the course of their radio­
therapy. Intense staining for phosphorylated histone
H2AX (γ-H2AX), a biomarker for DNA double-strand
breaks71,72
, showed that mobilized CTCs were derived
from the irradiated tumour, with the highest number of
CTCs mobilized after the first and second daily fractions
of radiotherapy — a sublethal dose for many tumour
cells (FIG. 1). Mobilized CTCs had an increased capacity
to grow in culture both as attached cultures and orga-
noids, compared with CTCs collected before treatment,
(O. A. Martin, unpublished work); such attributes can be
associated with unfavourable patient outcomes34
.
In lung cancer, strong clinical evidence indicates
a link between the fractionation schedule employed
in radiotherapy and the subsequent risk of distant
Nature Reviews | Clinical Oncology
Radiation-induced
DNA damage
Cancer
non-stem
cells
Cancer
stem cells
DNA damage repaired
DNA damage unrepaired Radiation-induced
cell death
DNA damage misrepaired
• EMT
• Hypoxia
• Motility
• Invasiveness
Initial 2 Gy
radiotherapy
fractions
Accumulated radiotherapy fractions
Increasing number of treatment fractions over time
Early radiotherapy
survival (<50%)
Figure 1 | Progressive effects of fractionated radiotherapy on tumour cells in vivo. Up to one-half of the malignant
cells in an irradiated tumour can survive the first radiotherapy fractions; they can subsequently acquire a more-aggressive
phenotype, becoming circulating tumour cells that are detectable during the course of radiotherapy. Radiotherapy affects
the regulation of genes associated with radioresistance, tumour aggressiveness, and enhanced metastatic potential,
including signatures associated with hypoxia, invasiveness and motility, and epithelial-to‑mesenchymal transition (EMT).
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 35
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
metastasis. Continuous hyperfractionated accelerated
radiotherapy (CHART)73
, a very intense but brief treat-
ment regimen, improved survival and local disease con-
trol compared with conventional prolonged radiotherapy
in patients with locally advanced NSCLC. CHART was
also associ­
ated with a 24% reduction in the relative risk
of death, equivalent to an absolute improvement in 2‑year
survival from 20% to 29% (P=0.004) between conven-
tional radio­therapy and CHART. With mature follow‑­­up
(at least 3 years), in patients with squamous carcinoma
a 24% reduction in the relative risk of distant metasta-
sis was noted (P=0.043)74
. The reduction in the distant
metastatic rate in patients treated with CHART has sev-
eral explanations, including a reduced probability of late
metastasis from uncontrolled, compared with controlled,
local disease. CHART was delivered in three fractions per
day; CTCs mobilized by this radiotherapy regimen pre-
sumably acquired more damage at any given time than
cells exposed to daily radiotherapy — and might have a
reduced capacity to form metastases.
To our knowledge, no other research group has sys-
tematically explored changes in CTCs during the early
phase of radiotherapy. Using a telomerase-based assay,
Dorsey et al.75
detected viable CTCs in serial blood
samples of patients with NCSLC receiving radiotherapy
(telo­
merase contributes to cancer cell immortality and is
present in almost all cancer cells, but at very low levels
in normal cells). Lowes et al.76
investigated CTCs using
the CellSearch system (Janssen Diagnostics, Raritan, New
Jersey, USA) in patients with prostate cancer before and
3 months after completion, but not during radiotherapy;
patients who had increased or unchanged CTC numbers
after radiotherapy experienced treatment failure. These
preliminary data76
are derived from a combined popu­
lation of only 19 patients, but are consistent with the
hypothesis that failure to clear CTCs might be associated
with an unfavourable prognosis, an important subject for
future research.
Effects of radiotherapy on immunity
Immune responses can be finely balanced between pro-
moting metastasis and inhibiting signalling pathways
affected in cancer; antitumour therapy can perturb this
balance, with a positive or negative effect on the risk
of metastasis77
. Systemic immunosuppression owing
to antitumour therapy, including radiotherapy, could
potentially increase the risk of metastasis. Individuals
who have received renal and cardiac transplants have an
increased incidence of cancer compared with the gen-
eral population, owing to immunosuppressive thera­
pies associated with transplantation. In a study with
381 patients who received a cardiac transplant, 130
(34%) had developed a malignancy after a median fol-
low up of 9.7 years78
. Immunosuppressed patients have
a worse prognosis than immunocompetent patients
with cancer79
. Radiotherapy induces a pronounced
local inflammatory response77
, enhancing expression of
inflammation-related cytokines, including TGFβ, which
mediates EMT in mammalian cells80
. Several studies
have examined radiation-induced EMT. For example,
Park et al.49
established a mouse xenograft model using
a cancer cell line expressing luciferase. After irradiation
of primary tumours, the authors observed luminescent
signals identified as intestinal and pulmonary metas-
tases; these lesions expressed EMT markers and were
histologically confirmed as metastatic tumours. The
expression of several proteins50,81–83
has been associated
with the acquisition of EMT characteristics by irradiated
tumour cells, and could be potentially targeted to atten-
uate the harmful effects of radiation and/or to enhance
the efficacy of radiotherapy.
Traditionally, ionizing radiation has been considered
immunosuppressive because of the inherent susceptibil-
ity of naive immune cells to radiation84
and thus, radio­
therapy was not deemed to contribute to anti­
tumour
immunity. A growing body of evidence, however, sup-
ports the possibility of irradiated tumour cells becom-
ing a robust source of neoantigens, which can prompt
local and systemic antitumour immune responses, in
addition to direct radiation-induced lethality77
. Immune
responses can depend greatly on the type of radiation-­
induced cell death (that is, mitotic catastrophe, apoptosis,
necrosis, or autophagy). DNA released from irradiated
tumour cells activates both innate and adoptive immune
cells via different mechanisms85–87
, including boosting
cytotoxic T lymphocytes that contribute to tumour-cell
eradication77
. The fate of a cell damaged by radiation
depends mainly on the extent and type of DNA damage
and, therefore, on the absorbed dose of radiation and the
regimen used88
. The DNA-damage response and signal-
ling pathways involved in the immune response have
been proposed to cooperate to cause cancer cell death89
.
While lower radiotherapy doses (<2 Gy) can promote
anti-inflammatory responses77,90
, higher doses (>15 Gy)
might induce an intense inflammatory response that can
influence the survival of residual tumour cells inside and
outside of the irradiated area91
. Thus, a carefully selected
combination of radiotherapy fractionation schedule and
immune-modulating agents could enhance systemic
antitumour immune responses.
The most intense form of large-fraction radio­therapy
is stereotactic ablative body radiotherapy (SABR),
which is leading to a revolution in the management
of oligometa­
static solid tumours and small primary
tumours unsuitable for surgical resection, especially in
lung cancer92,93
. SABR can provide non-inferior survival
outcomes and local disease control comparable to sur-
gery94,95
owing to improvements in linear-accelerator
technology, cancer imaging, and radiotherapy-­
planning
software. The delivery of very large radiotherapy frac-
tions (>20 Gy), leads to high levels of cell death in both
tumour and stromal cells — thereby justifying the title
‘ablative’. The profound local inflammatory response
resulting from rapid cell death by SABR likely contributes
to both local control and induction of a systemic immune
response. This response can sometimes be capable of
causing disease regression at sites distant from the irradi­
ated volume, the so‑called ‘abscopal’ effect of radiother-
apy96
. The potential effect of SABR on CTC mobilization
has not been studied; future comparisons of the rates of
distant metastasis in matched patients treated with either
surgery or SABR will be of particular interest.
REVIEWS
36 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
Radiotherapy–metastasis interactions
Direct evidence for mobilization of CTCs during radio­
therapy in humans is at present limited to reports from
our own group in patients with NSCLC31
. This mobil­
ization might be due to the effect of radiation on tumour
architecture,especiallyonthetumourvasculature.During
fractionated radiotherapy, actively proliferating tumour
and stromal cells (both radiosensitive) increasingly
undergo cell death with accompanying disruption of
tumour architecture, which could promote direct entry
of tumour fragments into the circulation. Furthermore,
tumour debris, including irradiated stromal and viable
neoplastic cells, might be transported to the terminal lym-
phaticsandsubsequentlyformcoloniesindraininglymph
nodes, owing to mutations or other adaptations enabling
them to survive in this harsh environment. Viable tumour
cells might also be capable of forming distant metastases
upon entry into the ­
peripheral circulation (FIG. 2).
Patients with detectable lymph-node metastasis at
the time of locoregional treatments are much more likely
to experience distant failure than patients with node-­
negative cancer. For example, in a multivariate analysis
of known prognostic factors, only lymph-node involve-
ment was associated with distant relapse in patients with
locally advanced cervical cancer treated with definitive
radiotherapy97
. Similarly, the distant site relapse rate in
patients with endometrial cancer following curative treat-
ment was >66% when both lymphovascular invasion and
lymph-node metastasis were present, but only 7% when
these parameters were negative98
.
Radiotherapy might also negatively affect CTC biol-
ogy. In NSCLC, radiotherapy increased the number of
CTCs detected in clusters31
, which are associated with
an increased rate of metastasis in several cancers30,99,100
.
Radiotherapy was also associated with an increase in
the number of CTCs displaying mesenchymal-like
­characteristics31
.
No direct clinical evidence exists to link an increase in
radiotherapy-released CTCs or mesenchymal-like CTC
characteristics associated with a worse prognosis. If the
Nature Reviews | Clinical Oncology
Primary
lymph node
Secondary
lymph node
Direct tumour
cell entry into
draining veins
Venus circulation
via LV anastomoses
Venus circulation
via LV anastomoses
Venus circulation
via thoracic duct
Disrupted tumour
blood vessel
• Disrupted tumour architecture
• Elevated tumour interstitial pressure
• Opening of lymphatics
• Entry of tumour components into lymphvasculature
a Venous
b Lymphatic
Figure 2 | Routes of cancer cell mobilisation during treatment of an intact tumour. Tumour irradiation with doses
insufficient to eliminate all tumour cells can result in the disruption of the tumour architecture and, subsequently, lead to
the entry of tumour cells and other tumour components into the peripheral circulation, either directly into the venous
system (panel a) or indirectly via the lymphatic system (panel b) through lymphaticovenous (LV) anastomoses, or the
thoracic duct, following hydrostatic pressure gradients. Other therapeutic interventions can have a similar effect on
tumour cell mobilization.
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 37
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
release of CTCs during radiotherapy does have prog-
nostic value, then strategies designed to target CTCs or
to ensure that they are critically damaged before entry
into the circulation would become appealing. Such strat­
egies could include delivery of radiotherapy with larger
or more-frequent fractions, appropriately timed co‑­
administration of systemic agents to eliminate mobilized
CTCs,immunemodulationtoeliminateCTCs,andthera­
pies targeted at the prevention of EMT or ­
elimination of
­
mesenchymal-like tumour cells.
Surgical procedures
Patients with cancer can undergo a wide variety of pre-
surgical and surgical procedures — ranging from external
manipulation (such as endorectal ultrasound) to radical
resection with extensive lymph-node dissection — all of
which have the potential to perturb the tumour and lead
to the release of CTCs. After resection, the surgical bed
can contain, in addition to viable tumour cells, a mixture
of blood, extracellular fluid and a wide range of cytokines
and inflammatory cells, which can promote entry of via-
ble tumour cells into the local and regional lymphatic
vessels and peripheral circulation101
. Tumour cells can be
inadvert­
ently implanted into surgical wounds and drain-
age tube tracks, leading to loco­
regional recurrence102
.
Several groups have reported that increased numbers of
CTCscanbedetectedduringandaftersurgery(compared
with the levels before surgery), leading to the hypoth­
esis that surgery itself could promote distant metastasis.
Invasive surgical procedures are generally performed
under some form of regional or general anaesthesia; the
physiological effects of anaesthetic and surgical pro­
cedurescouldpotentiallyinfluencetheriskofmetastasis103
.
Perioperative factors, such as paracrine and neuroendo-
crine responses to surgery, could facilitate the metastatic
process by directly affecting malignant tissues, or through
indirect pathways, such as immunological perturbations.
Surgery-related anxiety and stress, and the nutritional sta-
tus might also be important factors104
. Anaesthesia itself
might also impair various immune functions, including
those of neutrophils, macrophages, dendritic cells, T‑cells,
andnaturalkiller(NK)cells105
.Intravenouslyadministered
induction agents (such as ketamine and thiopentone)
and inhalational agents (such as halothane) can suppress
NK‑cell activity in animal studies, and the upregulation of
hypoxia-inducible factors by volatile anaesthetics can be
cytoprotective for residual tumour cells106
. The pathways
involved in these processes, as well as their clinical rele-
vance,haveyettobeelucidated.Finally,bloodtransfusions
given in the perioperative period have been shown to be
associated with higher disease recurrence (odds ratio 1.6)
in patients with head and neck cancer and hepatocellular
carcinoma,aphenomenonattributedbysomeresearchers
to immunosuppression107
.
Preclinical studies
Detailed reviews on the effect of surgery on tumour
growth and metastasis using preclinical models have
been published108,109
. Evidence supports both a favour-
able and an unfavourable role of surgery in cancer out-
comes. Studies that conclude a detrimental effect of
surgery on subsequent outcome in preclinical models
invoke Folkman’s reports of the release of antiangiogenic
proteins (such as angiostatin and endostatin) from the
primary tumour to prevent the expansion of metastatic
lesions110
. Removal of the primary tumour relieves this
brake on angiogenesis, thus accelerating metastasis.
In other ­
studies, stimulation of angiogenesis through
wound-healing mechanisms triggered by the surgical
procedure has been demonstrated111
. A study from 2014
that used a preclinical cancer model112
described that the
procedure of core-­
needle biopsies can induce distant
metastasis, mediated by inflammatory cytokines secreted
as a result of the wound112
.
Effect of surgery on patients’ CTCs
Biopsies and other diagnostic procedures. Direct evi-
dence has linked needle biopsies (for example, in patients
with prostate cancer3,113,114
), and incisional biopsies (for
example, in patients with oral cancers115,116
), with the
detection of increased numbers of CTCs in the circula-
tion.Zoubekandcolleagues117
reportedachildwithpelvic
Ewing sarcoma who had CTCs detected in the peripheral
blood collected during open tumour biopsy (identified
using RT‑PCR of specific hybrid transcripts), but not
before nor 6 days after surgery117
. Tumours can also form
along the track left by a biopsy needle, as described in a
case report118
. Jones and colleagues119
retrospectively ana-
lysed patients with colorectal cancer liver metastases who
had undergone resection with or without a preoperative
needle biopsy. These investigators reported evidence of
tumours along the needle track in 17% of patients who
had undergone biopsy. The survival rate was worse in
patients who had undergone preopera­
tive biopsies com-
pared with those patients who had not119
. A remarkably
congruent local dissemination rate of 16% after liver
biopsy was reported in a similar trial120
. In response to
the increased awareness of tumour seeding along needle
tracks, ‘anti-seeding technology’ using radiofrequency
pulses has been proposed as a method to sterilize the
needle and its track121,122
. In prospective studies, Koch
and colleagues reported that the proportion of patients
having detectable CTCs increased after colonoscopy123
and endorectal ultrasound124
.
Cancer surgery. The number of publications relating
CTCs, surgery, and metastasis risk has increased con-
siderably over the years; herein, we discuss a selection
of those studies. The effects of surgery on CTCs have
been investigated extensively in patients with gastro-
intestinal tumours, especially in those with colorectal
cancer (CRC)125
. CRC cells often disseminate through
draining nodes and vessels; the most frequent distant
meta­
static site in patients with CRC is the liver, which
can be accessible by laparotomy. The presence of both
CTCs and disseminated tumour cells in lymph nodes
and bone marrow has negative prognostic significance
in CRC126,127
. Increased numbers of CTCs have been
reported during and after surgery compared with base-
line levels128
. At the time of surgery for CRC, CTCs
can reach the liver through the portal venous circula-
tion; CTCs are detected more frequently and in higher
REVIEWS
38 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
numbers in the portal venous compartment than in the
central venous compartment during surgery for CRC129
.
Trapping of CTCs in the liver, however, does not neces­
sarily lead to liver metastases. Koch and colleagues130
identified CRC-derived cells in liver biopsies of 10 of 100
patients with UICC stage I–III CRC, but no correlation
between the presence of these cells and the survival rate
of the patients was found, suggesting that most tumour
cells trapped in the liver did not survive to cause metas-
tasis. In a study of 37 patients who underwent resection
of liver metastases131
, however, multi­
variate analysis
confirmed that tumour-cell detection in intraoperative
blood and in bone marrow were independent prognos-
tic factors for tumour relapse. Interestingly, Wind et al.131
reported a difference in CTC mobilization between open
and laparo­
scopic surgical resections, with fewer CTCs
detected after laparoscopy.
If CTCs mobilized by surgery in CRC are, indeed, a
cause of metastases, then preoperative treatment could
potentially modulate the metastatic risk. The number of
CTCs detectable during and after surgery can be reduced
by the use of preoperative chemoradiotherapy in rectal
cancer132
. Kienle and colleagues132
assessed the blood and
bone marrow samples from 142 patients collected before,
during, and after surgery. Tumour cells were detected
in 34 of 103 (33%) bone marrow and 65 of 117 (55.6%)
blood samples of patients who did not receive treatment,
compared with 4 of 24 (16.7%) bone marrow and 10 of
25 (40%) blood samples of patients receiving treatment.
The tumour-cell detection rate was lower in the chemo-
radiotherapy group compared with untreated patients,
but the survival rate was significantly worse in patients
with tumour cells in the bone marrow after neoadjuvant
therapy compared with those without detectable bone
marrow involvement132
. Preoperative radiotherapy and
chemoradiotherapy both improved local disease control,
but the roles of these treatments in metastasis prevention
or overall-survival improvements is unclear133
. In a study
of 162 patients with rectal cancer treated with preopera-
tive radiotherapy, Nesteruk and colleagues134
found that
thedetectionofCTCs7daysaftersurgerywasofprognos-
tic value for local recurrence, whereas detection of CTCs
before or 24 hours after surgery was not.
In breast cancer, a bimodal pattern of distant metas-
tasis after treatment has been reported in surgical ­studies
involving large cohorts (ranging between 1,526–3,921
patients)135–137
, a phenomenon that could be related
to events occurring at the time of surgery. Demicheli
and colleagues138
analysed the results of trials in which
patients with axillary node-positive breast cancer had
received either adjuvant chemotherapy after breast
­
cancer surgery, or mastectomy and nodal dissection
alone. These investigators proposed that the meta-
static process could be driven by mastectomy, and that
micrometastases could undergo sustained periods
of dormancy.
In NSCLC, both open surgery and video-assisted
surgery139,140
have been associated with increased CTC
counts in venous blood. Hashimoto and colleagues141
found that, on average, surgery caused increases of CTC
numbers in the pulmonary veins in 30 patients treated
with lobectomy for peripheral NSCLC. Dong and col-
leagues142
studiedacohortof31patientswithNSCLCwho
had blood collected from a pulmonary vein during open
thoracic surgery; of these 31 patients, 15 had positive test
results for CTCs. The median survival and 2‑year survival
rates for patients with positive versus negative findings
were11monthsand26.7%, ­respectively,versus27months
and 62.5%, respectively.
In organ-confined prostate cancer, Eschwege et al.143
reported that CTC numbers were increased in 12 of 14
patients who underwent surgery compared with before
surgery. Other situations in which CTC counts might
become elevated during or soon after surgery include
breast cancer resection144
, transurethral bladder cancer
resection145
and oesopha­
geal cancer resection146
. The
surgical community has become increasingly aware of
the potential association between surgical intervention,
CTCs and metastasis risk, and is actively seeking ways to
­
investigate and reduce CTC mobilization.
Surgery–metastasis interaction
The mechanisms by which therapeutic interventions
can promote CTC mobilization are unknown and are
likely to vary between and within modalities. The worst
outcomes observed in patients with mobilized CTCs
might reflect the negative impact of underlying vascular
invasion rather than a sudden large influx of CTCs. For
example, mobilization of CTCs after rectal ultrasound,
which involves external pressure on the tumour, could
reflect an early release of CTCs already primed within
blood vessels. Direct extension of macroscopic tumours
into large veins is an extreme example of such vascular
invasion (FIG. 3). In renal carcinomas, life-­
threatening
tumour embolisation to the lung can occur, before
or during surgical resection147
. In patients receiving
curative-­
intent surgery, the severing of blood and lym-
phatic vessels in proximity to the tumour can lead to the
formation of a fluid-filled cavity containing tumour cells
that might subsequently gain access to draining lym-
phatic channels148
. In addition, surgery induces a local
inflammatory response in the wound, characterised by
the influx of immune cells and a release of cytokines that
can ­
influence the risk of metastasis149
.
Nature Reviews | Clinical Oncology
a b
SVC
SVC
T
T
A
A
Figure 3 | Tumour extension into the venous system. a | Coronal CT and
b | 18
F-FDG-PET/CT images of a patient with direct extension of a lung cancer (T) into
the superior vena cava (SVC). The tumour causes a filling defect within the SVC and,
unlike a simple clot (which would be metabolically inactive), is FDG-avid.
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 39
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
A causal link between surgery and metastasis has not
beenestablishedtodate.Nevertheless,proposedstrategies
designed to minimise the entry of CTCs into the vascu-
lar system at the time of surgery, include clamping the
blood vessels that drain the tumour at the earliest possible
time point, and isolating the tumour from the circulation
before resection150
.
Systemic antitumour therapies
Evidence from studies of both surgery and radiotherapy
indicates that disturbance of the tumour or its environ-
ment by therapeutic intervention might, under some
circumstances, promote metastasis. Our group has, there-
fore, investigated the possibility that systemic anticancer
therapies might also occasionally have this effect, despite
the counterintuitive nature of such a hypothesis. A very
limited number of publications have addressed this topic.
Unlike surgery and radiotherapy, systemic treatments for
cancer — including chemotherapy and targeted thera-
pies — can have a direct effect not only on cancer cells in
the primary tumour, but also on CTCs and micrometas-
tases present at the time of treatment. In a growing num-
ber of clinical studies, the CTC count has been used to
monitor response to systemic therapy, but the potential
prognostic and predictive value of CTC release early dur-
ing chemotherapy or targeted therapies has not been fully
explored. In many studies, the presence of CTCs before
chemotherapy is associated with a worse overall survival,
compared with that of patients in whom CTCs are not
detectable initially151
. Smerage and colleagues151
reported
that, in patients with increased CTC counts (≥5 CTCs per
7.5ml of whole blood) detected before the commence-
ment of chemotherapy, the number of CTCs often failed
to decrease after treatment. Paradoxically, the results of
somepreclinicalandclinicalstudiesonthemobilizationof
haematopoieticstemcellsbeforehigh-dosechemotherapy
and autologous stem-cell transplant suggest that chemo-
therapy has the potential to increase the number of CTCs.
These studies are ­
summarised in the following sections.
Preclinical studies
A series of studies in preclinical metastasis models have
indicated that disruption of angiogenesis or induction
of damage to endothelial cells can enhance metastasis.
Two important studies showed that inhibition of angio-
genesis was able to reduce primary tumour growth152,153
;
­
however, by inducing an hypoxic environment in the
residual tumour, the therapy promoted invasion and
metastasis. Treatment of RIP1‑Tag2 mice, which develop
pancreatic tumours spontaneously, with a neutralising
antibody targeting VEGFR‑2 led to a reduction of pri-
mary tumour volume152
, but also resulted in an increase in
local invasion and distant metastasis. Similar results were
obtained using sunitinib, a small-molecule inhibitor of
both VEGFR and PDGFR152
. Ebos et al.153
demonstrated
a similar phenom­enon in transplantable models of breast
cancer and melanoma using the angiogenesis inhibitors
sunitinib, sorafenib, and SU10944. Antiangiogenic thera-
pies for cancer have generally not proved to be as effective
in improving the survival of patients with cancer as was
originally hoped154
. One of the reasons for this limited
effectiveness could be that the beneficial effects of target-
ing the tumour vasculature are offset by unwanted effects
on tumour cell hypoxia, invasion, and metastasis.
Other targeted therapies can also promote metastasis
in preclinical models. Treatment of mice bearing pri-
mary tumours derived from injection of A-375 human
melanoma cells with the BRAF inhibitor vemurafenib,
resulted in the suppression of tumour growth. When a
small proportion of vemurafenib-resistant A-375 cells
were injected together with parental A-375 cells, however,
treatment with vemurafenib resulted in the vemurafenib-­
sensitive cells secreting factors that promoted the growth
and metastasis of vemurafenib-­
resistant tumours155
. The
secreted factors activated AKT signalling in resistant cells,
thereby driving their ­
continued growth and metastasis.
Individual cytotoxic chemotherapy agents (such as
cyclophosphamide) have also been shown to promote
metastasis under certain conditions. High-dose cyclo-
phosphamide is effective in inducing treatment responses
in some cancers, although this agent can also cause
immunosuppression and, thus, is used to manage graft-
versus-hostdisease156
.Conversely,lowdosesofcyclophos-
phamide can enhance antitumour immune responses in
animal models157
. Man and colleagues158
showed that,
in addition to causing primary tumour shrinkage, both
low-dose and high-dose cyclophosphamide promoted
metastasis of subcutaneously implanted lung adeno­
carci­nomas to the lung. Other studies have demonstrated
that an injection of cyclophosphamide 24h before intra­
venous inoculation of fibrosarcoma-derived cells mark-
edly enhanced tumour-cell colonization of the lung159
.
Another example is the heat shock protein 90 inhibitor
17‑AAG, which inhibited the growth of primary breast
tumours, but also promoted metastasis to the bone by
enhancing osteoclast formation160
.
Clinical studies with chemotherapy
In patients with detectable CTCs, a reduction of CTC
numberswithchemotherapyisassociatedwithfavourable
responses to treatment and improved survival compared
withpatientswithoutadeclineinCTCs142,152,153
.Thechange
in CTC numbers might even constitute a surro­
gate end
pointforchemotherapyresponsiveness151,161,162
.Inpatients
which CTC numbers increase steadily after chemother-
apy, the increase is probably caused by the release of CTCs
from progressive disease. Direct evidence for mobiliza-
tion of CTCs by chemotherapy in humans is scarce, but
provocative findings have been reported for high-dose
chemotherapy and autologous stem-cell transplantation
for the treatment of advanced-stage or high-risk solid
tumours. For instance, Brugger and colleagues163
­studied
the outcomes of 46 patients with NSCLC, SCLC, or breast
cancer who underwent peripheral blood stem-cell har-
vesting and CTC analysis after treatment with the cyto-
toxic chemotherapy drugs VP‑16, ifosfamide, cisplatin
and G‑CSF (administered to mobilize blood progeni­
tor
cells for subsequent harvesting). Of 42 patients with no
detectable CTCs at baseline, 13 patients had detectable
CTCs after chemotherapy. In the remaining four patients
(with detectable baseline CTCs), CTC numbers increased
after chemotherapy164
.
REVIEWS
40 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
Mobilization of CTCs by chemotherapy might be
irrelevant in patients who already have metastatic disease;
however, when chemotherapy is given as neoadjuvant
therapy (before planned curative surgery or radiother-
apy), with the intention of treating both the local disease
andoccultmetastasis,aparadoxicalmetastasis-promoting
effect of CTC mobilization could be clinically important.
Inhestern and colleagues164
studied CTCs in patients with
advanced-stage oral and oropharyngeal squamous-cell
carcinoma treated with neoadjuvant chemotherapy,
surgery, and postoperative radiotherapy. These investi­
gators found that CTCs were detectable in about 80%
of patients at baseline and that CTC numbers declined
during chemotherapy in most patients. After surgery and
before the planned adjuvant radiotherapy, however, CTC
numbers had increased to above baselinelevels.Thecause
of this phenomenon is unknown but in these patients,
who had potentially curable disease, chemotherapy was
unable to eradicate CTCs — CTC mobilization should be
considered as a possible cause for this observation.
Clinical studies: biological therapies
In the study by Brugger et al.163
, the relative contributions
of chemotherapy and G‑CSF to CTC mobilization could
not be separated. In another study165
, cells identified as
epithelial because they expressed epithelial cell-adhesion
molecule(EpCAM),werefoundtocontaminateharvested
haematopoietic progenitor cells mobilized by G‑CSF
alone in patients with metastatic breast cancer before
high-dose chemotherapy and autologous stem-cell trans-
plantation. Thus, G‑CSF alone might also mobilize CTCs
from the bone marrow or other metastatic sites. High-
dose chemotherapy and stem-cell transplantation are
no longer part of the standard therapy used for patients
with epithelial cancers because of the negative results
of randomised clinical trials166
; thus, the contribution of
mobilized tumour cells to disease relapse after ­autologous
stem-cell ­
transplantation has not been established.
G‑CSF-based therapy is commonly administered
to patients to treat or prevent neutropenia caused by
myelosuppressive chemotherapy. A meta-analysis of 17
randomized controlled trials of patients receiving chemo-
therapy concluded that the mortality rate associated with
infection (P=0.018) and febrile neutropenia (P<0.001)
was significantly reduced by the administration of G‑CSF;
however, insufficient data were available to reach any
conclusions about the effect of G‑CSF on overall sur-
vival167
. In preclinical studies, we and others have shown
that the administration of G‑CSF promotes metastasis of
mammary tumours168,169
and cell migration induced by
low-dose radiation, for example, is dependent on G‑CSF
signalling in NSCLC cells62
. As well as acting on tumour
cells that express the G‑CSF receptor, G‑CSF can mobilize
myeloid cells that have potent immunosuppressive activ-
ity against T cells, thereby enhancing metastasis168
. The
implications of these findings in the treatment of patients
with cancer are unclear.
Erythropoietin is another haematopoietic growth
factor that has been used to treat patients with cancer.
Erythropoietin receptors can be found on the surface
of different cancer cell lines, leading to concerns about
a potential role in tumour growth stimulation170
. In a
randomised trial comparing erythropoietin plus radio­
therapy with radiotherapy alone in patients with head and
neck cancer171
, the 5‑year estimate of locoregional failure
was 46.2% versus 39.4% (P=0.42); therefore, a detri­
mental effect of erythropoietin was not ruled out by the
study investigators.
The publication of reports on the use of molecularly
targeted agents in the treatment of patients with cancer
has rapidly grown in the past few years. The mechanisms
of action of such agents are completely different from
those of conventional cytotoxic agents. No good clinical
evidence exists on the potential role of these agents in
CTC dissemination, but this possibility should be con-
sidered when evaluating long-term results of treatments
with molecularly targeted agents.
Systemic therapy–metastasis interactions
The possibility of a metastasis-promoting effect from
systemic therapy in patients with cancer is a challen­
ging research question, especially because patients often
derive a benefit from such therapies. Nevertheless, seek-
ing evidence for such an effect is justifiable to improve
the overall benefits of antitumour therapies. A potential
cancer-­
promoting effect derived from systemic cytotoxic
anticancer therapies seems paradoxical because of the
therapeutic nature of such therapies; if this effect does
occur,theimmediateclinicalcoursewouldbedetermined
by the pre-existing macroscopic tumour burden.
CTC mobilization by chemotherapy could poten-
tially be important when neoadjuvant chemotherapy
is used to treat locoregionally confined advanced-stage
tumours before administering curative-intent surgery or
radiotherapy and/or chemoradiotherapy. In patients with
macroscopic stage IIIA NSCLC, chemotherapy followed
bysurgerysubstantiallyimprovedsurvivalcomparedwith
surgery alone172
. By contrast, only a small improvement in
survival has been observed when surgery is followed by
adjuvant chemotherapy173
. In breast cancer, however, the
benefits of neoadjuvant and adjuvant chemotherapy are
similar174
. The interactions between therapeutic modal-
ities are complex, and likely to vary substantially across
cancer types.
The superior outcomes reported across a range of epi-
thelial cancers for concurrent versus sequential chemo-
radiotherapy are usually attributed solely to an effect on
local disease control, but accurate information on a direct
link between local disease control and systemic disease
can be difficult to obtain from data generated in clinical
trials. Most frequently, only the site of first failure (local
or systemic) is reported, and subsequent local or distant
failures are not described. Nevertheless, distant and local
treatment failure have been recorded separately and
analysed according to FDG-PET response in a study
involving patients with locoregionally advanced-stage
NSCLC treated with concurrent chemoradiotherapy175
.
A powerful negative correlation between metastasis and
a favourable local treatment response emerged from this
study, indicating that chemoradiotherapy-responsive
tumour cells were less likely to metastasise during or
after treatment175
. Concurrent chemoradiotherapy could
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 41
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
reduce the risk of late distant relapse both by decreasing
the probability of spread from uncontrolled local disease,
and by ensuring that CTCs mobilized during treatment
have sustained more damage — and, therefore, would
be less capable of forming distant metastasis than CTCs
­
mobilized during radiotherapy as a single modality.
Conclusions
The relationships between frontline treatments, local
tumour control, and metastasis in patients with solid
tumours are complex and remain poorly understood.
Broad generalisations cannot be made from the studies
addressing these relationships because of the diversity
of cancers and antitumour therapies involved, as well as
variations in treatment delivery, CTC detection methods,
and time points analysed, together with the long time
period spanned by the publication of relevant observa-
tions. A common finding in these studies, however, is
that widely used antitumour therapies might be associ-
ated with an early increase in either the number or the
prolifera­
tive capacity of CTCs. In studies analysing both
changes in CTC numbers and survival, an increase in
CTCs was in general associated with a less-beneficial out-
come. None of these studies has established a direct link
between metastasis and CTC mobilization by treatment,
but they do indicate an association between increased
CTC numbers after the initiation of therapy and an
­
elevated risk of metastasis.
The results of these studies do not imply that the cur-
rent development of antitumour treatment methods,
often the hard-won results of painstaking sequential ran-
domized trials, should be abandoned because they might
promote CTC mobilization. These treatments enable
many patients with locoregionally confined advanced-
stage solid tumours, who would otherwise suffer and
die from progressive disease, to be cured or have long-
term disease-free survival. The best available treatment
cannot be administered without a diagnosis, but, if CTC
mobilization by invasive medical interventions or ther-
apy represents a major cause of antitumour treatment
failure, then this subject is worthy of intensive study. In
the meantime, using the least invasive means of effective
diagnosis and treatment in patients with cancer seems the
most prudent option.
1. Pantel, K. et al. Circulating epithelial cells in patients
with benign colon diseases. Clin. Chem. 58, 936–940
(2012).
2. Sanger, N. et al. Disseminated tumor cells in the bone
marrow of patients with ductal carcinoma in situ.
Int. J. Cancer 129, 2522–2526 (2011).
3. Husemann, Y. et al. Systemic spread is an early step
in breast cancer. Cancer Cell 13, 58–68 (2008).
4. Weigelt, B. et al. Gene expression profiles of primary
breast tumors maintained in distant metastases.
Proc. Natl Acad. Sci. USA 100, 15901–15905
(2003).
5. Allard, W. J. et al. Tumor cells circulate in the
peripheral blood of all major carcinomas but not
in healthy subjects or patients with nonmalignant
diseases. Clin. Cancer Res. 10, 6897–6904 (2004).
6. Lucci, A. et al. Circulating tumour cells in non-
metastatic breast cancer: a prospective study.
Lancet Oncol. 13, 688–695 (2012).
7. Fidler, I. J. The pathogenesis of cancer metastasis: the
‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 3,
453–458 (2003).
8. Luzzi, K. J. et al. Multistep nature of metastatic
inefficiency: dormancy of solitary cells after
successful extravasation and limited survival of early
micrometastases. Am. J. Pathol. 153, 865–873
(1998).
9. Coumans, F. A., Siesling, S. & Terstappen, L. W.
Detection of cancer before distant metastasis.
BMC Cancer 13, 283 (2013).
10. Klein, C. A. Parallel progression of primary tumours
and metastases. Nat. Rev. Cancer 9, 302–312 (2009).
11. Naxerova, K. & Jain, R. K. Using tumour phylogenetics
to identify the roots of metastasis in humans.
Nat. Rev. Clin. Oncol. 12, 258–272 (2015).
12. Sosa, M. S., Bragado, P. & Aguirre-Ghiso, J. A.
Mechanisms of disseminated cancer cell dormancy:
an awakening field. Nat. Rev. Cancer 14, 611–622
(2014).
13. Comen, E., Norton, L. & Massague, J. Clinical
implications of cancer self-seeding. Nat. Rev. Clin.
Oncol. 8, 369–377 (2011).
14. Gundem, G. et al. The evolutionary history of lethal
metastatic prostate cancer. Nature 520, 353–357
(2015).
15. Toss, A., Mu, Z., Fernandez, S. & Cristofanilli, M.
CTC enumeration and characterization: moving toward
personalized medicine. Ann. Transl Med. 2, 108
(2014).
16. Schwarzenbach, H. et al. Loss of heterozygosity at
tumor suppressor genes detectable on fractionated
circulating cell-free tumor DNA as indicator of breast
cancer progression. Clin. Cancer Res. 18, 5719–5730
(2012).
17. Spindler, K. L., Pallisgaard, N., Andersen, R. F. &
Jakobsen, A. Changes in mutational status during third-
line treatment for metastatic colorectal cancer—results
of consecutive measurement of cell free DNA, KRAS and
BRAF in the plasma. Int. J. Cancer 135, 2215–2222
(2014).
18. Murtaza, M. et al. Non-invasive analysis of acquired
resistance to cancer therapy by sequencing of plasma
DNA. Nature 497, 108–112 (2013).
19. Bonnomet, A. et al. Epithelial-to‑mesenchymal
transitions and circulating tumor cells. J. Mammary
Gland Biol. Neoplasia 15, 261–273 (2010).
20. Yu, M. et al. Circulating breast tumor cells exhibit
dynamic changes in epithelial and mesenchymal
composition. Science 339, 580–584 (2013).
21. Lafrenie, R., Shaughnessy, S. G. & Orr, F. W. Cancer cell
interactions with injured or activated endothelium.
Cancer Metastasis Rev. 11, 377–388 (1992).
22. Friedl, P., Locker, J., Sahai, E. & Segall, J. E. Classifying
collective cancer cell invasion. Nat. Cell Biol. 14,
777–783 (2012).
23. Rofstad, E. K., Galappathi, K. & Mathiesen, B. S.
Tumor interstitial fluid pressure‑a link between tumor
hypoxia, microvascular density, and lymph node
metastasis. Neoplasia 16, 586–594 (2014).
24. Robinson, B. D. et al. Tumor microenvironment of
metastasis in human breast carcinoma: a potential
prognostic marker linked to hematogenous
dissemination. Clin. Cancer Res. 15, 2433–2441
(2009).
25. Roussos, E. T. et al. Mena invasive (MenaINV) promotes
multicellular streaming motility and transendothelial
migration in a mouse model of breast cancer. J. Cell Sci.
124, 2120–2131 (2011).
26. Armstrong, A. J. et al. Circulating tumor cells from
patients with advanced prostate and breast cancer
display both epithelial and mesenchymal markers.
Mol. Cancer Res. 9, 997–1007 (2011).
27. Hou, J. M. et al. Circulating tumor cells as a window on
metastasis biology in lung cancer. Am. J. Pathol. 178,
989–996 (2011).
28. Brandt, B. et al. Isolation of prostate-derived single cells
and cell clusters from human peripheral blood. Cancer
Res. 56, 4556–4561 (1996).
29. Hou, J. M. et al. Clinical significance and molecular
characteristics of circulating tumor cells and circulating
tumor microemboli in patients with small-cell lung
cancer. J. Clin. Oncol. 30, 525–532 (2012).
30. Aceto, N. et al. Circulating tumor cell clusters are
oligoclonal precursors of breast cancer metastasis.
Cell 158, 1110–1122 (2014).
31. Martin, O. A. et al. Mobilization of viable tumor
cells into the circulation during radiation therapy.
Int. J. Radiat. Oncol. Biol. Phys. 88, 395–403 (2014).
32. Pretlow, T. G. et al. Prostate cancer and other xenografts
from cells in peripheral blood of patients. Cancer Res.
60, 4033–4036 (2000).
33. Baccelli, I. et al. Identification of a population of blood
circulating tumor cells from breast cancer patients that
initiates metastasis in a xenograft assay.
Nat. Biotechnol. 31, 539–544 (2013).
34. Hodgkinson, C. L. et al. Tumorigenicity and genetic
profiling of circulating tumor cells in small-cell lung
cancer. Nat. Med. 20, 897–903 (2014).
35. Yu, M. et al. Cancer therapy. Ex vivo culture of
circulating breast tumor cells for individualized testing
of drug susceptibility. Science 345, 216–220 (2014).
36. Gao, D. et al. Organoid cultures derived from patients
with advanced prostate cancer. Cell 159, 176–187
(2014).
37. Levina, V., Marrangoni, A. M., DeMarco, R., Gorelik, E.
& Lokshin, A. E. Drug-selected human lung cancer stem
cells: cytokine network, tumorigenic and metastatic
properties. PLoS ONE 3, e3077 (2008).
38. Gomez-Casal, R. et al. Non-small cell lung cancer cells
survived ionizing radiation treatment display cancer
stem cell and epithelial-mesenchymal transition
phenotypes. Mol. Cancer 12, 94 (2013).
39. Paget, S. The distribution of secondary growths in
cancer of the breast. Lancet 1, 571–573 (1889).
40. Kaplan, R. N. et al. VEGFR1-positive haematopoietic
bone marrow progenitors initiate the pre-metastatic
niche. Nature 438, 820–827 (2005).
41. Hiratsuka, S. et al. The S100A8‑serum amyloid
A3‑TLR4 paracrine cascade establishes a pre-metastatic
phase. Nat. Cell Biol. 10, 1349–1355 (2008).
42. Hood, J. L., San, R. S. & Wickline, S. A. Exosomes
released by melanoma cells prepare sentinel lymph
nodes for tumor metastasis. Cancer Res. 71,
3792–3801 (2011).
43. Fong, M. Y. et al. Breast-cancer-secreted miR‑122
reprograms glucose metabolism in premetastatic niche
to promote metastasis. Nat. Cell Biol. 17,
183–194 (2015).
44. von Essen, C. F. Radiation enhancement of metastasis:
a review. Clin. Exp. Metastasis 9, 77–104 (1991).
45. Kaplan, H. S. & Murphy, E. D. The effect of local
roentgen irradiation on the biological behavior of a
transplantable mouse carcinoma; increased frequency of
pulmonary metastasis. J. Natl Cancer Inst. 9, 407–413
(1949).
46. Sheldon, P. W. & Fowler, J. F. The effect of low-dose
pre-operative X‑irradiation of implanted mouse
mammary carcinomas on local recurrence and
metastasis. Br. J. Cancer 34, 401–407 (1976).
47. Camphausen, K. et al. Radiation therapy to a
primary tumor accelerates metastatic growth in mice.
Cancer Res. 61, 2207–2211 (2001).
REVIEWS
42 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
48. Rofstad, E. K., Mathiesen, B. & Galappathi, K.
Increased metastatic dissemination in human
melanoma xenografts after subcurative radiation
treatment: radiation-induced increase in fraction of
hypoxic cells and hypoxia-induced up‑regulation
of urokinase-type plasminogen activator receptor.
Cancer Res. 64, 13–18 (2004).
49. Park, J. K. et al. Establishment of animal model for the
analysis of cancer cell metastasis during radiotherapy.
Radiat. Oncol. 7, 153 (2012).
50. Li, T. et al. Radiation enhances long-term metastasis
potential of residual hepatocellular carcinoma in nude
mice through TMPRSS4-induced epithelial–
mesenchymal transition. Cancer Gene Ther. 18,
617–626 (2011).
51. Woods, G. M. et al. Radiation therapy may increase
metastatic potential in alveolar rhabdomyosarcoma.
Pediatr. Blood Cancer 62, 1550–1554 (2015).
52. Bernier, J., Hall, E. J. & Giaccia, A. Radiation oncology:
a century of achievements. Nat. Rev. Cancer 4,
737–747 (2004).
53. Eriksson, D. & Stigbrand, T. Radiation-induced cell
death mechanisms. Tumour Biol. 31, 363–372 (2010).
54. Dewey, W. C., Ling, C. C. & Meyn, R. E. Radiation-
induced apoptosis: relevance to radiotherapy.
Int. J. Radiat. Oncol. Biol. Phys. 33, 781–796 (1995).
55. Palumbo, S. & Comincini, S. Autophagy and ionizing
radiation in tumors: the “survive or not survive”
dilemma. J. Cell. Physiol. 228, 1–8 (2013).
56. Brunner, T. B., Kunz-Schughart, L. A., Grosse-
Gehling, P. & Baumann, M. Cancer stem cells as a
predictive factor in radiotherapy. Semin. Radiat.
Oncol. 22, 151–174 (2012).
57. Butof, R., Dubrovska, A. & Baumann, M. Clinical
perspectives of cancer stem cell research in radiation
oncology. Radiother. Oncol. 108, 388–396 (2013).
58. Jackson, S. P. Sensing and repairing DNA double-
strand breaks. Carcinogenesis 23, 687–696 (2002).
59. Roy, K., Kodama, S., Suzuki, K. & Watanabe, M.
Delayed cell death, giant cell formation and
chromosome instability induced by X‑irradiation in
human embryo cells. J. Radiat. Res. 40, 311–322
(1999).
60. De Bacco, F. et al. Induction of MET by ionizing
radiation and its role in radioresistance and invasive
growth of cancer. J. Natl Cancer Inst. 103, 645–661
(2011).
61. Zheng, Q. et al. X‑ray radiation promotes the
metastatic potential of tongue squamous cell
carcinoma cells via modulation of biomechanical
and cytoskeletal properties. Hum. Exp. Toxicol. 34,
894–903 (2015).
62. Cui, Y. H. et al. Radiation promotes invasiveness of
non-small-cell lung cancer cells through granulocyte-
colony-stimulating factor. Oncogene 34, 5372–5382
(2015).
63. Vilalta, M., Rafat, M., Giaccia, A. J. & Graves, E. E.
Recruitment of circulating breast cancer cells is
stimulated by radiotherapy. Cell Rep. 8, 402–409
(2014).
64. Gorski, D. H. et al. Blockage of the vascular
endothelial growth factor stress response increases
the antitumor effects of ionizing radiation. Cancer Res.
59, 3374–3378 (1999).
65. Moeller, B. J., Cao, Y., Li, C. Y. & Dewhirst, M. W.
Radiation activates HIF‑1 to regulate vascular
radiosensitivity in tumors: role of reoxygenation, free
radicals, and stress granules. Cancer Cell 5, 429–441
(2004).
66. Chung, Y. L. et al. Sublethal irradiation induces
vascular endothelial growth factor and promotes
growth of hepatoma cells: implications for
radiotherapy of hepatocellular carcinoma. Clin. Cancer
Res. 12, 2706–2715 (2006).
67. Sofia Vala, I. et al. Low doses of ionizing radiation
promote tumor growth and metastasis by enhancing
angiogenesis. PLoS ONE 5, e11222 (2010).
68. Shen, C. J. et al. Ionizing radiation induces tumor cell
lysyl oxidase secretion. BMC Cancer 14, 532 (2014).
69. Budach, W., Kammers, K., Boelke, E. & Matuschek, C.
Adjuvant radiotherapy of regional lymph nodes in
breast cancer - a meta-analysis of randomized trials.
Radiat. Oncol. 8, 267 (2013).
70. Errico, A. Radiotherapy: a double-edged sword for
NSCLC? Nat. Rev. Clin. Oncol. 11, 66 (2014).
71. Bonner, W. M. et al. γH2AX and cancer. Nat. Rev.
Cancer 8, 957–967 (2008).
72. Ivashkevich, A., Redon, C. E., Nakamura, A. J.,
Martin, R. F. & Martin, O. A. Use of the γ‑H2AX assay
to monitor DNA damage and repair in translational
cancer research. Cancer Lett. 327, 123–133 (2012).
73. Saunders, M. et al. Continuous hyperfractionated
accelerated radiotherapy (CHART) versus conventional
radiotherapy in non-small-cell lung cancer:
a randomised multicentre trial. CHART Steering
Committee. Lancet 350, 161–165 (1997).
74. Saunders, M. et al. Continuous, hyperfractionated,
accelerated radiotherapy (CHART) versus conventional
radiotherapy in non-small cell lung cancer: mature
data from the randomised multicentre trial. CHART
Steering committee. Radiother. Oncol. 52, 137–148
(1999).
75. Dorsey, J. F. et al. Tracking viable circulating tumor
cells (CTCs) in the peripheral blood of non-small cell
lung cancer (NSCLC) patients undergoing definitive
radiation therapy: pilot study results. Cancer 121,
139–149 (2015).
76. Lowes, L. E. et al. Circulating tumour cells in prostate
cancer patients receiving salvage radiotherapy.
Clin. Transl Oncol. 14, 150–156 (2012).
77. Haikerwal, S. J., Hagekyriakou, J., MacManus, M.,
Martin, O. A. & Haynes, N. M. Building immunity to
cancer with radiation therapy. Cancer Lett. 368,
198–208 (2015).
78. Rivinius, R. et al. Analysis of malignancies in patients
after heart transplantation with subsequent
immunosuppressive therapy. Drug Des. Devel. Ther. 9,
93–102 (2015).
79. Engels, E. A. et al. Spectrum of cancer risk among
US solid organ transplant recipients. JAMA 306,
1891–1901 (2011).
80. Andarawewa, K. L. et al. Ionizing radiation predisposes
nonmalignant human mammary epithelial cells to
undergo transforming growth factor β-induced
epithelial to mesenchymal transition. Cancer Res. 67,
8662–8670 (2007).
81. Yuan, W., Yuan, Y., Zhang, T. & Wu, S. Role of Bmi‑1 in
regulation of ionizing irradiation-induced epithelial-
mesenchymal transition and migration of breast
cancer cells. PLoS ONE 10, e0118799 (2015).
82. Kim, R. K. et al. Radiation promotes malignant
phenotypes through SRC in breast cancer cells.
Cancer Sci. 106, 78–85 (2015).
83. Qian, L. W. et al. Radiation-induced increase in invasive
potential of human pancreatic cancer cells and its
blockade by a matrix metalloproteinase inhibitor,
CGS27023. Clin. Cancer Res. 8, 1223–1227
(2002).
84. Wasserman, J., Blomgren, H., Rotstein, S., Petrini, B.
& Hammarstrom, S. Immunosuppression in irradiated
breast cancer patients: in vitro effect of
cyclooxygenase inhibitors. Bull. NY Acad. Med. 65,
36–44 (1989).
85. Reits, E. A. et al. Radiation modulates the peptide
repertoire, enhances MHC class I expression, and
induces successful antitumor immunotherapy.
J. Exp. Med. 203, 1259–1271 (2006).
86. Zhang, B. et al. Induced sensitization of tumor stroma
leads to eradication of established cancer by T cells.
J. Exp. Med. 204, 49–55 (2007).
87. Chakraborty, M. et al. Irradiation of tumor cells
up‑regulates Fas and enhances CTL lytic activity and
CTL adoptive immunotherapy. J. Immunol. 170,
6338–6347 (2003).
88. Hall, E. J. Radiobiology for the Radiologist (Medical
Department, Harper and Row, 1973).
89. Pateras, I. S. et al. The DNA damage response and
immune signaling alliance: is it good or bad? Nature
decides when and where. Pharmacol. Ther. 154,
36–56 (2015).
90. Scheithauer, H., Belka, C., Lauber, K. & Gaipl, U. S.
Immunological aspects of radiotherapy. Radiat. Oncol.
9, 185 (2014).
91. Brown, J. M. The hypoxic cell: a target for selective
cancer therapy—eighteenth Bruce, F. Cain Memorial
Award lecture. Cancer Res. 59, 5863–5870 (1999).
92. Shultz, D. B., Diehn, M. & Loo, B. W. Jr. To SABR or
not to SABR? Indications and contraindications for
stereotactic ablative radiotherapy in the treatment of
early-stage, oligometastatic, or oligoprogressive non-
small cell lung cancer. Semin. Radiat. Oncol. 25,
78–86 (2015).
93. Bertolaccini, L., Terzi, A., Ricchetti, F. & Alongi, F.
Surgery or stereotactic ablative radiation therapy:
how will be treated operable patients with early stage
not small cell lung cancer in the next future?
Ann. Transl Med. 3, 25 (2015).
94. Cao, C. et al. Surgery versus SABR for resectable non-
small-cell lung cancer. Lancet Oncol. 16, e370–e371
(2015).
95. Zheng, X. et al. Survival outcome after stereotactic
body radiation therapy and surgery for stage I
non-small cell lung cancer: a meta-analysis.
Int. J. Radiat. Oncol. Biol. Phys. 90, 603–611 (2014).
96. Rekers, N. H. et al. Stereotactic ablative body
radiotherapy combined with immunotherapy: present
status and future perspectives. Cancer Radiother. 18,
391–395 (2014).
97. Narayan, K., Fisher, R. J., Bernshaw, D., Shakher, R. &
Hicks, R. J. Patterns of failure and prognostic factor
analyses in locally advanced cervical cancer patients
staged by positron emission tomography and treated
with curative intent. Int. J. Gynecol. Cancer 19,
912–918 (2009).
98. Narayan, K., Khaw, P., Bernshaw, D., Mileshkin, L.
& Kondalsamy-Chennakesavan, S. Prognostic
significance of lymphovascular space invasion and
nodal involvement in intermediate- and high-risk
endometrial cancer patients treated with curative
intent using surgery and adjuvant radiotherapy.
Int. J. Gynecol. Cancer 22, 260–266 (2012).
99. Fidler, I. J. The relationship of embolic homogeneity,
number, size and viability to the incidence of
experimental metastasis. Eur. J. Cancer 9, 223–227
(1973).
100. Liotta, L. A., Saidel, M. G. & Kleinerman, J. The
significance of hematogenous tumor cell clumps in
the metastatic process. Cancer Res. 36, 889–894
(1976).
101. Ceelen, W., Pattyn, P. & Mareel, M. Surgery, wound
healing, and metastasis: recent insights and clinical
implications. Crit. Rev. Oncol. Hematol. 89, 16–26
(2014).
102. Alagaratnam, T. T. & Ong, G. B. Wound implantation
— a surgical hazard. Br. J. Surg. 64, 872–875 (1977).
103. Heaney, A. & Buggy, D. J. Can anaesthetic and
analgesic techniques affect cancer recurrence or
metastasis? Br. J. Anaesthesia 109 (Suppl. 1),
i17–i28 (2012).
104. Ben-Eliyahu, S. The promotion of tumor metastasis
by surgery and stress: immunological basis and
implications for psychoneuroimmunology. Brain
Behav. Immun. 17 (Suppl. 1), 27–36 (2003).
105. Buckley, A., McQuaid, S., Johnson, P. & Buggy, D. J.
Effect of anaesthetic technique on the natural killer
cell anti-tumour activity of serum from women
undergoing breast cancer surgery: a pilot study.
Br. J. Anaesthesia 113 (Suppl. 1), i56–62 (2014).
106. Divatia, J. V. & Ambulkar, R. Anesthesia and cancer
recurrence: what is the evidence? J. Anaesthesiol. Clin.
Pharmacol. 30, 147–150 (2014).
107. Cata, J. P., Wang, H., Gottumukkala, V., Reuben, J.
& Sessler, D. I. Inflammatory response,
immunosuppression, and cancer recurrence after
perioperative blood transfusions. Br. J. Anaesthesia
110, 690–701 (2013).
108. Demicheli, R., Retsky, M. W., Hrushesky, W. J.,
Baum, M. & Gukas, I. D. The effects of surgery on
tumor growth: a century of investigations. Ann. Oncol.
19, 1821–1828 (2008).
109. Horowitz, M., Neeman, E., Sharon, E. &
Ben-Eliyahu, S. Exploiting the critical perioperative
period to improve long-term cancer outcomes.
Nat. Rev. Clin. Oncol. 12, 213–226 (2015).
110. Holmgren, L., O’Reilly, M. S. & Folkman, J. Dormancy
of micrometastases: balanced proliferation and
apoptosis in the presence of angiogenesis
suppression. Nat. Med. 1, 149–153 (1995).
111. Abramovitch, R., Marikovsky, M., Meir, G. &
Neeman, M. Stimulation of tumour growth by wound-
derived growth factors. Br. J. Cancer 79, 1392–1398
(1999).
112. Mathenge, E. G. et al. Core needle biopsy of breast
cancer tumors increases distant metastases in a
mouse model. Neoplasia 16, 950–960 (2014).
113. Moreno, J. G. et al. Transrectal ultrasound-guided
biopsy causes hematogenous dissemination of
prostate cells as determined by RT‑PCR. Urology 49,
515–520 (1997).
114. Polascik, T. J. et al. Influence of sextant prostate
needle biopsy or surgery on the detection and harvest
of intact circulating prostate cancer cells. J. Urol. 162,
749–752 (1999).
115. Dyavanagoudar, S., Kale, A., Bhat, K. &
Hallikerimath, S. Reverse transcriptase polymerase
chain reaction study to evaluate dissemination of
cancer cells into circulation after incision biopsy in oral
squamous cell carcinoma. Indian J. Dental Res. 19,
315–319 (2008).
116. Kusukawa, J. et al. Dissemination of cancer cells into
circulation occurs by incisional biopsy of oral
squamous cell carcinoma. J. Oral Pathol. Med. 29,
303–307 (2000).
REVIEWS
NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 43
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
117. Zoubek, A. et al. Mobilization of tumour cells during
biopsy in an infant with Ewing sarcoma. Eur. J. Pediatr.
155, 373–376 (1996).
118. Uchida, N., Suda, T., Inoue, T., Fujiwara, Y. &
Ishiguro, K. Needle track dissemination of follicular
thyroid carcinoma following fine-needle aspiration
biopsy: report of a case. Surg. Today 37, 34–37
(2007).
119. Jones, O. M., Rees, M., John, T. G., Bygrave, S.
& Plant, G. Biopsy of resectable colorectal liver
metastases causes tumour dissemination and
adversely affects survival after liver resection.
Br. J. Surg. 92, 1165–1168 (2005).
120. Rodgers, M. S., Collinson, R., Desai, S., Stubbs, R. S.
& McCall, J. L. Risk of dissemination with biopsy of
colorectal liver metastases. Dis. Colon Rectum 46,
454–458; discussion 458–459 (2003).
121. Myrvang, H. Diagnosis: novel anti-seeding technology
to prevent dissemination of tumor cells after a fine-
needle aspiration biopsy. Nat. Rev. Clin. Oncol. 8, 64
(2011).
122. Wiksell, H. et al. Prevention of tumour cell
dissemination in diagnostic needle procedures.
Br. J. Cancer 103, 1706–1709 (2010).
123. Koch, M. et al. Hematogenous tumor cell
dissemination during colonoscopy for colorectal
cancer. Surg. Endosc. 18, 587–591 (2004).
124. Koch, M. et al. Increased detection rate and potential
prognostic impact of disseminated tumor cells in
patients undergoing endorectal ultrasound for
rectal cancer. Int. J. Colorectal Dis. 22, 359–365
(2007).
125. Peach, G., Kim, C., Zacharakis, E., Purkayastha, S. &
Ziprin, P. Prognostic significance of circulating tumour
cells following surgical resection of colorectal cancers:
a systematic review. Br. J. Cancer 102, 1327–1334
(2010).
126. Rahbari, N. N. et al. Meta-analysis shows that
detection of circulating tumor cells indicates poor
prognosis in patients with colorectal cancer.
Gastroenterology 138, 1714–1726 (2010).
127. Steinert, G., Scholch, S., Koch, M. & Weitz, J. Biology
and significance of circulating and disseminated
tumour cells in colorectal cancer. Langenbecks Arch.
Surg. 397, 535–542 (2012).
128. Weitz, J. et al. Dissemination of tumor cells in patients
undergoing surgery for colorectal cancer. Clin. Cancer
Res. 4, 343–348 (1998).
129. Rahbari, N. N. et al. Compartmental differences of
circulating tumor cells in colorectal cancer. Ann. Surg.
Oncol. 19, 2195–2202 (2012).
130. Koch, M. et al. Detection of disseminated tumor cells
in liver biopsies of colorectal cancer patients is not
associated with a worse prognosis. Ann. Surg. Oncol.
14, 810–817 (2007).
131. Wind, J. et al. Circulating tumour cells during
laparoscopic and open surgery for primary colonic
cancer in portal and peripheral blood. Eur. J. Surg.
Oncol. 35, 942–950 (2009).
132. Kienle, P. et al. Decreased detection rate of
disseminated tumor cells of rectal cancer patients
after preoperative chemoradiation: a first step
towards a molecular surrogate marker for neoadjuvant
treatment in colorectal cancer. Ann. Surg. 238,
324–330; discussion 330–321 (2003).
133. Rahbari, N. N. et al. Neoadjuvant radiotherapy
for rectal cancer: meta-analysis of randomized
controlled trials. Ann. Surg. Oncol. 20, 4169–4182
(2013).
134. Nesteruk, D. et al. Evaluation of prognostic
significance of circulating tumor cells detection in
rectal cancer patients treated with preoperative
radiotherapy: prospectively collected material data.
BioMed Res. Int. 2014, 712827 (2014).
135. Demicheli, R., Biganzoli, E., Boracchi, P., Greco, M. &
Retsky, M. W. Recurrence dynamics does not depend
on the recurrence site. Breast Cancer Res. 10, R83
(2008).
136. Fortin, A., Larochelle, M., Laverdiere, J., Lavertu, S.
& Tremblay, D. Local failure is responsible for the
decrease in survival for patients with breast
cancer treated with conservative surgery and
postoperative radiotherapy. J. Clin. Oncol. 17,
101–109 (1999).
137. Jatoi, I., Tsimelzon, A., Weiss, H., Clark, G. M. &
Hilsenbeck, S. G. Hazard rates of recurrence following
diagnosis of primary breast cancer. Breast Cancer
Res. Treat. 89, 173–178 (2005).
138. Demicheli, R. et al. Breast cancer recurrence dynamics
following adjuvant CMF is consistent with tumor
dormancy and mastectomy-driven acceleration of
the metastatic process. Ann. Oncol. 16, 1449–1457
(2005).
139. Hermansson, U., Konstantinov, I. E. & Aren, C. Tumor
dissemination after video-assisted thoracic surgery:
what does it mean? J. Thorac. Cardiovasc. Surg. 114,
300–302 (1997).
140. Yamashita, J. I., Kurusu, Y., Fujino, N., Saisyoji, T. &
Ogawa, M. Detection of circulating tumor cells in
patients with non-small cell lung cancer undergoing
lobectomy by video-assisted thoracic surgery:
a potential hazard for intraoperative hematogenous
tumor cell dissemination. J. Thorac. Cardiovasc. Surg.
119, 899–905 (2000).
141. Hashimoto, M. et al. Significant increase in circulating
tumour cells in pulmonary venous blood during
surgical manipulation in patients with primary lung
cancer. Interact. Cardiovasc. Thorac. Surg. 18,
775–783 (2014).
142. Dong, Q. et al. Hematogenous dissemination of lung
cancer cells during surgery: quantitative detection by
flow cytometry and prognostic significance. Lung Cancer
37, 293–301 (2002).
143. Eschwege, P. et al. Haematogenous dissemination of
prostatic epithelial cells during radical prostatectomy.
Lancet 346, 1528–1530 (1995).
144. Daskalakis, M. et al. Assessment of the effect of
surgery on the kinetics of circulating tumour cells
in patients with operable breast cancer based on
cytokeratin‑19 mRNA detection. Eur. J. Surg. Oncol.
37, 404–410 (2011).
145. Engilbertsson, H. et al. Transurethral bladder tumor
resection can cause seeding of cancer cells into the
bloodstream. J. Urol. 193, 53–57 (2015).
146. Nakashima, S. et al. Clinical significance of circulating
tumor cells in blood by molecular detection and tumor
markers in esophageal cancer. Surgery 133, 162–169
(2003).
147. Kayalar, N. et al. Concomitant surgery for renal
neoplasm with pulmonary tumor embolism.
J. Thorac. Cardiovasc. Surg. 139, 320–325 (2010).
148. Podgrabinska, S. & Skobe, M. Role of lymphatic
vasculature in regional and distant metastases.
Microvasc. Res. 95, 46–52 (2014).
149. Liao, C. et al. Prognostic value of circulating
inflammatory factors in non-small cell lung cancer:
a systematic review and meta-analysis. Cancer
Biomarkers 14, 469–481 (2014).
150. Song, P. P., Zhang, W., Zhang, B., Liu, Q. & Du, J.
Effects of different sequences of pulmonary artery
and vein ligations during pulmonary lobectomy on
blood micrometastasis of non-small cell lung cancer.
Oncol. Lett. 5, 463–468 (2013).
151. Smerage, J. B. et al. Circulating tumor cells and
response to chemotherapy in metastatic breast
cancer: SWOG S0500. J. Clin. Oncol. 32, 3483–3489
(2014).
152. Paez-Ribes, M. et al. Antiangiogenic therapy elicits
malignant progression of tumors to increased local
invasion and distant metastasis. Cancer Cell 15,
220–231 (2009).
153. Ebos, J. M. et al. Accelerated metastasis after short-
term treatment with a potent inhibitor of tumor
angiogenesis. Cancer Cell 15, 232–239 (2009).
154. Ebos, J. M. & Kerbel, R. S. Antiangiogenic therapy:
impact on invasion, disease progression, and
metastasis. Nat. Rev. Clin. Oncol. 8, 210–221 (2011).
155. Obenauf, A. C. et al. Therapy-induced tumour
secretomes promote resistance and tumour
progression. Nature 520, 368–372 (2015).
156. Itescu, S. et al. Intravenous pulse administration of
cyclophosphamide is an effective and safe treatment
for sensitized cardiac allograft recipients. Circulation
105, 1214–1219 (2002).
157. Schiavoni, G. et al. Cyclophosphamide induces type I
interferon and augments the number of CD44hi
T lymphocytes in mice: implications for strategies
of chemoimmunotherapy of cancer. Blood 95,
2024–2030 (2000).
158. Man, S., Zhang, Y., Gao, W., Yan, L. & Ma, C.
Cyclophosphamide promotes pulmonary metastasis
on mouse lung adenocarcinoma. Clin. Exp. Metastasis
25, 855–864 (2008).
159. Yamauchi, K. et al. Induction of cancer metastasis
by cyclophosphamide pretreatment of host mice:
an opposite effect of chemotherapy. Cancer Res. 68,
516–520 (2008).
160. Price, J. T. et al. The heat shock protein 90 inhibitor,
17‑allylamino-17‑demethoxygeldanamycin, enhances
osteoclast formation and potentiates bone metastasis
of a human breast cancer cell line. Cancer Res. 65,
4929–4938 (2005).
161. Cristofanilli, M. et al. Circulating tumor cells, disease
progression, and survival in metastatic breast cancer.
N. Engl. J. Med. 351, 781–791 (2004).
162. Cohen, S. J. et al. Relationship of circulating tumor
cells to tumor response, progression-free survival, and
overall survival in patients with metastatic colorectal
cancer. J. Clin. Oncol. 26, 3213–3221 (2008).
163. Brugger, W. et al. Mobilization of peripheral blood
progenitor cells by sequential administration of
interleukin‑3 and granulocyte-macrophage colony-
stimulating factor following polychemotherapy with
etoposide, ifosfamide, and cisplatin. Blood 79,
1193–1200 (1992).
164. Inhestern, J. et al. Prognostic role of circulating tumor
cells during induction chemotherapy followed by
curative surgery combined with postoperative
radiotherapy in patients with locally advanced oral
and oropharyngeal squamous cell cancer. PLoS ONE
10, e0132901 (2015).
165. Mego, M. et al. Prognostic value of EMT-circulating
tumor cells in metastatic breast cancer patients
undergoing high-dose chemotherapy with autologous
hematopoietic stem cell transplantation. J. Cancer 3,
369–380 (2012).
166. Hamilton, B. K. et al. Long-term survival after high-
dose chemotherapy with autologous hematopoietic
cell transplantation in metastatic breast cancer.
Hematol. Oncol. Stem Cell Ther. 8, 115–124
(2015).
167. Kuderer, N. M., Dale, D. C., Crawford, J. &
Lyman, G. H. Impact of primary prophylaxis with
granulocyte colony-stimulating factor on febrile
neutropenia and mortality in adult cancer patients
receiving chemotherapy: a systematic review.
J. Clin. Oncol. 25, 3158–3167 (2007).
168. Cao, Y. et al. BMP4 inhibits breast cancer metastasis
by blocking myeloid-derived suppressor cell activity.
Cancer Res. 74, 5091–5102 (2014).
169. Kowanetz, M. et al. Granulocyte-colony stimulating
factor promotes lung metastasis through mobilization
of Ly6G+Ly6C+ granulocytes. Proc. Natl Acad. Sci.
USA 107, 21248–21255 (2010).
170. Debeljak, N., Solar, P. & Sytkowski, A. J.
Erythropoietin and cancer: the unintended
consequences of anemia correction. Front. Immunol.
5, 563 (2014).
171. Shenouda, G. et al. Long-term results of radiation
therapy oncology group 9903: a randomized phase 3
trial to assess the effect of erythropoietin on local-
regional control in anemic patients treated with
radiation therapy for squamous cell carcinoma of the
head and neck. Int. J. Radiat. Oncol. Biol. Phys. 91,
907–915 (2015).
172. NSCLC Meta-analysis Collaborative Group.
Preoperative chemotherapy for non-small-cell lung
cancer: a systematic review and meta-analysis of
individual participant data. Lancet 383, 1561–1571
(2014).
173. Liang, Y. & Wakelee, H. A. Adjuvant chemotherapy of
completely resected early stage non-small cell lung
cancer (NSCLC). Transl Lung Cancer Res. 2, 403–410
(2013).
174. Mauri, D., Pavlidis, N. & Ioannidis, J. P. Neoadjuvant
versus adjuvant systemic treatment in breast cancer:
ameta-analysis. J. Natl Cancer Inst. 97, 188–194
(2005).
175. Mac Manus, M. P. et al. Metabolic (FDG-PET)
response after radical radiotherapy/
chemoradiotherapy for non-small cell lung cancer
correlates with patterns of failure. Lung Cancer 49,
95–108 (2005).
Acknowledgements
We are grateful to David Ball for fruitful discussions and his
continuous support of our scientific initiatives, and to
Bernhard Riedel for his critical reading of the manuscript. We
thank Tim Akhurst for his assistance with Figure 3. O.A.M.
and M.P.M. receive support from the Australian National
Health and Medical Research Council (NHMRC) grant
1104139, and the Peter MacCallum Cancer Foundation
grant 1218. R.L.A. receives fellowship support from the
National Breast Cancer Foundation of Australia.
Author contributions
O.A.M., R.L.A. and M.P.M. researched data for the article,
and K.N. contributed to discussion of the article’s content. All
authors wrote, reviewed and edited the manuscript before
submission.
Competing interests statement
The authors declare no competing interests.
REVIEWS
44 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
View publication stats

More Related Content

Similar to Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nature reviews Clinical oncology. nrclinonc_2016.pdf

Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Peter Pachmann
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...daranisaha
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...AnonIshanvi
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...JohnJulie1
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...semualkaira
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...semualkaira
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...NainaAnon
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...semualkaira
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...EditorSara
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...EditorSara
 
Comprehensive Notes on the Molecular Basis of Cancer
Comprehensive Notes on the Molecular Basis of CancerComprehensive Notes on the Molecular Basis of Cancer
Comprehensive Notes on the Molecular Basis of Cancermeducationdotnet
 
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview 1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview KiyokoSlagleis
 
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview 1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview AnastaciaShadelb
 
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer Metastasis (PMMH)
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer  Metastasis (PMMH)Introducing the Prodotis M1-Macrophage Hypothesis of Cancer  Metastasis (PMMH)
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer Metastasis (PMMH)banafsheh61
 
METASTATIC TUMORS OF THE JAW
METASTATIC  TUMORS OF THE JAWMETASTATIC  TUMORS OF THE JAW
METASTATIC TUMORS OF THE JAWUpama Sishan
 
Potential and Common Sites for Metastasis      The potential sit.docx
Potential and Common Sites for Metastasis      The potential sit.docxPotential and Common Sites for Metastasis      The potential sit.docx
Potential and Common Sites for Metastasis      The potential sit.docxLacieKlineeb
 
Liquid Biopsy Cancer Prevention and Monitoring
Liquid Biopsy Cancer Prevention and MonitoringLiquid Biopsy Cancer Prevention and Monitoring
Liquid Biopsy Cancer Prevention and MonitoringDavid Tjahjono,MD,MBA(UK)
 

Similar to Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nature reviews Clinical oncology. nrclinonc_2016.pdf (20)

Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
Chemosensitivity Testing of Circulating Epithelial Tumor Cells (CETC) in Vitr...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
Circulating Tumor Cells and Cell-Free Nucleic Acids as Predictor Factors for ...
 
Comprehensive Notes on the Molecular Basis of Cancer
Comprehensive Notes on the Molecular Basis of CancerComprehensive Notes on the Molecular Basis of Cancer
Comprehensive Notes on the Molecular Basis of Cancer
 
Advances in thymoma_imaging
Advances in thymoma_imagingAdvances in thymoma_imaging
Advances in thymoma_imaging
 
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview 1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
 
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview 1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
1578185 - McGraw-Hill Professional ©CHAPTER 175Overview
 
LRP research plan final
LRP research plan finalLRP research plan final
LRP research plan final
 
Nrclinonc.2011.30
Nrclinonc.2011.30Nrclinonc.2011.30
Nrclinonc.2011.30
 
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer Metastasis (PMMH)
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer  Metastasis (PMMH)Introducing the Prodotis M1-Macrophage Hypothesis of Cancer  Metastasis (PMMH)
Introducing the Prodotis M1-Macrophage Hypothesis of Cancer Metastasis (PMMH)
 
METASTATIC TUMORS OF THE JAW
METASTATIC  TUMORS OF THE JAWMETASTATIC  TUMORS OF THE JAW
METASTATIC TUMORS OF THE JAW
 
Potential and Common Sites for Metastasis      The potential sit.docx
Potential and Common Sites for Metastasis      The potential sit.docxPotential and Common Sites for Metastasis      The potential sit.docx
Potential and Common Sites for Metastasis      The potential sit.docx
 
Liquid Biopsy Cancer Prevention and Monitoring
Liquid Biopsy Cancer Prevention and MonitoringLiquid Biopsy Cancer Prevention and Monitoring
Liquid Biopsy Cancer Prevention and Monitoring
 

Recently uploaded

User Guide: Pulsar™ Weather Station (Columbia Weather Systems)
User Guide: Pulsar™ Weather Station (Columbia Weather Systems)User Guide: Pulsar™ Weather Station (Columbia Weather Systems)
User Guide: Pulsar™ Weather Station (Columbia Weather Systems)Columbia Weather Systems
 
OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024innovationoecd
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxNandakishor Bhaurao Deshmukh
 
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...D. B. S. College Kanpur
 
Neurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trNeurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trssuser06f238
 
User Guide: Orion™ Weather Station (Columbia Weather Systems)
User Guide: Orion™ Weather Station (Columbia Weather Systems)User Guide: Orion™ Weather Station (Columbia Weather Systems)
User Guide: Orion™ Weather Station (Columbia Weather Systems)Columbia Weather Systems
 
Pests of jatropha_Bionomics_identification_Dr.UPR.pdf
Pests of jatropha_Bionomics_identification_Dr.UPR.pdfPests of jatropha_Bionomics_identification_Dr.UPR.pdf
Pests of jatropha_Bionomics_identification_Dr.UPR.pdfPirithiRaju
 
Topic 9- General Principles of International Law.pptx
Topic 9- General Principles of International Law.pptxTopic 9- General Principles of International Law.pptx
Topic 9- General Principles of International Law.pptxJorenAcuavera1
 
Functional group interconversions(oxidation reduction)
Functional group interconversions(oxidation reduction)Functional group interconversions(oxidation reduction)
Functional group interconversions(oxidation reduction)itwameryclare
 
Davis plaque method.pptx recombinant DNA technology
Davis plaque method.pptx recombinant DNA technologyDavis plaque method.pptx recombinant DNA technology
Davis plaque method.pptx recombinant DNA technologycaarthichand2003
 
Environmental Biotechnology Topic:- Microbial Biosensor
Environmental Biotechnology Topic:- Microbial BiosensorEnvironmental Biotechnology Topic:- Microbial Biosensor
Environmental Biotechnology Topic:- Microbial Biosensorsonawaneprad
 
Bioteknologi kelas 10 kumer smapsa .pptx
Bioteknologi kelas 10 kumer smapsa .pptxBioteknologi kelas 10 kumer smapsa .pptx
Bioteknologi kelas 10 kumer smapsa .pptx023NiWayanAnggiSriWa
 
The dark energy paradox leads to a new structure of spacetime.pptx
The dark energy paradox leads to a new structure of spacetime.pptxThe dark energy paradox leads to a new structure of spacetime.pptx
The dark energy paradox leads to a new structure of spacetime.pptxEran Akiva Sinbar
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝soniya singh
 
User Guide: Magellan MX™ Weather Station
User Guide: Magellan MX™ Weather StationUser Guide: Magellan MX™ Weather Station
User Guide: Magellan MX™ Weather StationColumbia Weather Systems
 
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuine
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 GenuineCall Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuine
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuinethapagita
 
Harmful and Useful Microorganisms Presentation
Harmful and Useful Microorganisms PresentationHarmful and Useful Microorganisms Presentation
Harmful and Useful Microorganisms Presentationtahreemzahra82
 
Speech, hearing, noise, intelligibility.pptx
Speech, hearing, noise, intelligibility.pptxSpeech, hearing, noise, intelligibility.pptx
Speech, hearing, noise, intelligibility.pptxpriyankatabhane
 
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxGenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxBerniceCayabyab1
 
Good agricultural practices 3rd year bpharm. herbal drug technology .pptx
Good agricultural practices 3rd year bpharm. herbal drug technology .pptxGood agricultural practices 3rd year bpharm. herbal drug technology .pptx
Good agricultural practices 3rd year bpharm. herbal drug technology .pptxSimeonChristian
 

Recently uploaded (20)

User Guide: Pulsar™ Weather Station (Columbia Weather Systems)
User Guide: Pulsar™ Weather Station (Columbia Weather Systems)User Guide: Pulsar™ Weather Station (Columbia Weather Systems)
User Guide: Pulsar™ Weather Station (Columbia Weather Systems)
 
OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024OECD bibliometric indicators: Selected highlights, April 2024
OECD bibliometric indicators: Selected highlights, April 2024
 
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptxTHE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
THE ROLE OF PHARMACOGNOSY IN TRADITIONAL AND MODERN SYSTEM OF MEDICINE.pptx
 
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...
Fertilization: Sperm and the egg—collectively called the gametes—fuse togethe...
 
Neurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 trNeurodevelopmental disorders according to the dsm 5 tr
Neurodevelopmental disorders according to the dsm 5 tr
 
User Guide: Orion™ Weather Station (Columbia Weather Systems)
User Guide: Orion™ Weather Station (Columbia Weather Systems)User Guide: Orion™ Weather Station (Columbia Weather Systems)
User Guide: Orion™ Weather Station (Columbia Weather Systems)
 
Pests of jatropha_Bionomics_identification_Dr.UPR.pdf
Pests of jatropha_Bionomics_identification_Dr.UPR.pdfPests of jatropha_Bionomics_identification_Dr.UPR.pdf
Pests of jatropha_Bionomics_identification_Dr.UPR.pdf
 
Topic 9- General Principles of International Law.pptx
Topic 9- General Principles of International Law.pptxTopic 9- General Principles of International Law.pptx
Topic 9- General Principles of International Law.pptx
 
Functional group interconversions(oxidation reduction)
Functional group interconversions(oxidation reduction)Functional group interconversions(oxidation reduction)
Functional group interconversions(oxidation reduction)
 
Davis plaque method.pptx recombinant DNA technology
Davis plaque method.pptx recombinant DNA technologyDavis plaque method.pptx recombinant DNA technology
Davis plaque method.pptx recombinant DNA technology
 
Environmental Biotechnology Topic:- Microbial Biosensor
Environmental Biotechnology Topic:- Microbial BiosensorEnvironmental Biotechnology Topic:- Microbial Biosensor
Environmental Biotechnology Topic:- Microbial Biosensor
 
Bioteknologi kelas 10 kumer smapsa .pptx
Bioteknologi kelas 10 kumer smapsa .pptxBioteknologi kelas 10 kumer smapsa .pptx
Bioteknologi kelas 10 kumer smapsa .pptx
 
The dark energy paradox leads to a new structure of spacetime.pptx
The dark energy paradox leads to a new structure of spacetime.pptxThe dark energy paradox leads to a new structure of spacetime.pptx
The dark energy paradox leads to a new structure of spacetime.pptx
 
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
Call Girls in Munirka Delhi 💯Call Us 🔝8264348440🔝
 
User Guide: Magellan MX™ Weather Station
User Guide: Magellan MX™ Weather StationUser Guide: Magellan MX™ Weather Station
User Guide: Magellan MX™ Weather Station
 
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuine
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 GenuineCall Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuine
Call Girls in Majnu Ka Tilla Delhi 🔝9711014705🔝 Genuine
 
Harmful and Useful Microorganisms Presentation
Harmful and Useful Microorganisms PresentationHarmful and Useful Microorganisms Presentation
Harmful and Useful Microorganisms Presentation
 
Speech, hearing, noise, intelligibility.pptx
Speech, hearing, noise, intelligibility.pptxSpeech, hearing, noise, intelligibility.pptx
Speech, hearing, noise, intelligibility.pptx
 
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptxGenBio2 - Lesson 1 - Introduction to Genetics.pptx
GenBio2 - Lesson 1 - Introduction to Genetics.pptx
 
Good agricultural practices 3rd year bpharm. herbal drug technology .pptx
Good agricultural practices 3rd year bpharm. herbal drug technology .pptxGood agricultural practices 3rd year bpharm. herbal drug technology .pptx
Good agricultural practices 3rd year bpharm. herbal drug technology .pptx
 

Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nature reviews Clinical oncology. nrclinonc_2016.pdf

  • 1. See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/306401849 Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Article in Nature Reviews Clinical Oncology · August 2016 DOI: 10.1038/nrclinonc.2016.128 CITATIONS 144 READS 1,508 4 authors: Olga A Martin University of Wollongong 143 PUBLICATIONS 13,568 CITATIONS SEE PROFILE Robin L Anderson Olivia Newton-John Cancer Research Institute 234 PUBLICATIONS 9,019 CITATIONS SEE PROFILE Kailash Narayan Peter MacCallum Cancer Centre 124 PUBLICATIONS 2,517 CITATIONS SEE PROFILE Michael P Macmanus University of Melbourne 333 PUBLICATIONS 9,527 CITATIONS SEE PROFILE All content following this page was uploaded by Olga A Martin on 26 June 2018. The user has requested enhancement of the downloaded file.
  • 2. Patients with the most common carcinomas, such as those affecting the lung, prostate, bowel or breast, fre- quently die from distant metastasis, even after curative-­ intent surgery, radiotherapy, or chemoradiotherapy for apparently locoregionally confined disease. Distant metastases that become apparent after successful treat- ment to achieve locoregional control are considered to derive from micrometastases that were present before treatment commenced1–3 . In patients for whom attempts to eliminate locoregionally confined disease have failed, distant metastasis might also derive from residual local disease4 . Another unwelcome possibility is that metastasis can sometimes be caused or promoted by the antitumour treatment itself. Events surrounding treatment with sur- gery, radiotherapy, and systemic antitumour therapies can perturb tumours in complex ways that might influence either positively or negatively the risk of distant metas- tasis. Evidence indicates that all three modalities might, under some ­ circumstances, mobilize tumour cells into the bloodstream. Circulating tumour cells (CTCs) can be detected in patients with all of the major carcinomas and are necessary for distant metastasis to occur5 . In general, although patients with high numbers of CTCs have a poor prognosis6 , the mobilization of CTCs induced by a therapeutic or diagnostic intervention is not necessarily associated with an adverse outcome. Metastasis is some- times regarded as a linear process, whereby tumour cells acquire enhanced migratory and invasive capacities, enter lymphatic and blood vessels, attain both the capacity to survive the harsh environment of the circulation and the ability to extravasate and proliferate to form a metastatic lesion, in a new and potentially unfavourable microenvi- ronment7 . Luzzi et al.8 reported that most CTCs can sur- vivetheshearforcesthattheyencounterinthecirculation, enabling them to lodge in capillary beds and often extra­ vasate into tissues. The most common outcome in such situations is cell death, but a small proportion of tumour cells (approximately 0.02%) survive and, after a vari­ able period of latency, expand into clinically detectable lesions. In a modelling study, Coumans et al.9 assessed data from a large cohort of patients with breast cancer and estimated thatthemetastaticrateismuchlowerthanthatreportedin thestudydiscussed4 :onlyonemetastasisformsfromevery 60 million cells that disseminate from the primary site. The time to first metastasis varies widely among different malignancies, owing to their diverse natural histories. In many patients with apparently localised 1 Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre. 2 Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre. 3 Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia. 4 The Sir Peter MacCallum Department of Oncology, University of Melbourne. 5 Department of Obstetrics and Gynaecology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia. Correspondence to M.P.M. Michael.macmanus@ petermac.org doi:10.1038/nrclinonc.2016.128 Published online 23 Aug 2016 Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Olga A. Martin1,2,4 , Robin L. Anderson3,4 , Kailash Narayan1,4,5 and Michael P. MacManus1,4 Abstract|Despiteprogressiveimprovementsinthemanagementofpatientswithlocoregionally confined,advanced-stagesolidtumours,distantmetastasisremainsaverycommon—andusually fatal—modeoffailureafterattemptedcurativetreatment.Surgeryandradiotherapyarethe primarycurativemodalitiesforthesepatients,oftencombinedwitheachotherand/orwith chemotherapy.Distantmetastasisoccurringaftertreatmentcanarisefrompreviouslyundetected micrometastasesor,alternatively,frompersistentlocoregionaldisease.Anotherpossibilityisthat treatmentitselfmightsometimescauseorpromotemetastasis.Surgicalinterventionsinpatients withcancer,includingbiopsies,arecommonlyassociatedwithincreasedconcentrationsof circulatingtumourcells(CTCs).HighCTCnumbersareassociatedwithanunfavourableprognosis inmanycancers.RadiotherapyandsystemicantitumourtherapiesmightalsomobilizeCTCs.We reviewthepreclinicalandclinicaldataconcerningcancertreatments,CTCmobilizationandother factorsthatmightpromotemetastasis.Contemporarytreatmentregimensrepresentthebest availablecurativeoptionsforpatientswhomightotherwisediefromlocallyconfined, advanced-stagecancers;however,ifsuchtreatmentscanpromotemetastasis,thisprocessmust beunderstoodandaddressedtherapeuticallytoimprovepatientsurvival. REVIEWS 32 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 3. cancers, metastasis occurs early, even before diagnosis3 . This discovery has invoked an alternative parallel pro- gression model of metastasis, whereby tumour cells that disseminate early evolve independently, but in parallel, from the primary tumour10,11 . Disseminated tumour cells might lie dormant in bone marrow and/or other tissues for many years. These occult cells can enter a G0-like state and remain as quiescent single cells, or they can expand to form a cluster that is unable to expand further, possibly owing to effective immune surveillance, or the inability to trigger angiogenesis12 . In either case, these micrometastases are presumed to be the source of the CTCs that are detected after primary tumour control in many patients. Genomic analyses13,14 have reported ‘early’ and ‘late’ metastases, an observa- tion confounded by the fact that a metastasis can seed ­secondary metastases. Curing the most-common epithelial and mesen- chymal malignancies is generally impossible without loco­ regional control provided by surgery and/or radio- therapy; however, if such potentially curative therapies are also able to mobilize tumour cells into the circulation, or to promote the growth of dormant micro­ metastases, a detailed understanding of these events is essential. This knowledge could lead to the design of new thera­ peutic approaches capable of increasing the cure rates for ­ cancers for which metastasis is a frequent cause of treatment failure. In this Review, we examine the pre- clinical and clinical effects of the major modalities used in curative treatment of epithelial cancers on the mobil­ ization of tumour cells into the circulation, and consider other treatment-related factors that could potentially modulate the risk of metastasis. When evaluating these data, we have considered in vitro and animal studies sep- arately from clinical studies, with the latter constituting a higher level of evidence because of their direct relevance to patients. Circulating tumour cells Both CTCs and cell-free tumour-derived DNA are com- monly found in the blood of patients with carcinoma, including those who will never develop overt metastatic disease. This observation reflects the extremely inefficient natureofmetastasis8 .Despitethisinefficiency,thenumber of CTCs and cell-free tumour DNA found in the circula- tion has both prognostic and predictive value in several different cancer types15–18 . CTCs escape from the primary tumour environment through an active process, in which they acquire expres- sion of genes involved in migration and invasion, and of genes that switch their morphology to a mesenchymal-­ like motile phenotype. Epithelial-to‑mesenchymal tran- sition (EMT) has been investigated in many settings and evidence for its role in driving the release of CTCs is growing19,20 . However, EMT is not essential for tumour cell escape; an elevated intratumour interstitial pressure, an incompetent or damaged tumour vasculature, and migration of actively motile cells can collectively result in the entry of single CTCs or clusters of cells into the termi- nal lymphatics or bloodstream21–23 . Both in clinical sam- ples and in preclinical tumour models, tumour cells have been shown to be associated with macrophages during migration towards blood vessels, where they interact with endothelial cells before intravasation24,25 . The presence of CTCs with both epithelial and mesenchymal properties has been reported in blood samples from patients with breast20,26 , prostate26 and lung27 ­ cancer; cell clusters, com- posed in part of tumour cells, have also been detected in the circulation of these patients, and their presence is associated with an earlier onset of metastatic disease ­ compared with single CTCs28–31 . The ability of CTCs to lodge in capillaries and expand to form macrometastases has been demonstrated in sev- eral studies, including a report using tumour xenografts generated from blood samples of patients with metastatic prostate or colon cancer32 . In subsequent studies, CTCs from patients with breast and small-cell lung ­ cancer (SCLC) injected into immunologically ­ compromised mice resulted in tumour formation33–35 . Researchers have reported that breast and prostate CTCs can be propagated in vitro for prolonged ­ periods of time (>6 months in some cases) as spheroids or ­organoids32,36 — a hallmark of cancer stem cells (CSCs) that are known to have a high capacity for self-renewal, a radioresistance and chemoresistance phenotype, and a high metastatic potential37,38 . The cell lines and tumours derived from these CTCs share histological and immuno­ chemical features and a mutational landscape similar to the primary tumour, indicating that their biologi- cal properties remain after long-term culture. In one study35 , multiple tumour cell lines could be established from six out of 36 patients with breast cancer from whom CTCs had been obtained during chemotherapy, but not from CTCs harvested before treatment, indicating that the CTCs isolated during treatment were more numer- ous and/or more autonomous in their growth require- ments than those obtained before treatment. Taken together, these data demonstrate that, in addition to being prognostic or predictive markers, CTCs can also be ­metastasis-initiating cells. While, theoretically, CTCs can access all tissues in the body, secondary tumours tend to arise in selected tissues, depending on the type of cancer. This observation is the basis of the famous ‘seed and soil’ hypothesis proposed over 125 years ago by Stephen Paget39 . Studies conducted Key points • Distant metastasis remains a frequent cause of death, even after locoregional disease control is achieved using surgery, radiotherapy, and/or systemic therapy • Antitumour therapies can, under some circumstances, mobilize tumour cells into the peripheral circulation that might influence the risk of distant metastasis • Irradiation can enable tumour cells to acquire properties that facilitate their dissemination and the subsequent generation of metastases • Severalmechanismsmightexplainametastasis-promotingeffectderivedfromsurgical procedures,suchastissuedisruptionandleakageofbloodcontainingtumourcells • A potential link between systemic therapies and metastasis has not been established, but the results of isolated studies indicate that this question needs to be addressed • The risk of distant failure from antitumour therapy can potentially be reduced if treatment-related factors capable of promoting metastasis are identified, and targeted therapeutically REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 33 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 4. over the past decade have indicated that CTCs ‘home’ to sites within tissues in which tumour-promoting stro- mal cells — including cells of the myeloid, fibroblast and endothelial lineages — have already been recruited and provide a supportive niche for the tumour cell40,41 . The proposed mechanism by which the primary tumour initiates the formation of the premetastatic niche is by secreting factors, either as freely circulating molecules or encapsulated within exosomes, that direct stromal cells to tissue sites (or modulate the phenotypes of cells resident at the site) that later attract CTCs42,43 . Whether the for- mation of a niche is essential for metastases to develop, and whether the early arrival of CTCs might stimulate the initiation of the niche remains to be determined. Radiotherapy Preclinical studies In the 1940s, the suspicion that an increased rate of metastasis might follow radiotherapy led to testing of this hypothesis in animal tumour models. The initial investi­ gations, summarized by von Essen44 , involved a range of tumour-bearing animals irradiated at vari­ ous doses in single or multiple fractions. In general, these ­ studies indicated that irradiation of primary tumours with lower doses (insufficient for local ­ control) was associated with the highest risk of metastases. Kaplan and Murphy45 observed an unexpectedly high number of early distant metastases in patients with epidermoid carcinoma of the lower lip and buccal mucosa treated with radiotherapy. In 1949, they conducted a series of animal experiments using subcutaneously transplanted murine mammary carcinomas that were locally irradiated with 4–10Gy (non-curative) in single X‑ray doses. Within 8 weeks, pulmonary metastases were found in 43.5% of irradiated mice and 9.6% of controls. Those metastases were exclu- sively pulmonary and grew intravascularly in arteries and arterioles. The irradiated tumours recurred locally within 2 weeks. In 1976, Sheldon and Fowler46 irradiated mammary carcinomas implanted on the anterior chest wall of syngeneic mice using either a single dose of 5Gy or two fractions of 3.5Gy; excision of the tumours was performed while the mice were under anaesthesia for the second (real or sham) irradiation. Pulmonary metastasis occurred 10 weeks later in 20% of non-­ irradiated mice, 25% of mice that received 5Gy and 30% of those that received two doses of 3.5Gy, indicating that the metas- tasis rate was enhanced by ­ two-fraction irradiation ­ compared with surgical removal alone. In the past 15 years47–51 , investigators have sought to explain this phenomenon. For example, Camphausen and colleagues47 injected Lewis lung carcinoma cells into the hindlimbs of mice and irradiated the resulting tumours. Local disease control was achieved in 71% of the irradiated mice, but these mice had a higher number of lung metastases when compared with non-­ irradiated mice. In further experiments involving mice that had been injected with fibrosarcoma-derived cells, irradiation markedly increased the numbers of pul- monary metastases compared with those detected in non-­ irradiated tumour-bearing mice. The researchers suggested that previously dormant metastases had been activated by radiation47 , in agreement with von Essen and colleagues44 , who showed that both tumour cells and stroma are modified by radiation, promoting the selection of ­ more-aggressive and invasive cells with ­metastatic potential. Studies of tumour cell biology indicate that radio­ therapy can alter tumour cells, making them more aggressive than non-irradiated cells. The two phases of a typical course of conventionally fractionated high-dose radiotherapy (~2Gy per fraction) can have different bio- logical effects (FIG. 1). In the later phase of radiotherapy (for example, after 30 daily fractions of 2Gy), the high cumulative radiation doses will have caused permanent loss of reproductive capacity in all clonogenic tumour cells, if the treatment is successful. Tumour cell death occurs primarily through the induction of irreparable DNA damage and cell cycle arrest52 . Irradiated cells usu- ally undergo one or two error-prone cell cycles before succumbing to mitotic catastrophe, apoptosis, necrosis, or autophagy53–55 , depending on the dose. In the early stages of fractionated radiotherapy (that is, after delivery of only sublethal cumulative radiation doses, such as 2–6 Gy in 1–3 fractions), a significant proportion of the irradiated tumour cells can repair the DNA damage; tumour cells are much more likely to survive if they escape into the circulation at this stage than the more-heavily irradiated cells that enter the circulation later in treatment. Cellular radioresistance and radiosensitivity depend on many factors, including cell type and origin, cell-­cycle phase, tissue oxygenation, and genetic background. CSCs are more radioresistant than non-stem cells56,57 and can selectively survive irradiation. If they are not elimi­ nated or do not undergo permanent cell-cycle arrest (senes- cence or dormancy), CSCs have the capacity to cause disease recurrence. Furthermore, non-small-cell lung carcinoma (NSCLC)-derived cells have been reported to survive 5Gy irradiation by acquiring new CSC charac­ teristics, including the capacity to form 3D spheroids in vitro, to self-renew and to generate differentiated progeny. These cells also acquire the ­more-mesenchymal phenotype typical of EMT38 . Failure to repair DNA double-strand breaks caused by radiotherapy is a major threat to genomic integrity58,59 . Irradiated tumour cells have increased genome instability and plasticity compared with unirradiated cells, and thus can become more radioresistant. Rofstad et al.48 reported that irradiation of mouse melanoma xenografts induced tumour hypoxia that, in turn, promoted metastasis by upregulating the expression of urokinase plasminogen activator surface receptor (uPAR). Injections of anti- uPAR antibodies almost completely blocked metastasis. Changes in invasiveness and biomechanical properties might also occur in irradiated cells. For example, the expression levels of the proto-oncogene MET (encoding HGFR, a known driver cell invasiveness), were fivefold higher in tumour cells irradiated at 10Gy than in non-­ irradiated tumour cells60 ; silencing of MET expression prevented radiation-induced proliferation and invasive- ness,andpromotedapoptosis.Zheng et al.61 ,reportedthat low-doseirradiation(1–4Gy)ofcellsderivedfromtongue squamous-cellcarcinomaincreasedtheirinvasivenessina REVIEWS 34 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 5. dose-dependent manner and changed some of their bio- mechanical properties, including the ­organization of their actin cytoskeleton. In 2015, a study revealed that the migratory capacity of tumour cells induced by low doses of radiation was medi- ated by G‑CSF secreted by the tumour cells62 . Human NSCLC cells expressing G‑CSF receptors were stimulated with radiation to release G‑CSF, which subsequently pro- moted EMT through an autocrine mechanism involving JAK/STAT3 signalling62 . Radiotherapy can also stimu- late primary tumour self-seeding from CTCs released either before or after radiation exposure. Vilalta and co-­workers63 used preclinical models to demonstrate the release of GM‑CSF from irradiated tumour cells stimu­ lated the migration of tumour cells into the irradiated site, thus triggering local tumour recurrence. Abundant evidence indicates that radiation can indi- rectly affect metastasis by modulating angio­ genesis, thereby affecting the biological behaviour of tumour cells. For example, radiation stimulates hypoxia-­inducible factor 1 (HIF‑1) and, subsequently, the expression of VEGF64,65 . In a clinical trial in which patients with hepato­ cellular carcinoma received either chemotherapy alone or chemotherapy plus conformal radiotherapy, an associ­ationwasobservedbetweencombinedchemoradio­ therapy and rapid tumour progression outside the irradi­ ated field66 . In cell-culture experiments performed in hypoxia-mimicking conditions, Sofia Vala et al.67 demon- strated that low doses of radiation (<0.8 Gy) led to the rapid phosphorylation of VEGFR‑2 and induced VEGF production — thereby, enhancing endothelial-cell migra- tion. Moreover, Shen et al.68 reported that irradiation of cultured cancer cells induces secretion of protein-lysine 6‑oxidase (LOX), a key player in hypoxia-dependent tumour-celldisseminationandmetastasis.IncreasedLOX secretion was also detected in mouse serum after tumour xenografts were exposed to localized radiation63 . Clinical studies of radiotherapy In some clinical situations, locoregional radiotherapy can reduce the risk of distant metastasis. For example, in patients with stage I–III breast cancer, adjuvant regional radiotherapy to the draining lymph nodes is associated with a substantial improvement in distant metastasis-­ free survival (hazard ratio (HR) 0.82), leading to a sig- nificant improvement of overall survival (HR 0.85, 95% CI 0.75–0.96; P=0.011) at 10 years69 . Nevertheless, our group reported the first clinical evidence that localized radiotherapy, when delivered to an intact tumour, can mobilize viable tumour cells into the circulation31,70 , a phenomenon that could potentially increase the risk of distant metastasis. We detected increased numbers of CTCs, both as single entities or in clusters, in the blood of patients with NSCLC early in the course of their radio­ therapy. Intense staining for phosphorylated histone H2AX (γ-H2AX), a biomarker for DNA double-strand breaks71,72 , showed that mobilized CTCs were derived from the irradiated tumour, with the highest number of CTCs mobilized after the first and second daily fractions of radiotherapy — a sublethal dose for many tumour cells (FIG. 1). Mobilized CTCs had an increased capacity to grow in culture both as attached cultures and orga- noids, compared with CTCs collected before treatment, (O. A. Martin, unpublished work); such attributes can be associated with unfavourable patient outcomes34 . In lung cancer, strong clinical evidence indicates a link between the fractionation schedule employed in radiotherapy and the subsequent risk of distant Nature Reviews | Clinical Oncology Radiation-induced DNA damage Cancer non-stem cells Cancer stem cells DNA damage repaired DNA damage unrepaired Radiation-induced cell death DNA damage misrepaired • EMT • Hypoxia • Motility • Invasiveness Initial 2 Gy radiotherapy fractions Accumulated radiotherapy fractions Increasing number of treatment fractions over time Early radiotherapy survival (<50%) Figure 1 | Progressive effects of fractionated radiotherapy on tumour cells in vivo. Up to one-half of the malignant cells in an irradiated tumour can survive the first radiotherapy fractions; they can subsequently acquire a more-aggressive phenotype, becoming circulating tumour cells that are detectable during the course of radiotherapy. Radiotherapy affects the regulation of genes associated with radioresistance, tumour aggressiveness, and enhanced metastatic potential, including signatures associated with hypoxia, invasiveness and motility, and epithelial-to‑mesenchymal transition (EMT). REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 35 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 6. metastasis. Continuous hyperfractionated accelerated radiotherapy (CHART)73 , a very intense but brief treat- ment regimen, improved survival and local disease con- trol compared with conventional prolonged radiotherapy in patients with locally advanced NSCLC. CHART was also associ­ ated with a 24% reduction in the relative risk of death, equivalent to an absolute improvement in 2‑year survival from 20% to 29% (P=0.004) between conven- tional radio­therapy and CHART. With mature follow‑­­up (at least 3 years), in patients with squamous carcinoma a 24% reduction in the relative risk of distant metasta- sis was noted (P=0.043)74 . The reduction in the distant metastatic rate in patients treated with CHART has sev- eral explanations, including a reduced probability of late metastasis from uncontrolled, compared with controlled, local disease. CHART was delivered in three fractions per day; CTCs mobilized by this radiotherapy regimen pre- sumably acquired more damage at any given time than cells exposed to daily radiotherapy — and might have a reduced capacity to form metastases. To our knowledge, no other research group has sys- tematically explored changes in CTCs during the early phase of radiotherapy. Using a telomerase-based assay, Dorsey et al.75 detected viable CTCs in serial blood samples of patients with NCSLC receiving radiotherapy (telo­ merase contributes to cancer cell immortality and is present in almost all cancer cells, but at very low levels in normal cells). Lowes et al.76 investigated CTCs using the CellSearch system (Janssen Diagnostics, Raritan, New Jersey, USA) in patients with prostate cancer before and 3 months after completion, but not during radiotherapy; patients who had increased or unchanged CTC numbers after radiotherapy experienced treatment failure. These preliminary data76 are derived from a combined popu­ lation of only 19 patients, but are consistent with the hypothesis that failure to clear CTCs might be associated with an unfavourable prognosis, an important subject for future research. Effects of radiotherapy on immunity Immune responses can be finely balanced between pro- moting metastasis and inhibiting signalling pathways affected in cancer; antitumour therapy can perturb this balance, with a positive or negative effect on the risk of metastasis77 . Systemic immunosuppression owing to antitumour therapy, including radiotherapy, could potentially increase the risk of metastasis. Individuals who have received renal and cardiac transplants have an increased incidence of cancer compared with the gen- eral population, owing to immunosuppressive thera­ pies associated with transplantation. In a study with 381 patients who received a cardiac transplant, 130 (34%) had developed a malignancy after a median fol- low up of 9.7 years78 . Immunosuppressed patients have a worse prognosis than immunocompetent patients with cancer79 . Radiotherapy induces a pronounced local inflammatory response77 , enhancing expression of inflammation-related cytokines, including TGFβ, which mediates EMT in mammalian cells80 . Several studies have examined radiation-induced EMT. For example, Park et al.49 established a mouse xenograft model using a cancer cell line expressing luciferase. After irradiation of primary tumours, the authors observed luminescent signals identified as intestinal and pulmonary metas- tases; these lesions expressed EMT markers and were histologically confirmed as metastatic tumours. The expression of several proteins50,81–83 has been associated with the acquisition of EMT characteristics by irradiated tumour cells, and could be potentially targeted to atten- uate the harmful effects of radiation and/or to enhance the efficacy of radiotherapy. Traditionally, ionizing radiation has been considered immunosuppressive because of the inherent susceptibil- ity of naive immune cells to radiation84 and thus, radio­ therapy was not deemed to contribute to anti­ tumour immunity. A growing body of evidence, however, sup- ports the possibility of irradiated tumour cells becom- ing a robust source of neoantigens, which can prompt local and systemic antitumour immune responses, in addition to direct radiation-induced lethality77 . Immune responses can depend greatly on the type of radiation-­ induced cell death (that is, mitotic catastrophe, apoptosis, necrosis, or autophagy). DNA released from irradiated tumour cells activates both innate and adoptive immune cells via different mechanisms85–87 , including boosting cytotoxic T lymphocytes that contribute to tumour-cell eradication77 . The fate of a cell damaged by radiation depends mainly on the extent and type of DNA damage and, therefore, on the absorbed dose of radiation and the regimen used88 . The DNA-damage response and signal- ling pathways involved in the immune response have been proposed to cooperate to cause cancer cell death89 . While lower radiotherapy doses (<2 Gy) can promote anti-inflammatory responses77,90 , higher doses (>15 Gy) might induce an intense inflammatory response that can influence the survival of residual tumour cells inside and outside of the irradiated area91 . Thus, a carefully selected combination of radiotherapy fractionation schedule and immune-modulating agents could enhance systemic antitumour immune responses. The most intense form of large-fraction radio­therapy is stereotactic ablative body radiotherapy (SABR), which is leading to a revolution in the management of oligometa­ static solid tumours and small primary tumours unsuitable for surgical resection, especially in lung cancer92,93 . SABR can provide non-inferior survival outcomes and local disease control comparable to sur- gery94,95 owing to improvements in linear-accelerator technology, cancer imaging, and radiotherapy-­ planning software. The delivery of very large radiotherapy frac- tions (>20 Gy), leads to high levels of cell death in both tumour and stromal cells — thereby justifying the title ‘ablative’. The profound local inflammatory response resulting from rapid cell death by SABR likely contributes to both local control and induction of a systemic immune response. This response can sometimes be capable of causing disease regression at sites distant from the irradi­ ated volume, the so‑called ‘abscopal’ effect of radiother- apy96 . The potential effect of SABR on CTC mobilization has not been studied; future comparisons of the rates of distant metastasis in matched patients treated with either surgery or SABR will be of particular interest. REVIEWS 36 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 7. Radiotherapy–metastasis interactions Direct evidence for mobilization of CTCs during radio­ therapy in humans is at present limited to reports from our own group in patients with NSCLC31 . This mobil­ ization might be due to the effect of radiation on tumour architecture,especiallyonthetumourvasculature.During fractionated radiotherapy, actively proliferating tumour and stromal cells (both radiosensitive) increasingly undergo cell death with accompanying disruption of tumour architecture, which could promote direct entry of tumour fragments into the circulation. Furthermore, tumour debris, including irradiated stromal and viable neoplastic cells, might be transported to the terminal lym- phaticsandsubsequentlyformcoloniesindraininglymph nodes, owing to mutations or other adaptations enabling them to survive in this harsh environment. Viable tumour cells might also be capable of forming distant metastases upon entry into the ­ peripheral circulation (FIG. 2). Patients with detectable lymph-node metastasis at the time of locoregional treatments are much more likely to experience distant failure than patients with node-­ negative cancer. For example, in a multivariate analysis of known prognostic factors, only lymph-node involve- ment was associated with distant relapse in patients with locally advanced cervical cancer treated with definitive radiotherapy97 . Similarly, the distant site relapse rate in patients with endometrial cancer following curative treat- ment was >66% when both lymphovascular invasion and lymph-node metastasis were present, but only 7% when these parameters were negative98 . Radiotherapy might also negatively affect CTC biol- ogy. In NSCLC, radiotherapy increased the number of CTCs detected in clusters31 , which are associated with an increased rate of metastasis in several cancers30,99,100 . Radiotherapy was also associated with an increase in the number of CTCs displaying mesenchymal-like ­characteristics31 . No direct clinical evidence exists to link an increase in radiotherapy-released CTCs or mesenchymal-like CTC characteristics associated with a worse prognosis. If the Nature Reviews | Clinical Oncology Primary lymph node Secondary lymph node Direct tumour cell entry into draining veins Venus circulation via LV anastomoses Venus circulation via LV anastomoses Venus circulation via thoracic duct Disrupted tumour blood vessel • Disrupted tumour architecture • Elevated tumour interstitial pressure • Opening of lymphatics • Entry of tumour components into lymphvasculature a Venous b Lymphatic Figure 2 | Routes of cancer cell mobilisation during treatment of an intact tumour. Tumour irradiation with doses insufficient to eliminate all tumour cells can result in the disruption of the tumour architecture and, subsequently, lead to the entry of tumour cells and other tumour components into the peripheral circulation, either directly into the venous system (panel a) or indirectly via the lymphatic system (panel b) through lymphaticovenous (LV) anastomoses, or the thoracic duct, following hydrostatic pressure gradients. Other therapeutic interventions can have a similar effect on tumour cell mobilization. REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 37 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 8. release of CTCs during radiotherapy does have prog- nostic value, then strategies designed to target CTCs or to ensure that they are critically damaged before entry into the circulation would become appealing. Such strat­ egies could include delivery of radiotherapy with larger or more-frequent fractions, appropriately timed co‑­ administration of systemic agents to eliminate mobilized CTCs,immunemodulationtoeliminateCTCs,andthera­ pies targeted at the prevention of EMT or ­ elimination of ­ mesenchymal-like tumour cells. Surgical procedures Patients with cancer can undergo a wide variety of pre- surgical and surgical procedures — ranging from external manipulation (such as endorectal ultrasound) to radical resection with extensive lymph-node dissection — all of which have the potential to perturb the tumour and lead to the release of CTCs. After resection, the surgical bed can contain, in addition to viable tumour cells, a mixture of blood, extracellular fluid and a wide range of cytokines and inflammatory cells, which can promote entry of via- ble tumour cells into the local and regional lymphatic vessels and peripheral circulation101 . Tumour cells can be inadvert­ ently implanted into surgical wounds and drain- age tube tracks, leading to loco­ regional recurrence102 . Several groups have reported that increased numbers of CTCscanbedetectedduringandaftersurgery(compared with the levels before surgery), leading to the hypoth­ esis that surgery itself could promote distant metastasis. Invasive surgical procedures are generally performed under some form of regional or general anaesthesia; the physiological effects of anaesthetic and surgical pro­ cedurescouldpotentiallyinfluencetheriskofmetastasis103 . Perioperative factors, such as paracrine and neuroendo- crine responses to surgery, could facilitate the metastatic process by directly affecting malignant tissues, or through indirect pathways, such as immunological perturbations. Surgery-related anxiety and stress, and the nutritional sta- tus might also be important factors104 . Anaesthesia itself might also impair various immune functions, including those of neutrophils, macrophages, dendritic cells, T‑cells, andnaturalkiller(NK)cells105 .Intravenouslyadministered induction agents (such as ketamine and thiopentone) and inhalational agents (such as halothane) can suppress NK‑cell activity in animal studies, and the upregulation of hypoxia-inducible factors by volatile anaesthetics can be cytoprotective for residual tumour cells106 . The pathways involved in these processes, as well as their clinical rele- vance,haveyettobeelucidated.Finally,bloodtransfusions given in the perioperative period have been shown to be associated with higher disease recurrence (odds ratio 1.6) in patients with head and neck cancer and hepatocellular carcinoma,aphenomenonattributedbysomeresearchers to immunosuppression107 . Preclinical studies Detailed reviews on the effect of surgery on tumour growth and metastasis using preclinical models have been published108,109 . Evidence supports both a favour- able and an unfavourable role of surgery in cancer out- comes. Studies that conclude a detrimental effect of surgery on subsequent outcome in preclinical models invoke Folkman’s reports of the release of antiangiogenic proteins (such as angiostatin and endostatin) from the primary tumour to prevent the expansion of metastatic lesions110 . Removal of the primary tumour relieves this brake on angiogenesis, thus accelerating metastasis. In other ­ studies, stimulation of angiogenesis through wound-healing mechanisms triggered by the surgical procedure has been demonstrated111 . A study from 2014 that used a preclinical cancer model112 described that the procedure of core-­ needle biopsies can induce distant metastasis, mediated by inflammatory cytokines secreted as a result of the wound112 . Effect of surgery on patients’ CTCs Biopsies and other diagnostic procedures. Direct evi- dence has linked needle biopsies (for example, in patients with prostate cancer3,113,114 ), and incisional biopsies (for example, in patients with oral cancers115,116 ), with the detection of increased numbers of CTCs in the circula- tion.Zoubekandcolleagues117 reportedachildwithpelvic Ewing sarcoma who had CTCs detected in the peripheral blood collected during open tumour biopsy (identified using RT‑PCR of specific hybrid transcripts), but not before nor 6 days after surgery117 . Tumours can also form along the track left by a biopsy needle, as described in a case report118 . Jones and colleagues119 retrospectively ana- lysed patients with colorectal cancer liver metastases who had undergone resection with or without a preoperative needle biopsy. These investigators reported evidence of tumours along the needle track in 17% of patients who had undergone biopsy. The survival rate was worse in patients who had undergone preopera­ tive biopsies com- pared with those patients who had not119 . A remarkably congruent local dissemination rate of 16% after liver biopsy was reported in a similar trial120 . In response to the increased awareness of tumour seeding along needle tracks, ‘anti-seeding technology’ using radiofrequency pulses has been proposed as a method to sterilize the needle and its track121,122 . In prospective studies, Koch and colleagues reported that the proportion of patients having detectable CTCs increased after colonoscopy123 and endorectal ultrasound124 . Cancer surgery. The number of publications relating CTCs, surgery, and metastasis risk has increased con- siderably over the years; herein, we discuss a selection of those studies. The effects of surgery on CTCs have been investigated extensively in patients with gastro- intestinal tumours, especially in those with colorectal cancer (CRC)125 . CRC cells often disseminate through draining nodes and vessels; the most frequent distant meta­ static site in patients with CRC is the liver, which can be accessible by laparotomy. The presence of both CTCs and disseminated tumour cells in lymph nodes and bone marrow has negative prognostic significance in CRC126,127 . Increased numbers of CTCs have been reported during and after surgery compared with base- line levels128 . At the time of surgery for CRC, CTCs can reach the liver through the portal venous circula- tion; CTCs are detected more frequently and in higher REVIEWS 38 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 9. numbers in the portal venous compartment than in the central venous compartment during surgery for CRC129 . Trapping of CTCs in the liver, however, does not neces­ sarily lead to liver metastases. Koch and colleagues130 identified CRC-derived cells in liver biopsies of 10 of 100 patients with UICC stage I–III CRC, but no correlation between the presence of these cells and the survival rate of the patients was found, suggesting that most tumour cells trapped in the liver did not survive to cause metas- tasis. In a study of 37 patients who underwent resection of liver metastases131 , however, multi­ variate analysis confirmed that tumour-cell detection in intraoperative blood and in bone marrow were independent prognos- tic factors for tumour relapse. Interestingly, Wind et al.131 reported a difference in CTC mobilization between open and laparo­ scopic surgical resections, with fewer CTCs detected after laparoscopy. If CTCs mobilized by surgery in CRC are, indeed, a cause of metastases, then preoperative treatment could potentially modulate the metastatic risk. The number of CTCs detectable during and after surgery can be reduced by the use of preoperative chemoradiotherapy in rectal cancer132 . Kienle and colleagues132 assessed the blood and bone marrow samples from 142 patients collected before, during, and after surgery. Tumour cells were detected in 34 of 103 (33%) bone marrow and 65 of 117 (55.6%) blood samples of patients who did not receive treatment, compared with 4 of 24 (16.7%) bone marrow and 10 of 25 (40%) blood samples of patients receiving treatment. The tumour-cell detection rate was lower in the chemo- radiotherapy group compared with untreated patients, but the survival rate was significantly worse in patients with tumour cells in the bone marrow after neoadjuvant therapy compared with those without detectable bone marrow involvement132 . Preoperative radiotherapy and chemoradiotherapy both improved local disease control, but the roles of these treatments in metastasis prevention or overall-survival improvements is unclear133 . In a study of 162 patients with rectal cancer treated with preopera- tive radiotherapy, Nesteruk and colleagues134 found that thedetectionofCTCs7daysaftersurgerywasofprognos- tic value for local recurrence, whereas detection of CTCs before or 24 hours after surgery was not. In breast cancer, a bimodal pattern of distant metas- tasis after treatment has been reported in surgical ­studies involving large cohorts (ranging between 1,526–3,921 patients)135–137 , a phenomenon that could be related to events occurring at the time of surgery. Demicheli and colleagues138 analysed the results of trials in which patients with axillary node-positive breast cancer had received either adjuvant chemotherapy after breast ­ cancer surgery, or mastectomy and nodal dissection alone. These investigators proposed that the meta- static process could be driven by mastectomy, and that micrometastases could undergo sustained periods of dormancy. In NSCLC, both open surgery and video-assisted surgery139,140 have been associated with increased CTC counts in venous blood. Hashimoto and colleagues141 found that, on average, surgery caused increases of CTC numbers in the pulmonary veins in 30 patients treated with lobectomy for peripheral NSCLC. Dong and col- leagues142 studiedacohortof31patientswithNSCLCwho had blood collected from a pulmonary vein during open thoracic surgery; of these 31 patients, 15 had positive test results for CTCs. The median survival and 2‑year survival rates for patients with positive versus negative findings were11monthsand26.7%, ­respectively,versus27months and 62.5%, respectively. In organ-confined prostate cancer, Eschwege et al.143 reported that CTC numbers were increased in 12 of 14 patients who underwent surgery compared with before surgery. Other situations in which CTC counts might become elevated during or soon after surgery include breast cancer resection144 , transurethral bladder cancer resection145 and oesopha­ geal cancer resection146 . The surgical community has become increasingly aware of the potential association between surgical intervention, CTCs and metastasis risk, and is actively seeking ways to ­ investigate and reduce CTC mobilization. Surgery–metastasis interaction The mechanisms by which therapeutic interventions can promote CTC mobilization are unknown and are likely to vary between and within modalities. The worst outcomes observed in patients with mobilized CTCs might reflect the negative impact of underlying vascular invasion rather than a sudden large influx of CTCs. For example, mobilization of CTCs after rectal ultrasound, which involves external pressure on the tumour, could reflect an early release of CTCs already primed within blood vessels. Direct extension of macroscopic tumours into large veins is an extreme example of such vascular invasion (FIG. 3). In renal carcinomas, life-­ threatening tumour embolisation to the lung can occur, before or during surgical resection147 . In patients receiving curative-­ intent surgery, the severing of blood and lym- phatic vessels in proximity to the tumour can lead to the formation of a fluid-filled cavity containing tumour cells that might subsequently gain access to draining lym- phatic channels148 . In addition, surgery induces a local inflammatory response in the wound, characterised by the influx of immune cells and a release of cytokines that can ­ influence the risk of metastasis149 . Nature Reviews | Clinical Oncology a b SVC SVC T T A A Figure 3 | Tumour extension into the venous system. a | Coronal CT and b | 18 F-FDG-PET/CT images of a patient with direct extension of a lung cancer (T) into the superior vena cava (SVC). The tumour causes a filling defect within the SVC and, unlike a simple clot (which would be metabolically inactive), is FDG-avid. REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 39 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 10. A causal link between surgery and metastasis has not beenestablishedtodate.Nevertheless,proposedstrategies designed to minimise the entry of CTCs into the vascu- lar system at the time of surgery, include clamping the blood vessels that drain the tumour at the earliest possible time point, and isolating the tumour from the circulation before resection150 . Systemic antitumour therapies Evidence from studies of both surgery and radiotherapy indicates that disturbance of the tumour or its environ- ment by therapeutic intervention might, under some circumstances, promote metastasis. Our group has, there- fore, investigated the possibility that systemic anticancer therapies might also occasionally have this effect, despite the counterintuitive nature of such a hypothesis. A very limited number of publications have addressed this topic. Unlike surgery and radiotherapy, systemic treatments for cancer — including chemotherapy and targeted thera- pies — can have a direct effect not only on cancer cells in the primary tumour, but also on CTCs and micrometas- tases present at the time of treatment. In a growing num- ber of clinical studies, the CTC count has been used to monitor response to systemic therapy, but the potential prognostic and predictive value of CTC release early dur- ing chemotherapy or targeted therapies has not been fully explored. In many studies, the presence of CTCs before chemotherapy is associated with a worse overall survival, compared with that of patients in whom CTCs are not detectable initially151 . Smerage and colleagues151 reported that, in patients with increased CTC counts (≥5 CTCs per 7.5ml of whole blood) detected before the commence- ment of chemotherapy, the number of CTCs often failed to decrease after treatment. Paradoxically, the results of somepreclinicalandclinicalstudiesonthemobilizationof haematopoieticstemcellsbeforehigh-dosechemotherapy and autologous stem-cell transplant suggest that chemo- therapy has the potential to increase the number of CTCs. These studies are ­ summarised in the following sections. Preclinical studies A series of studies in preclinical metastasis models have indicated that disruption of angiogenesis or induction of damage to endothelial cells can enhance metastasis. Two important studies showed that inhibition of angio- genesis was able to reduce primary tumour growth152,153 ; ­ however, by inducing an hypoxic environment in the residual tumour, the therapy promoted invasion and metastasis. Treatment of RIP1‑Tag2 mice, which develop pancreatic tumours spontaneously, with a neutralising antibody targeting VEGFR‑2 led to a reduction of pri- mary tumour volume152 , but also resulted in an increase in local invasion and distant metastasis. Similar results were obtained using sunitinib, a small-molecule inhibitor of both VEGFR and PDGFR152 . Ebos et al.153 demonstrated a similar phenom­enon in transplantable models of breast cancer and melanoma using the angiogenesis inhibitors sunitinib, sorafenib, and SU10944. Antiangiogenic thera- pies for cancer have generally not proved to be as effective in improving the survival of patients with cancer as was originally hoped154 . One of the reasons for this limited effectiveness could be that the beneficial effects of target- ing the tumour vasculature are offset by unwanted effects on tumour cell hypoxia, invasion, and metastasis. Other targeted therapies can also promote metastasis in preclinical models. Treatment of mice bearing pri- mary tumours derived from injection of A-375 human melanoma cells with the BRAF inhibitor vemurafenib, resulted in the suppression of tumour growth. When a small proportion of vemurafenib-resistant A-375 cells were injected together with parental A-375 cells, however, treatment with vemurafenib resulted in the vemurafenib-­ sensitive cells secreting factors that promoted the growth and metastasis of vemurafenib-­ resistant tumours155 . The secreted factors activated AKT signalling in resistant cells, thereby driving their ­ continued growth and metastasis. Individual cytotoxic chemotherapy agents (such as cyclophosphamide) have also been shown to promote metastasis under certain conditions. High-dose cyclo- phosphamide is effective in inducing treatment responses in some cancers, although this agent can also cause immunosuppression and, thus, is used to manage graft- versus-hostdisease156 .Conversely,lowdosesofcyclophos- phamide can enhance antitumour immune responses in animal models157 . Man and colleagues158 showed that, in addition to causing primary tumour shrinkage, both low-dose and high-dose cyclophosphamide promoted metastasis of subcutaneously implanted lung adeno­ carci­nomas to the lung. Other studies have demonstrated that an injection of cyclophosphamide 24h before intra­ venous inoculation of fibrosarcoma-derived cells mark- edly enhanced tumour-cell colonization of the lung159 . Another example is the heat shock protein 90 inhibitor 17‑AAG, which inhibited the growth of primary breast tumours, but also promoted metastasis to the bone by enhancing osteoclast formation160 . Clinical studies with chemotherapy In patients with detectable CTCs, a reduction of CTC numberswithchemotherapyisassociatedwithfavourable responses to treatment and improved survival compared withpatientswithoutadeclineinCTCs142,152,153 .Thechange in CTC numbers might even constitute a surro­ gate end pointforchemotherapyresponsiveness151,161,162 .Inpatients which CTC numbers increase steadily after chemother- apy, the increase is probably caused by the release of CTCs from progressive disease. Direct evidence for mobiliza- tion of CTCs by chemotherapy in humans is scarce, but provocative findings have been reported for high-dose chemotherapy and autologous stem-cell transplantation for the treatment of advanced-stage or high-risk solid tumours. For instance, Brugger and colleagues163 ­studied the outcomes of 46 patients with NSCLC, SCLC, or breast cancer who underwent peripheral blood stem-cell har- vesting and CTC analysis after treatment with the cyto- toxic chemotherapy drugs VP‑16, ifosfamide, cisplatin and G‑CSF (administered to mobilize blood progeni­ tor cells for subsequent harvesting). Of 42 patients with no detectable CTCs at baseline, 13 patients had detectable CTCs after chemotherapy. In the remaining four patients (with detectable baseline CTCs), CTC numbers increased after chemotherapy164 . REVIEWS 40 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 11. Mobilization of CTCs by chemotherapy might be irrelevant in patients who already have metastatic disease; however, when chemotherapy is given as neoadjuvant therapy (before planned curative surgery or radiother- apy), with the intention of treating both the local disease andoccultmetastasis,aparadoxicalmetastasis-promoting effect of CTC mobilization could be clinically important. Inhestern and colleagues164 studied CTCs in patients with advanced-stage oral and oropharyngeal squamous-cell carcinoma treated with neoadjuvant chemotherapy, surgery, and postoperative radiotherapy. These investi­ gators found that CTCs were detectable in about 80% of patients at baseline and that CTC numbers declined during chemotherapy in most patients. After surgery and before the planned adjuvant radiotherapy, however, CTC numbers had increased to above baselinelevels.Thecause of this phenomenon is unknown but in these patients, who had potentially curable disease, chemotherapy was unable to eradicate CTCs — CTC mobilization should be considered as a possible cause for this observation. Clinical studies: biological therapies In the study by Brugger et al.163 , the relative contributions of chemotherapy and G‑CSF to CTC mobilization could not be separated. In another study165 , cells identified as epithelial because they expressed epithelial cell-adhesion molecule(EpCAM),werefoundtocontaminateharvested haematopoietic progenitor cells mobilized by G‑CSF alone in patients with metastatic breast cancer before high-dose chemotherapy and autologous stem-cell trans- plantation. Thus, G‑CSF alone might also mobilize CTCs from the bone marrow or other metastatic sites. High- dose chemotherapy and stem-cell transplantation are no longer part of the standard therapy used for patients with epithelial cancers because of the negative results of randomised clinical trials166 ; thus, the contribution of mobilized tumour cells to disease relapse after ­autologous stem-cell ­ transplantation has not been established. G‑CSF-based therapy is commonly administered to patients to treat or prevent neutropenia caused by myelosuppressive chemotherapy. A meta-analysis of 17 randomized controlled trials of patients receiving chemo- therapy concluded that the mortality rate associated with infection (P=0.018) and febrile neutropenia (P<0.001) was significantly reduced by the administration of G‑CSF; however, insufficient data were available to reach any conclusions about the effect of G‑CSF on overall sur- vival167 . In preclinical studies, we and others have shown that the administration of G‑CSF promotes metastasis of mammary tumours168,169 and cell migration induced by low-dose radiation, for example, is dependent on G‑CSF signalling in NSCLC cells62 . As well as acting on tumour cells that express the G‑CSF receptor, G‑CSF can mobilize myeloid cells that have potent immunosuppressive activ- ity against T cells, thereby enhancing metastasis168 . The implications of these findings in the treatment of patients with cancer are unclear. Erythropoietin is another haematopoietic growth factor that has been used to treat patients with cancer. Erythropoietin receptors can be found on the surface of different cancer cell lines, leading to concerns about a potential role in tumour growth stimulation170 . In a randomised trial comparing erythropoietin plus radio­ therapy with radiotherapy alone in patients with head and neck cancer171 , the 5‑year estimate of locoregional failure was 46.2% versus 39.4% (P=0.42); therefore, a detri­ mental effect of erythropoietin was not ruled out by the study investigators. The publication of reports on the use of molecularly targeted agents in the treatment of patients with cancer has rapidly grown in the past few years. The mechanisms of action of such agents are completely different from those of conventional cytotoxic agents. No good clinical evidence exists on the potential role of these agents in CTC dissemination, but this possibility should be con- sidered when evaluating long-term results of treatments with molecularly targeted agents. Systemic therapy–metastasis interactions The possibility of a metastasis-promoting effect from systemic therapy in patients with cancer is a challen­ ging research question, especially because patients often derive a benefit from such therapies. Nevertheless, seek- ing evidence for such an effect is justifiable to improve the overall benefits of antitumour therapies. A potential cancer-­ promoting effect derived from systemic cytotoxic anticancer therapies seems paradoxical because of the therapeutic nature of such therapies; if this effect does occur,theimmediateclinicalcoursewouldbedetermined by the pre-existing macroscopic tumour burden. CTC mobilization by chemotherapy could poten- tially be important when neoadjuvant chemotherapy is used to treat locoregionally confined advanced-stage tumours before administering curative-intent surgery or radiotherapy and/or chemoradiotherapy. In patients with macroscopic stage IIIA NSCLC, chemotherapy followed bysurgerysubstantiallyimprovedsurvivalcomparedwith surgery alone172 . By contrast, only a small improvement in survival has been observed when surgery is followed by adjuvant chemotherapy173 . In breast cancer, however, the benefits of neoadjuvant and adjuvant chemotherapy are similar174 . The interactions between therapeutic modal- ities are complex, and likely to vary substantially across cancer types. The superior outcomes reported across a range of epi- thelial cancers for concurrent versus sequential chemo- radiotherapy are usually attributed solely to an effect on local disease control, but accurate information on a direct link between local disease control and systemic disease can be difficult to obtain from data generated in clinical trials. Most frequently, only the site of first failure (local or systemic) is reported, and subsequent local or distant failures are not described. Nevertheless, distant and local treatment failure have been recorded separately and analysed according to FDG-PET response in a study involving patients with locoregionally advanced-stage NSCLC treated with concurrent chemoradiotherapy175 . A powerful negative correlation between metastasis and a favourable local treatment response emerged from this study, indicating that chemoradiotherapy-responsive tumour cells were less likely to metastasise during or after treatment175 . Concurrent chemoradiotherapy could REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 41 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 12. reduce the risk of late distant relapse both by decreasing the probability of spread from uncontrolled local disease, and by ensuring that CTCs mobilized during treatment have sustained more damage — and, therefore, would be less capable of forming distant metastasis than CTCs ­ mobilized during radiotherapy as a single modality. Conclusions The relationships between frontline treatments, local tumour control, and metastasis in patients with solid tumours are complex and remain poorly understood. Broad generalisations cannot be made from the studies addressing these relationships because of the diversity of cancers and antitumour therapies involved, as well as variations in treatment delivery, CTC detection methods, and time points analysed, together with the long time period spanned by the publication of relevant observa- tions. A common finding in these studies, however, is that widely used antitumour therapies might be associ- ated with an early increase in either the number or the prolifera­ tive capacity of CTCs. In studies analysing both changes in CTC numbers and survival, an increase in CTCs was in general associated with a less-beneficial out- come. None of these studies has established a direct link between metastasis and CTC mobilization by treatment, but they do indicate an association between increased CTC numbers after the initiation of therapy and an ­ elevated risk of metastasis. The results of these studies do not imply that the cur- rent development of antitumour treatment methods, often the hard-won results of painstaking sequential ran- domized trials, should be abandoned because they might promote CTC mobilization. These treatments enable many patients with locoregionally confined advanced- stage solid tumours, who would otherwise suffer and die from progressive disease, to be cured or have long- term disease-free survival. The best available treatment cannot be administered without a diagnosis, but, if CTC mobilization by invasive medical interventions or ther- apy represents a major cause of antitumour treatment failure, then this subject is worthy of intensive study. In the meantime, using the least invasive means of effective diagnosis and treatment in patients with cancer seems the most prudent option. 1. Pantel, K. et al. Circulating epithelial cells in patients with benign colon diseases. Clin. Chem. 58, 936–940 (2012). 2. Sanger, N. et al. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. Int. J. Cancer 129, 2522–2526 (2011). 3. Husemann, Y. et al. Systemic spread is an early step in breast cancer. Cancer Cell 13, 58–68 (2008). 4. Weigelt, B. et al. Gene expression profiles of primary breast tumors maintained in distant metastases. Proc. Natl Acad. Sci. USA 100, 15901–15905 (2003). 5. Allard, W. J. et al. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 10, 6897–6904 (2004). 6. Lucci, A. et al. Circulating tumour cells in non- metastatic breast cancer: a prospective study. Lancet Oncol. 13, 688–695 (2012). 7. Fidler, I. J. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003). 8. Luzzi, K. J. et al. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 153, 865–873 (1998). 9. Coumans, F. A., Siesling, S. & Terstappen, L. W. Detection of cancer before distant metastasis. BMC Cancer 13, 283 (2013). 10. Klein, C. A. Parallel progression of primary tumours and metastases. Nat. Rev. Cancer 9, 302–312 (2009). 11. Naxerova, K. & Jain, R. K. Using tumour phylogenetics to identify the roots of metastasis in humans. Nat. Rev. Clin. Oncol. 12, 258–272 (2015). 12. Sosa, M. S., Bragado, P. & Aguirre-Ghiso, J. A. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat. Rev. Cancer 14, 611–622 (2014). 13. Comen, E., Norton, L. & Massague, J. Clinical implications of cancer self-seeding. Nat. Rev. Clin. Oncol. 8, 369–377 (2011). 14. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015). 15. Toss, A., Mu, Z., Fernandez, S. & Cristofanilli, M. CTC enumeration and characterization: moving toward personalized medicine. Ann. Transl Med. 2, 108 (2014). 16. Schwarzenbach, H. et al. Loss of heterozygosity at tumor suppressor genes detectable on fractionated circulating cell-free tumor DNA as indicator of breast cancer progression. Clin. Cancer Res. 18, 5719–5730 (2012). 17. Spindler, K. L., Pallisgaard, N., Andersen, R. F. & Jakobsen, A. Changes in mutational status during third- line treatment for metastatic colorectal cancer—results of consecutive measurement of cell free DNA, KRAS and BRAF in the plasma. Int. J. Cancer 135, 2215–2222 (2014). 18. Murtaza, M. et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013). 19. Bonnomet, A. et al. Epithelial-to‑mesenchymal transitions and circulating tumor cells. J. Mammary Gland Biol. Neoplasia 15, 261–273 (2010). 20. Yu, M. et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 339, 580–584 (2013). 21. Lafrenie, R., Shaughnessy, S. G. & Orr, F. W. Cancer cell interactions with injured or activated endothelium. Cancer Metastasis Rev. 11, 377–388 (1992). 22. Friedl, P., Locker, J., Sahai, E. & Segall, J. E. Classifying collective cancer cell invasion. Nat. Cell Biol. 14, 777–783 (2012). 23. Rofstad, E. K., Galappathi, K. & Mathiesen, B. S. Tumor interstitial fluid pressure‑a link between tumor hypoxia, microvascular density, and lymph node metastasis. Neoplasia 16, 586–594 (2014). 24. Robinson, B. D. et al. Tumor microenvironment of metastasis in human breast carcinoma: a potential prognostic marker linked to hematogenous dissemination. Clin. Cancer Res. 15, 2433–2441 (2009). 25. Roussos, E. T. et al. Mena invasive (MenaINV) promotes multicellular streaming motility and transendothelial migration in a mouse model of breast cancer. J. Cell Sci. 124, 2120–2131 (2011). 26. Armstrong, A. J. et al. Circulating tumor cells from patients with advanced prostate and breast cancer display both epithelial and mesenchymal markers. Mol. Cancer Res. 9, 997–1007 (2011). 27. Hou, J. M. et al. Circulating tumor cells as a window on metastasis biology in lung cancer. Am. J. Pathol. 178, 989–996 (2011). 28. Brandt, B. et al. Isolation of prostate-derived single cells and cell clusters from human peripheral blood. Cancer Res. 56, 4556–4561 (1996). 29. Hou, J. M. et al. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J. Clin. Oncol. 30, 525–532 (2012). 30. Aceto, N. et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 158, 1110–1122 (2014). 31. Martin, O. A. et al. Mobilization of viable tumor cells into the circulation during radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 88, 395–403 (2014). 32. Pretlow, T. G. et al. Prostate cancer and other xenografts from cells in peripheral blood of patients. Cancer Res. 60, 4033–4036 (2000). 33. Baccelli, I. et al. Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat. Biotechnol. 31, 539–544 (2013). 34. Hodgkinson, C. L. et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 20, 897–903 (2014). 35. Yu, M. et al. Cancer therapy. Ex vivo culture of circulating breast tumor cells for individualized testing of drug susceptibility. Science 345, 216–220 (2014). 36. Gao, D. et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 159, 176–187 (2014). 37. Levina, V., Marrangoni, A. M., DeMarco, R., Gorelik, E. & Lokshin, A. E. Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS ONE 3, e3077 (2008). 38. Gomez-Casal, R. et al. Non-small cell lung cancer cells survived ionizing radiation treatment display cancer stem cell and epithelial-mesenchymal transition phenotypes. Mol. Cancer 12, 94 (2013). 39. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1, 571–573 (1889). 40. Kaplan, R. N. et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438, 820–827 (2005). 41. Hiratsuka, S. et al. The S100A8‑serum amyloid A3‑TLR4 paracrine cascade establishes a pre-metastatic phase. Nat. Cell Biol. 10, 1349–1355 (2008). 42. Hood, J. L., San, R. S. & Wickline, S. A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71, 3792–3801 (2011). 43. Fong, M. Y. et al. Breast-cancer-secreted miR‑122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 17, 183–194 (2015). 44. von Essen, C. F. Radiation enhancement of metastasis: a review. Clin. Exp. Metastasis 9, 77–104 (1991). 45. Kaplan, H. S. & Murphy, E. D. The effect of local roentgen irradiation on the biological behavior of a transplantable mouse carcinoma; increased frequency of pulmonary metastasis. J. Natl Cancer Inst. 9, 407–413 (1949). 46. Sheldon, P. W. & Fowler, J. F. The effect of low-dose pre-operative X‑irradiation of implanted mouse mammary carcinomas on local recurrence and metastasis. Br. J. Cancer 34, 401–407 (1976). 47. Camphausen, K. et al. Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res. 61, 2207–2211 (2001). REVIEWS 42 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 13. 48. Rofstad, E. K., Mathiesen, B. & Galappathi, K. Increased metastatic dissemination in human melanoma xenografts after subcurative radiation treatment: radiation-induced increase in fraction of hypoxic cells and hypoxia-induced up‑regulation of urokinase-type plasminogen activator receptor. Cancer Res. 64, 13–18 (2004). 49. Park, J. K. et al. Establishment of animal model for the analysis of cancer cell metastasis during radiotherapy. Radiat. Oncol. 7, 153 (2012). 50. Li, T. et al. Radiation enhances long-term metastasis potential of residual hepatocellular carcinoma in nude mice through TMPRSS4-induced epithelial– mesenchymal transition. Cancer Gene Ther. 18, 617–626 (2011). 51. Woods, G. M. et al. Radiation therapy may increase metastatic potential in alveolar rhabdomyosarcoma. Pediatr. Blood Cancer 62, 1550–1554 (2015). 52. Bernier, J., Hall, E. J. & Giaccia, A. Radiation oncology: a century of achievements. Nat. Rev. Cancer 4, 737–747 (2004). 53. Eriksson, D. & Stigbrand, T. Radiation-induced cell death mechanisms. Tumour Biol. 31, 363–372 (2010). 54. Dewey, W. C., Ling, C. C. & Meyn, R. E. Radiation- induced apoptosis: relevance to radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 33, 781–796 (1995). 55. Palumbo, S. & Comincini, S. Autophagy and ionizing radiation in tumors: the “survive or not survive” dilemma. J. Cell. Physiol. 228, 1–8 (2013). 56. Brunner, T. B., Kunz-Schughart, L. A., Grosse- Gehling, P. & Baumann, M. Cancer stem cells as a predictive factor in radiotherapy. Semin. Radiat. Oncol. 22, 151–174 (2012). 57. Butof, R., Dubrovska, A. & Baumann, M. Clinical perspectives of cancer stem cell research in radiation oncology. Radiother. Oncol. 108, 388–396 (2013). 58. Jackson, S. P. Sensing and repairing DNA double- strand breaks. Carcinogenesis 23, 687–696 (2002). 59. Roy, K., Kodama, S., Suzuki, K. & Watanabe, M. Delayed cell death, giant cell formation and chromosome instability induced by X‑irradiation in human embryo cells. J. Radiat. Res. 40, 311–322 (1999). 60. De Bacco, F. et al. Induction of MET by ionizing radiation and its role in radioresistance and invasive growth of cancer. J. Natl Cancer Inst. 103, 645–661 (2011). 61. Zheng, Q. et al. X‑ray radiation promotes the metastatic potential of tongue squamous cell carcinoma cells via modulation of biomechanical and cytoskeletal properties. Hum. Exp. Toxicol. 34, 894–903 (2015). 62. Cui, Y. H. et al. Radiation promotes invasiveness of non-small-cell lung cancer cells through granulocyte- colony-stimulating factor. Oncogene 34, 5372–5382 (2015). 63. Vilalta, M., Rafat, M., Giaccia, A. J. & Graves, E. E. Recruitment of circulating breast cancer cells is stimulated by radiotherapy. Cell Rep. 8, 402–409 (2014). 64. Gorski, D. H. et al. Blockage of the vascular endothelial growth factor stress response increases the antitumor effects of ionizing radiation. Cancer Res. 59, 3374–3378 (1999). 65. Moeller, B. J., Cao, Y., Li, C. Y. & Dewhirst, M. W. Radiation activates HIF‑1 to regulate vascular radiosensitivity in tumors: role of reoxygenation, free radicals, and stress granules. Cancer Cell 5, 429–441 (2004). 66. Chung, Y. L. et al. Sublethal irradiation induces vascular endothelial growth factor and promotes growth of hepatoma cells: implications for radiotherapy of hepatocellular carcinoma. Clin. Cancer Res. 12, 2706–2715 (2006). 67. Sofia Vala, I. et al. Low doses of ionizing radiation promote tumor growth and metastasis by enhancing angiogenesis. PLoS ONE 5, e11222 (2010). 68. Shen, C. J. et al. Ionizing radiation induces tumor cell lysyl oxidase secretion. BMC Cancer 14, 532 (2014). 69. Budach, W., Kammers, K., Boelke, E. & Matuschek, C. Adjuvant radiotherapy of regional lymph nodes in breast cancer - a meta-analysis of randomized trials. Radiat. Oncol. 8, 267 (2013). 70. Errico, A. Radiotherapy: a double-edged sword for NSCLC? Nat. Rev. Clin. Oncol. 11, 66 (2014). 71. Bonner, W. M. et al. γH2AX and cancer. Nat. Rev. Cancer 8, 957–967 (2008). 72. Ivashkevich, A., Redon, C. E., Nakamura, A. J., Martin, R. F. & Martin, O. A. Use of the γ‑H2AX assay to monitor DNA damage and repair in translational cancer research. Cancer Lett. 327, 123–133 (2012). 73. Saunders, M. et al. Continuous hyperfractionated accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small-cell lung cancer: a randomised multicentre trial. CHART Steering Committee. Lancet 350, 161–165 (1997). 74. Saunders, M. et al. Continuous, hyperfractionated, accelerated radiotherapy (CHART) versus conventional radiotherapy in non-small cell lung cancer: mature data from the randomised multicentre trial. CHART Steering committee. Radiother. Oncol. 52, 137–148 (1999). 75. Dorsey, J. F. et al. Tracking viable circulating tumor cells (CTCs) in the peripheral blood of non-small cell lung cancer (NSCLC) patients undergoing definitive radiation therapy: pilot study results. Cancer 121, 139–149 (2015). 76. Lowes, L. E. et al. Circulating tumour cells in prostate cancer patients receiving salvage radiotherapy. Clin. Transl Oncol. 14, 150–156 (2012). 77. Haikerwal, S. J., Hagekyriakou, J., MacManus, M., Martin, O. A. & Haynes, N. M. Building immunity to cancer with radiation therapy. Cancer Lett. 368, 198–208 (2015). 78. Rivinius, R. et al. Analysis of malignancies in patients after heart transplantation with subsequent immunosuppressive therapy. Drug Des. Devel. Ther. 9, 93–102 (2015). 79. Engels, E. A. et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 306, 1891–1901 (2011). 80. Andarawewa, K. L. et al. Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor β-induced epithelial to mesenchymal transition. Cancer Res. 67, 8662–8670 (2007). 81. Yuan, W., Yuan, Y., Zhang, T. & Wu, S. Role of Bmi‑1 in regulation of ionizing irradiation-induced epithelial- mesenchymal transition and migration of breast cancer cells. PLoS ONE 10, e0118799 (2015). 82. Kim, R. K. et al. Radiation promotes malignant phenotypes through SRC in breast cancer cells. Cancer Sci. 106, 78–85 (2015). 83. Qian, L. W. et al. Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin. Cancer Res. 8, 1223–1227 (2002). 84. Wasserman, J., Blomgren, H., Rotstein, S., Petrini, B. & Hammarstrom, S. Immunosuppression in irradiated breast cancer patients: in vitro effect of cyclooxygenase inhibitors. Bull. NY Acad. Med. 65, 36–44 (1989). 85. Reits, E. A. et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 203, 1259–1271 (2006). 86. Zhang, B. et al. Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J. Exp. Med. 204, 49–55 (2007). 87. Chakraborty, M. et al. Irradiation of tumor cells up‑regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J. Immunol. 170, 6338–6347 (2003). 88. Hall, E. J. Radiobiology for the Radiologist (Medical Department, Harper and Row, 1973). 89. Pateras, I. S. et al. The DNA damage response and immune signaling alliance: is it good or bad? Nature decides when and where. Pharmacol. Ther. 154, 36–56 (2015). 90. Scheithauer, H., Belka, C., Lauber, K. & Gaipl, U. S. Immunological aspects of radiotherapy. Radiat. Oncol. 9, 185 (2014). 91. Brown, J. M. The hypoxic cell: a target for selective cancer therapy—eighteenth Bruce, F. Cain Memorial Award lecture. Cancer Res. 59, 5863–5870 (1999). 92. Shultz, D. B., Diehn, M. & Loo, B. W. Jr. To SABR or not to SABR? Indications and contraindications for stereotactic ablative radiotherapy in the treatment of early-stage, oligometastatic, or oligoprogressive non- small cell lung cancer. Semin. Radiat. Oncol. 25, 78–86 (2015). 93. Bertolaccini, L., Terzi, A., Ricchetti, F. & Alongi, F. Surgery or stereotactic ablative radiation therapy: how will be treated operable patients with early stage not small cell lung cancer in the next future? Ann. Transl Med. 3, 25 (2015). 94. Cao, C. et al. Surgery versus SABR for resectable non- small-cell lung cancer. Lancet Oncol. 16, e370–e371 (2015). 95. Zheng, X. et al. Survival outcome after stereotactic body radiation therapy and surgery for stage I non-small cell lung cancer: a meta-analysis. Int. J. Radiat. Oncol. Biol. Phys. 90, 603–611 (2014). 96. Rekers, N. H. et al. Stereotactic ablative body radiotherapy combined with immunotherapy: present status and future perspectives. Cancer Radiother. 18, 391–395 (2014). 97. Narayan, K., Fisher, R. J., Bernshaw, D., Shakher, R. & Hicks, R. J. Patterns of failure and prognostic factor analyses in locally advanced cervical cancer patients staged by positron emission tomography and treated with curative intent. Int. J. Gynecol. Cancer 19, 912–918 (2009). 98. Narayan, K., Khaw, P., Bernshaw, D., Mileshkin, L. & Kondalsamy-Chennakesavan, S. Prognostic significance of lymphovascular space invasion and nodal involvement in intermediate- and high-risk endometrial cancer patients treated with curative intent using surgery and adjuvant radiotherapy. Int. J. Gynecol. Cancer 22, 260–266 (2012). 99. Fidler, I. J. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur. J. Cancer 9, 223–227 (1973). 100. Liotta, L. A., Saidel, M. G. & Kleinerman, J. The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res. 36, 889–894 (1976). 101. Ceelen, W., Pattyn, P. & Mareel, M. Surgery, wound healing, and metastasis: recent insights and clinical implications. Crit. Rev. Oncol. Hematol. 89, 16–26 (2014). 102. Alagaratnam, T. T. & Ong, G. B. Wound implantation — a surgical hazard. Br. J. Surg. 64, 872–875 (1977). 103. Heaney, A. & Buggy, D. J. Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br. J. Anaesthesia 109 (Suppl. 1), i17–i28 (2012). 104. Ben-Eliyahu, S. The promotion of tumor metastasis by surgery and stress: immunological basis and implications for psychoneuroimmunology. Brain Behav. Immun. 17 (Suppl. 1), 27–36 (2003). 105. Buckley, A., McQuaid, S., Johnson, P. & Buggy, D. J. Effect of anaesthetic technique on the natural killer cell anti-tumour activity of serum from women undergoing breast cancer surgery: a pilot study. Br. J. Anaesthesia 113 (Suppl. 1), i56–62 (2014). 106. Divatia, J. V. & Ambulkar, R. Anesthesia and cancer recurrence: what is the evidence? J. Anaesthesiol. Clin. Pharmacol. 30, 147–150 (2014). 107. Cata, J. P., Wang, H., Gottumukkala, V., Reuben, J. & Sessler, D. I. Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions. Br. J. Anaesthesia 110, 690–701 (2013). 108. Demicheli, R., Retsky, M. W., Hrushesky, W. J., Baum, M. & Gukas, I. D. The effects of surgery on tumor growth: a century of investigations. Ann. Oncol. 19, 1821–1828 (2008). 109. Horowitz, M., Neeman, E., Sharon, E. & Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 12, 213–226 (2015). 110. Holmgren, L., O’Reilly, M. S. & Folkman, J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat. Med. 1, 149–153 (1995). 111. Abramovitch, R., Marikovsky, M., Meir, G. & Neeman, M. Stimulation of tumour growth by wound- derived growth factors. Br. J. Cancer 79, 1392–1398 (1999). 112. Mathenge, E. G. et al. Core needle biopsy of breast cancer tumors increases distant metastases in a mouse model. Neoplasia 16, 950–960 (2014). 113. Moreno, J. G. et al. Transrectal ultrasound-guided biopsy causes hematogenous dissemination of prostate cells as determined by RT‑PCR. Urology 49, 515–520 (1997). 114. Polascik, T. J. et al. Influence of sextant prostate needle biopsy or surgery on the detection and harvest of intact circulating prostate cancer cells. J. Urol. 162, 749–752 (1999). 115. Dyavanagoudar, S., Kale, A., Bhat, K. & Hallikerimath, S. Reverse transcriptase polymerase chain reaction study to evaluate dissemination of cancer cells into circulation after incision biopsy in oral squamous cell carcinoma. Indian J. Dental Res. 19, 315–319 (2008). 116. Kusukawa, J. et al. Dissemination of cancer cells into circulation occurs by incisional biopsy of oral squamous cell carcinoma. J. Oral Pathol. Med. 29, 303–307 (2000). REVIEWS NATURE REVIEWS | CLINICAL ONCOLOGY VOLUME 14 | JANUARY 2017 | 43 © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d .
  • 14. 117. Zoubek, A. et al. Mobilization of tumour cells during biopsy in an infant with Ewing sarcoma. Eur. J. Pediatr. 155, 373–376 (1996). 118. Uchida, N., Suda, T., Inoue, T., Fujiwara, Y. & Ishiguro, K. Needle track dissemination of follicular thyroid carcinoma following fine-needle aspiration biopsy: report of a case. Surg. Today 37, 34–37 (2007). 119. Jones, O. M., Rees, M., John, T. G., Bygrave, S. & Plant, G. Biopsy of resectable colorectal liver metastases causes tumour dissemination and adversely affects survival after liver resection. Br. J. Surg. 92, 1165–1168 (2005). 120. Rodgers, M. S., Collinson, R., Desai, S., Stubbs, R. S. & McCall, J. L. Risk of dissemination with biopsy of colorectal liver metastases. Dis. Colon Rectum 46, 454–458; discussion 458–459 (2003). 121. Myrvang, H. Diagnosis: novel anti-seeding technology to prevent dissemination of tumor cells after a fine- needle aspiration biopsy. Nat. Rev. Clin. Oncol. 8, 64 (2011). 122. Wiksell, H. et al. Prevention of tumour cell dissemination in diagnostic needle procedures. Br. J. Cancer 103, 1706–1709 (2010). 123. Koch, M. et al. Hematogenous tumor cell dissemination during colonoscopy for colorectal cancer. Surg. Endosc. 18, 587–591 (2004). 124. Koch, M. et al. Increased detection rate and potential prognostic impact of disseminated tumor cells in patients undergoing endorectal ultrasound for rectal cancer. Int. J. Colorectal Dis. 22, 359–365 (2007). 125. Peach, G., Kim, C., Zacharakis, E., Purkayastha, S. & Ziprin, P. Prognostic significance of circulating tumour cells following surgical resection of colorectal cancers: a systematic review. Br. J. Cancer 102, 1327–1334 (2010). 126. Rahbari, N. N. et al. Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterology 138, 1714–1726 (2010). 127. Steinert, G., Scholch, S., Koch, M. & Weitz, J. Biology and significance of circulating and disseminated tumour cells in colorectal cancer. Langenbecks Arch. Surg. 397, 535–542 (2012). 128. Weitz, J. et al. Dissemination of tumor cells in patients undergoing surgery for colorectal cancer. Clin. Cancer Res. 4, 343–348 (1998). 129. Rahbari, N. N. et al. Compartmental differences of circulating tumor cells in colorectal cancer. Ann. Surg. Oncol. 19, 2195–2202 (2012). 130. Koch, M. et al. Detection of disseminated tumor cells in liver biopsies of colorectal cancer patients is not associated with a worse prognosis. Ann. Surg. Oncol. 14, 810–817 (2007). 131. Wind, J. et al. Circulating tumour cells during laparoscopic and open surgery for primary colonic cancer in portal and peripheral blood. Eur. J. Surg. Oncol. 35, 942–950 (2009). 132. Kienle, P. et al. Decreased detection rate of disseminated tumor cells of rectal cancer patients after preoperative chemoradiation: a first step towards a molecular surrogate marker for neoadjuvant treatment in colorectal cancer. Ann. Surg. 238, 324–330; discussion 330–321 (2003). 133. Rahbari, N. N. et al. Neoadjuvant radiotherapy for rectal cancer: meta-analysis of randomized controlled trials. Ann. Surg. Oncol. 20, 4169–4182 (2013). 134. Nesteruk, D. et al. Evaluation of prognostic significance of circulating tumor cells detection in rectal cancer patients treated with preoperative radiotherapy: prospectively collected material data. BioMed Res. Int. 2014, 712827 (2014). 135. Demicheli, R., Biganzoli, E., Boracchi, P., Greco, M. & Retsky, M. W. Recurrence dynamics does not depend on the recurrence site. Breast Cancer Res. 10, R83 (2008). 136. Fortin, A., Larochelle, M., Laverdiere, J., Lavertu, S. & Tremblay, D. Local failure is responsible for the decrease in survival for patients with breast cancer treated with conservative surgery and postoperative radiotherapy. J. Clin. Oncol. 17, 101–109 (1999). 137. Jatoi, I., Tsimelzon, A., Weiss, H., Clark, G. M. & Hilsenbeck, S. G. Hazard rates of recurrence following diagnosis of primary breast cancer. Breast Cancer Res. Treat. 89, 173–178 (2005). 138. Demicheli, R. et al. Breast cancer recurrence dynamics following adjuvant CMF is consistent with tumor dormancy and mastectomy-driven acceleration of the metastatic process. Ann. Oncol. 16, 1449–1457 (2005). 139. Hermansson, U., Konstantinov, I. E. & Aren, C. Tumor dissemination after video-assisted thoracic surgery: what does it mean? J. Thorac. Cardiovasc. Surg. 114, 300–302 (1997). 140. Yamashita, J. I., Kurusu, Y., Fujino, N., Saisyoji, T. & Ogawa, M. Detection of circulating tumor cells in patients with non-small cell lung cancer undergoing lobectomy by video-assisted thoracic surgery: a potential hazard for intraoperative hematogenous tumor cell dissemination. J. Thorac. Cardiovasc. Surg. 119, 899–905 (2000). 141. Hashimoto, M. et al. Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact. Cardiovasc. Thorac. Surg. 18, 775–783 (2014). 142. Dong, Q. et al. Hematogenous dissemination of lung cancer cells during surgery: quantitative detection by flow cytometry and prognostic significance. Lung Cancer 37, 293–301 (2002). 143. Eschwege, P. et al. Haematogenous dissemination of prostatic epithelial cells during radical prostatectomy. Lancet 346, 1528–1530 (1995). 144. Daskalakis, M. et al. Assessment of the effect of surgery on the kinetics of circulating tumour cells in patients with operable breast cancer based on cytokeratin‑19 mRNA detection. Eur. J. Surg. Oncol. 37, 404–410 (2011). 145. Engilbertsson, H. et al. Transurethral bladder tumor resection can cause seeding of cancer cells into the bloodstream. J. Urol. 193, 53–57 (2015). 146. Nakashima, S. et al. Clinical significance of circulating tumor cells in blood by molecular detection and tumor markers in esophageal cancer. Surgery 133, 162–169 (2003). 147. Kayalar, N. et al. Concomitant surgery for renal neoplasm with pulmonary tumor embolism. J. Thorac. Cardiovasc. Surg. 139, 320–325 (2010). 148. Podgrabinska, S. & Skobe, M. Role of lymphatic vasculature in regional and distant metastases. Microvasc. Res. 95, 46–52 (2014). 149. Liao, C. et al. Prognostic value of circulating inflammatory factors in non-small cell lung cancer: a systematic review and meta-analysis. Cancer Biomarkers 14, 469–481 (2014). 150. Song, P. P., Zhang, W., Zhang, B., Liu, Q. & Du, J. Effects of different sequences of pulmonary artery and vein ligations during pulmonary lobectomy on blood micrometastasis of non-small cell lung cancer. Oncol. Lett. 5, 463–468 (2013). 151. Smerage, J. B. et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J. Clin. Oncol. 32, 3483–3489 (2014). 152. Paez-Ribes, M. et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 15, 220–231 (2009). 153. Ebos, J. M. et al. Accelerated metastasis after short- term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239 (2009). 154. Ebos, J. M. & Kerbel, R. S. Antiangiogenic therapy: impact on invasion, disease progression, and metastasis. Nat. Rev. Clin. Oncol. 8, 210–221 (2011). 155. Obenauf, A. C. et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature 520, 368–372 (2015). 156. Itescu, S. et al. Intravenous pulse administration of cyclophosphamide is an effective and safe treatment for sensitized cardiac allograft recipients. Circulation 105, 1214–1219 (2002). 157. Schiavoni, G. et al. Cyclophosphamide induces type I interferon and augments the number of CD44hi T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer. Blood 95, 2024–2030 (2000). 158. Man, S., Zhang, Y., Gao, W., Yan, L. & Ma, C. Cyclophosphamide promotes pulmonary metastasis on mouse lung adenocarcinoma. Clin. Exp. Metastasis 25, 855–864 (2008). 159. Yamauchi, K. et al. Induction of cancer metastasis by cyclophosphamide pretreatment of host mice: an opposite effect of chemotherapy. Cancer Res. 68, 516–520 (2008). 160. Price, J. T. et al. The heat shock protein 90 inhibitor, 17‑allylamino-17‑demethoxygeldanamycin, enhances osteoclast formation and potentiates bone metastasis of a human breast cancer cell line. Cancer Res. 65, 4929–4938 (2005). 161. Cristofanilli, M. et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 351, 781–791 (2004). 162. Cohen, S. J. et al. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J. Clin. Oncol. 26, 3213–3221 (2008). 163. Brugger, W. et al. Mobilization of peripheral blood progenitor cells by sequential administration of interleukin‑3 and granulocyte-macrophage colony- stimulating factor following polychemotherapy with etoposide, ifosfamide, and cisplatin. Blood 79, 1193–1200 (1992). 164. Inhestern, J. et al. Prognostic role of circulating tumor cells during induction chemotherapy followed by curative surgery combined with postoperative radiotherapy in patients with locally advanced oral and oropharyngeal squamous cell cancer. PLoS ONE 10, e0132901 (2015). 165. Mego, M. et al. Prognostic value of EMT-circulating tumor cells in metastatic breast cancer patients undergoing high-dose chemotherapy with autologous hematopoietic stem cell transplantation. J. Cancer 3, 369–380 (2012). 166. Hamilton, B. K. et al. Long-term survival after high- dose chemotherapy with autologous hematopoietic cell transplantation in metastatic breast cancer. Hematol. Oncol. Stem Cell Ther. 8, 115–124 (2015). 167. Kuderer, N. M., Dale, D. C., Crawford, J. & Lyman, G. H. Impact of primary prophylaxis with granulocyte colony-stimulating factor on febrile neutropenia and mortality in adult cancer patients receiving chemotherapy: a systematic review. J. Clin. Oncol. 25, 3158–3167 (2007). 168. Cao, Y. et al. BMP4 inhibits breast cancer metastasis by blocking myeloid-derived suppressor cell activity. Cancer Res. 74, 5091–5102 (2014). 169. Kowanetz, M. et al. Granulocyte-colony stimulating factor promotes lung metastasis through mobilization of Ly6G+Ly6C+ granulocytes. Proc. Natl Acad. Sci. USA 107, 21248–21255 (2010). 170. Debeljak, N., Solar, P. & Sytkowski, A. J. Erythropoietin and cancer: the unintended consequences of anemia correction. Front. Immunol. 5, 563 (2014). 171. Shenouda, G. et al. Long-term results of radiation therapy oncology group 9903: a randomized phase 3 trial to assess the effect of erythropoietin on local- regional control in anemic patients treated with radiation therapy for squamous cell carcinoma of the head and neck. Int. J. Radiat. Oncol. Biol. Phys. 91, 907–915 (2015). 172. NSCLC Meta-analysis Collaborative Group. Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 383, 1561–1571 (2014). 173. Liang, Y. & Wakelee, H. A. Adjuvant chemotherapy of completely resected early stage non-small cell lung cancer (NSCLC). Transl Lung Cancer Res. 2, 403–410 (2013). 174. Mauri, D., Pavlidis, N. & Ioannidis, J. P. Neoadjuvant versus adjuvant systemic treatment in breast cancer: ameta-analysis. J. Natl Cancer Inst. 97, 188–194 (2005). 175. Mac Manus, M. P. et al. Metabolic (FDG-PET) response after radical radiotherapy/ chemoradiotherapy for non-small cell lung cancer correlates with patterns of failure. Lung Cancer 49, 95–108 (2005). Acknowledgements We are grateful to David Ball for fruitful discussions and his continuous support of our scientific initiatives, and to Bernhard Riedel for his critical reading of the manuscript. We thank Tim Akhurst for his assistance with Figure 3. O.A.M. and M.P.M. receive support from the Australian National Health and Medical Research Council (NHMRC) grant 1104139, and the Peter MacCallum Cancer Foundation grant 1218. R.L.A. receives fellowship support from the National Breast Cancer Foundation of Australia. Author contributions O.A.M., R.L.A. and M.P.M. researched data for the article, and K.N. contributed to discussion of the article’s content. All authors wrote, reviewed and edited the manuscript before submission. Competing interests statement The authors declare no competing interests. REVIEWS 44 | JANUARY 2017 | VOLUME 14 www.nature.com/nrclinonc © 2 0 1 6 M a c m i l l a n P u b l i s h e r s L i m i t e d , p a r t o f S p r i n g e r N a t u r e . A l l r i g h t s r e s e r v e d . View publication stats