Cancer and Metastasis Reviews
https://doi.org/10.1007/s10555-023-10123-0
NON-THEMATIC REVIEW
Is cancer an intelligent species?
Chiara Nicolazzo1
Paola Gazzaniga1
· Federica Francescangeli2
· Valentina Magri3 · Alessandro Giuliani4
· Ann Zeuner2
·
Received: 3 April 2023 / Accepted: 26 June 2023
© The Author(s) 2023
Abstract
Some relevant emerging properties of intelligent systems are “adaptation to a changing environment,” “reaction to unexpected
situations,” “capacity of problem solving,” and “ability to communicate.” Single cells have remarkable abilities to adapt,
make adequate context-dependent decision, take constructive actions, and communicate, thus theoretically meeting all the
above-mentioned requirements. From a biological point of view, cancer can be viewed as an invasive species, composed of
cells that move from primary to distant sites, being continuously exposed to changes in the environmental conditions. Blood
represents the first hostile habitat that a cancer cell encounters once detached from the primary site, so that cancer cells must
rapidly carry out multiple adaptation strategies to survive. The aim of this review was to deepen the adaptation mechanisms
of cancer cells in the blood microenvironment, particularly referring to four adaptation strategies typical of animal species
(phenotypic adaptation, metabolic adaptation, niche adaptation, and collective adaptation), which together define the broad
concept of biological intelligence. We provided evidence that the required adaptations (either structural, metabolic, and
related to metastatic niche formation) and “social” behavior are useful principles allowing putting into a coherent frame
many features of circulating cancer cells. This interpretative frame is described by the comparison with analog behavioral
traits typical of various animal models.
Keywords Intelligence · Adaptation · Circulating tumor cells · EMT
1 About species and intelligence:
a necessary premise
Although the question of what constitutes a species has a
long, argumentative history, a universal definition of species
is far from being established. Settling whether cancer is a
Chiara Nicolazzo, Federica Francescangeli, Ann Zeuner, and Paola
Gazzaniga contributed equally.
species in its own is beyond the scope of this review; nevertheless, the title we have chosen forces us to make a little
reflection. Since various concepts of species, some mutually
exclusive, some other interchangeable, have followed one
another over time, it is particularly hard to answer whether
cancer can be considered as a species. Nevertheless, cancer
evolutionary studies have recently produced some information that can help to untie the knot. The concept of cancer
* Paola Gazzaniga
paola.gazzaniga@uniroma1.it
1
Department of Molecular Medicine, Sapienza University
of Rome, 00161 Rome, Italy
Chiara Nicolazzo
chiara.nicolazzo@uniroma1.it
2
Federica Francescangeli
federica.francescangeli@iss.it
Department of Oncology and Molecular Medicine, Istituto
Superiore di Sanità, Viale Regina Elena 299, 00161 Rome,
Italy
3
Valentina Magri
valentina.magri@uniroma1.it
Department of Pathology, Oncology and Radiology, Sapienza
University of Rome, 00161 Rome, Italy
4
Environment and Health Department, Istituto Superiore di
Sanità, Viale Regina Elena 299, 00161 Rome, Italy
Alessandro Giuliani
alessandro.giuliani@iss.it
Ann Zeuner
a.zeuner@iss.it
13
Vol.:(0123456789)
Cancer and Metastasis Reviews
as a species is not new, having its roots very far back in time
with Boveri who for the first time assumed that aneuploidy,
the starting point of malignant transformation, delineates
cancer as a species [1]. Later, Duesberg and Rasnick [2],
who supported the theory that cancer cells derive their complex phenotypes from random chromosome number mutation, a process that is analogous to speciation, suggested
that a broad definition of species could be useful to describe
some relevant features of cancer. Mark Vincent, in the same
years, excellently reexamined the concept of “cancer as
species” supported from the gradually recognized species
distinctiveness of asexual organisms, enlarging the reach of
the “cancer as species” concept [3]. To delve deeper into
this thorny issue, we recommend the book “Debating Cancer: The Paradox in Cancer Research,” where the chapter
“Do Different Cancers Represent Different Species?” excellently illustrates the evidence for and against the concept of
cancer as a species [4]. Furthermore, Heng’s postulation of
the genome architecture theory [5], which we will discuss
later, conciliates cancer with biological evolution, definitely
reserving a place for cancer as a species in itself. Following
the assumption that cancer can be considered as a species, a
second theme of this review is whether this species shows a
somewhat “intelligent” behavior. A critical premise is that
intelligence is here to be intended as a metaphor and not as
a comparison with animal behaviors that are “symptoms”
of intelligence. The metaphor of cancer as an “intelligent
species” is an attempt to provide a different point of view
on cancer adaptation through a process of simplification
(by moving from complex cellular processes to macroscopic phenomena) and consilience (stimulating a “jumping
together” of knowledge in different fields of science to converge on the same conclusion) [6]. In fact, metaphors (from
the Greek metapherein, meaning “transference”) allow moving between different levels of knowledge, contributing to
the development of meaning through boundless interpretive
trajectories [7]. Moreover, metaphor has an instructive value,
namely caused by the pleasure of the moment of understanding that follows surprise (Aristotles, Ars Rhetorica).
1.1 About intelligence and species adaptation
Almost all definitions of intelligence agree on that, far from
being a mere set of cognitive abilities (general intelligence),
intelligence involves the ability to adapt to a changing environment (adaptive intelligence) [8]. From a biological
perspective, intelligence is often referred as “adapting” to
the environment, as the result of natural selection [9]. This
adaptation (“narrow adaptation”) represents an oversimplification of biological intelligence. Cephalocarids, populating our seas since 250 million years are excellent adapters, but not “intelligent” in the sense in which we usually
refer to intelligence. Still more cogent, the configuration
13
change of hemoglobin shifting from R to T folds adapting
to changes in oxygen partial pressure tells us that such a
refined adaptive behavior cannot be considered as a sign of a
molecular “intelligence” [10]. A broader (and more relevant
for our aims) view of biological adaptation can be further
defined as follows: (1) structural adaptation, i.e., the ability
to change oneself shape to fit the environment; (2) changing metabolism to increase oneself fitness; (3) changing the
environment; and (4) involve other individuals in a collective
adaptation [11]. As already stressed in the “Abstract,” we
concentrate on animal adaptive strategies, given the wide
range of remarkable examples of the above-sketched points.
Many birds, including parrots, belong to the first category,
undergoing phenotypic shaping and increasing their beaks
and limbs size in order to adapt to climate changing [12]. In
some birds, even hybridization is an adaptive solution when
landscape changes due to human interventions [13]. Naked
mole-rats (Heterocephalus glaber) have the ability defined
at point 2, changing metabolism to survive underground in
low-oxygen environments [14]. Australian frogs (Cyclorana
australis) belong to point 3, creating a new environment rich
of mucus to prevent skin from drying out in the hot climate
[15]. Other species, such as penguins in Antarctica, carry
out the amazing collective adaptation of point 4, crowding together to share their warmth and survive glaciation.
Table 1 reports the adaptive solutions to a changing environment employed by some animal species.
In a broader sense, adaptation (and the corresponding
implication of biological intelligence) also means rapidly
changing oneself when moving from one environment to a
different one. Animal species can react to an environmental change in three ways: moving, adapting, or dying [16].
Moving is not always easy, since colonizing a new territory
means competition for spaces and resources with unfamiliar
species. Many species are able to adapt to new environmental conditions through phenotypic plasticity—the ability
of an organism to modify its behavioral and physical features in response to changes in the environment. Coral reefs
adapted to oceans warm without any genetic change through
the expulsion of the symbiotic algae that live within them
[17]. Plasticity can allow genetic adaptations to conquer new
habitats subsequently. Although the definition of intelligence
is controversial, “adaptation to a changing environment,”
“reaction to unexpected situations,” “capacity of problem
solving,” and “ability to communicate” are the most frequently mentioned terms to define it [18]. Brian Ford in a
fascinating article published in 2009, while reintroducing the
ancient Virchow’s concept that “cell is an organism,” suggested that intelligence should be re-thinked and applied at
the cell level. Cell’s behavior and interactions during inflammation or wound repair are suggestive that living cells can
display an intelligent behavior since they have remarkable
abilities to make decisions and take constructive actions,
Cancer and Metastasis Reviews
Table 1 Adaptive solutions to changing environments in different animal species
ANIMALS
STRESSOR
ADAPTATION
TYPE
Structural
ADAPTATION
STRATEGY
Enlargement of
appendages
ADAPTATION
OUTCOME
Heat dispersion
Metabolic
Suppression of
metabolic rate
Reduced energy
supply
Australian frogs
Niche
Mucus niche
construction
Water saving
Penguins
Collective
Crowding
Generate and share
body heat
Birds
Naked mole
rats
O2
independently from brain regulation [19]. Cells usually
know how to be compliant in a community, respecting the
constraints imposed by the microenvironment. Cells know
how to be autonomous while responding to stressors: hypertrophy and hyperplasia are just two examples of how normal
cells adapt to damage, as a further demonstration that they
are often ingenious, being able to solve problems in unexpected situations [20]. Cells communicate with each other
by autocrine, paracrine, endocrine, and contact-mediated
signalling pathways, which are essential to maintain and promote homeostasis. A very peculiar category of cells is represented by invasive cancer cells, which move from native to
distant sites of the body thus being continuously exposed to
new environmental conditions [21]. Even cancer cells, similarly to animal species, can react to an environmental change
in three ways: moving, adapting, or dying. The choice is
often dictated by the fitness of the single tumor cell through
the interaction of its own phenotype with the local environmental conditions. As an example, hypoxic tumor regions
characterized by low perfusion might support the survival
of few, more fit cancer cells, able to adapt to low oxygen
concentrations by activating programs controlling glycolysis, angiogenesis, invasion, immune suppression, and treatment resistance. Conversely, hypoxia will induce detachment
of unfit tumor cells from the primary tumor mass [22, 23].
Whatever the stimulus that causes the detachment of tumor
cells from the primitive mass, once disconnected, cancer
cells enter into the bloodstream, becoming circulating tumor
cells (CTCs) [24]. Blood represents the first, extremely hostile environment that a cancer cell encounters once detached,
so that CTCs must rapidly carry specific adaptations to
survive to potentially destructive shearing forces to evade
the immune system surveillance and to resist to anticancer
drugs. In order to complete the metastatic program, CTCs
must rapidly adapt to this new habitat, react to unexpected
situations, and communicate with the new blood neighbors,
as well as with the tissue-resident neighbors living in premetastatic sites [25]. In other words, CTCs have to use a
complex and “intelligent” adaptation strategy to survive. The
aim of this review was to deepen the adaptation mechanisms
of cancer cells to the blood microenvironment metaphorically referring to four adaptation strategies used by animal
species: phenotypic adaptation, metabolic adaptation, niche
adaptation, and collective adaptation.
1.2 Phenotypic adaptation to stressors: birds
and circulating cancer cells as “shape‑shifters”
Phenotypic plasticity is one of the mechanisms that can produce species adaptations to environmental changes. Birds
are excellent examples of “shape-shifters,” having recently
seen an increase in the size of all appendages such as beaks,
legs, and ears due to climate change. This shape-shifting
plays an important role in regulating body heat dispersion
[26]. Like birds, CTCs are good example of “shape shifters,” adapting to different stressors, including anticancer
drugs, through the acquisition of epithelial mesenchymal
transition (EMT) features [27]. In cancer, EMT is a cellular strategy to adapt to a new environment and to react
to unexpected situations. It consists in a cellular program
that alters cancer cell shape, leading to an elongated form
with a front-back polarity, allowing motility and survival
13
Cancer and Metastasis Reviews
into the circulation through the acquisition of mesenchymal
traits [28]. During EMT, cytoskeletal intermediate filaments
undergo a compositional change as epithelial cells, loosing
keratin intermediate filaments (IFs), E-cadherin, and epithelial specific markers while acquiring mesenchymal markers
such as fibronectin, N-cadherin, and vimentin [29]. Cells
gain a fibroblastoid invasive phenotype, and become resistant to detachment-induced death (anoikis, as explained in
detail below) [30]. Due to the typical change in IF composition, vimentin is considered a typical EMT hallmark [31].
Several lines of evidence suggest that cancer cells exploit
EMT to gain resistance to several treatments, including
chemo-, radio-, and immunotherapy [32]. Whether EMT
features are acquired in the primary tumor or within the circulation is still controversial, although recent data suggest
that—at least in some tumor types—the EMT pathway is
acquired by cancer cells during the hematogenous spread
[33]. Specifically, it has been suggested that mesenchymal
transformation of CTCs might be mediated by transforming growth factor beta (TGF-β) released from platelets [34].
Regardless of EMT inducers, it is widely accepted that EMT
represents one of the key adaptation mechanisms of CTCs to
stressors, including anticancer drugs. In fact, EMT-positive
CTCs have increased expression of antiapoptotic proteins
and transporters belonging to ATP binding cassette family,
that are responsible of drug efflux [35]. Accordingly, several
studies have demonstrated that CTCs isolated from cancer
patients unresponsive to standard anticancer drugs manifest
EMT phenotypes [36]. A pivotal study conducted by Yu
et al. suggested an association of mesenchymal CTCs with
disease progression in breast cancer patients. The authors
compared CTC features in pre- and post-treatment blood
samples reporting an increased number of mesenchymallike CTCs in post-treatment samples [37]. Raimondi et al.
reported that CTCs isolated from patients not responding to
immune checkpoint inhibitors displayed an unusual elongated spindle-like morphology compared to those isolated
from responders, suggesting that elongated CTCs may represent a small population of partial EMT-transformed cancer
cells [38]. Mego et al. demonstrated that CTCs undergoing
EMT are associated to resistance to neoadjuvant treatments
Fig. 1 Parrot-like circulating
tumor cells. EMT markers have
been investigated in CTCs isolated from metastatic colorectal
cancer patients in pre-treatment
(panel A) compared to treatment failure (panel B). A strong
upregulation of the epithelial
marker CK-20 was observed at
treatment failure as compared to
baseline
13
in breast cancer, suggesting a link between therapeutic stress
and activation of EMT program in drug-surviving CTCs
[39]. Oliveras-Ferraros et al. provided experimental evidence that EMT features in CTCs isolated from basal-like
breast cancer patients recognize a new subgroup of HER2
gene-amplified breast carcinomas with primary resistance
to HER2-targeted therapies, such as trastuzumab [40]. Data
obtained from non-small cell lung cancer suggested that
an increase of EMT markers in CTC subpopulations might
be a cause of resistance to treatment with tyrosine kinase
inhibitors (TKIs) [41]. It is becoming increasingly clear that
carcinoma cells, rather than undergoing a full mesenchymal
transformation, often attain a hybrid epithelial/mesenchymal
(E/M) phenotype, also referred to as partial or incomplete
EMT [42]. These cells do not completely loose epithelial
features and do not completely attain mesenchymal traits,
with several studies demonstrating a higher malignant
potential of E/M hybrids as compared to fully mesenchymal
variants [43]. The enhanced drug resistance traits of E/M
hybrids as compared to fully epithelial or fully mesenchymal
cells has been reported in several cancer types. Evidence has
been also provided that cancer cells with EMT features need
to re-upregulate epithelial markers, undergoing mesenchymal-epithelial transition (MET) immediately before colonizing distant sites. Consistently, in a population of metastatic
colorectal cancer patients treated with chemotherapy and
antiangiogenic drugs, we recently observed that CTCs isolated at treatment failure re-upregulated epithelial features,
probably reflecting cells undergoing MET, characterized by
high plasticity and strongly committed to metastatic spread
(unpublished data and Fig. 1). Metaphorically speaking, the
phenotypic plasticity adopted by CTCs in response to a new
and hostile microenvironment is reminiscent of phenotypic
alterations described in parrots under climate change.
Taking into consideration the most popular signatures
of intelligence (“adaptation to a changing environment,”
“reaction to unexpected situations,” “capacity of problem
solving,” and “ability to communicate”), EMT plasticity of
circulating tumor cells seems to meet all these requirements
well. In fact, the phenotypic change typical of EMT represents a cell adaptation to a changing microenvironment,
Cancer and Metastasis Reviews
such as hypoxia, acidosis, and change in glucose levels [44].
Referring to the definition of intelligence as “capacity of
problem solving,” one of the main features of the EMT process is anoikis resistance [45]. Anoikis (from ancient Greek:
“without a home”) is a sort of apoptotic cell death occurring upon insufficient cell-matrix interactions, representing a major problem for a cancer cell leaving tissues [46].
The dramatic phenotypic alterations driven by EMT allow
cells to evade normal-tissue architectural constraints, and
to escape from the primary tumor. Some CTCs through the
activation of Akt, PI3K, or epidermal growth factor receptor
(EGFR) pathways trigger autonomous survival mechanisms
enabling EMT-shifted cells to better resist anoikis [47]. This
is a clear example of how to solve a problem in a short time.
Coming to the “ability to communicate,” the dynamic crosstalk between circulating tumor cells and other blood cells
is increasingly recognized as a key regulator of malignant
progression [48]. A large body of evidence has been provided that platelets are more than physical shields for CTCs,
actively communicating with CTCs through TGF-β release
to potently induce EMT phenotype [49]. CTCs “communicate” with platelets through their ability to express tissue
factor (TF), determinant for CTC survival and seeding [50].
Also, CTCs have been shown to communicate with other
immune cells, including neutrophils [51]. EMT induces several receptors mediating interactions between neutrophils
and CTCs, including CD44, ICAM-1, and VCAM1. These
results suggest that EMT-shifted CTCs are particularly
efficient in communicating with platelets and neutrophils,
which both support cancer cells during their journey in the
bloodstream, allowing survival, resistance to shear stress,
and initiation of the metastatic niche. From these reflections,
one could deduce that EMT adopted by circulating cancer
cells mimics an intelligent behavior (Fig. 2).
1.3 Metabolic adaptations to stressors: naked
mole‑rats and circulating tumor cells
as “metabolism switchers”
Hypoxia is one of the strongest environmental drivers of
cellular metabolic adaptation [52]. Although many species are generally intolerant to hypoxia, some have evolved
adaptive strategies to survive in hypoxic niches [53]. The
key to tolerating hypoxia is to match metabolic demand to
reduced energy supply. The naked mole-rat (Heterocephalus
glaber) is the most hypoxia-tolerant mammal that adapted to
subterranean environment remaining active despite a rapid
suppression of its metabolic rate [54]. In cancer, hypoxia
induces a plethora of cellular responses, including EMT
induction, suppression of apoptosis, enhanced angiogenesis,
malignant progression, and metabolic reprogramming, the
latter allowing cell proliferation despite low-oxygen concentration [55]. Hypoxia-inducible factor 1 (HIF-1) is a key
regulator of the metabolic reprogramming in hypoxic cancer
cells through the regulation of genes such as the glucose
transporters GLUT1 and GLUT3, hexokinase 1 and 2 (HK1
and HK2), and phosphoglycerate kinase 1 (PGK1), finally
orchestrating the metabolic changes necessary to adapt to
oxygen deprivation [56]. HIF is a heterodimeric complex
consisting of one of the oxygen-regulated α-subunit isoforms (HIF-1α, HIF-2α, or HIF-3α) and the constitutively
expressed subunit HIF-1β. While in normoxic conditions
Fig. 2 EMT as an “intelligent”
behavior of cancer cells. EMT is
a phenotypic adaptation of cancer cells to microenvironment
stressors (1). It also represents
a strategy of problem solving
since the phenotypic alterations
driven by EMT allow cells to
elude anoikis through the activation of Akt, PI3K, or EGFR
pathways (2). EMT-like CTCs
are able to “communicate” with
platelets through their ability
to express TF, determinant for
CTC survival and seeding. On
the other hand, platelets are
strong inducers of EMT-like
CTCs through the release of
TGF-β (3)
13
Cancer and Metastasis Reviews
HIF-1α is regulated by hydroxylation, under hypoxia,
HIF-1α hydroxylation is reduced, leading to its accumulation and nuclear translocation where the dimerization with
HIF-1β occurs [57]. The HIF-1α/β dimer binds to hypoxiaresponse elements (HREs) of oxygen-dependent genes,
including glucose transporter genes, glycolytic enzymes, and
angiogenic and hematopoietic growth factors [58]. HIF-1α
can upregulate pyruvate dehydrogenase kinase (PDK1),
lactic dehydrogenase A (LDHA), and pyruvate kinase M2
subtype (PKM2), a key enzyme implicated in the last irreversible step of glycolysis. In addition to enhanced glycolysis, hypoxia activates the pentose phosphate pathway
(PPP) [59]. HIF-1α is a common link between adaptation to
hypoxia, changes in cancer metabolism, and cancer progression [60–62]. In this context, evidence has been provided
that hypoxia is a major promoter of EMT through the HIF
pathway, being hypoxia and EMT strictly interconnected
[63]. HIF-1 induces EMT through several pathways, including TGF-β/SMAD, Wnt/beta catenin, Hedgehog, FOXM1,
and through activation of EMT-specific transcription factors
[64]. Notably, hypoxia can be induced by some anticancer
treatments, such as antiangiogenic drugs [65]. Evidence
has been provided that vascular endothelial growth factor
A (VEGFA)-targeted agents such as bevacizumab promote
intra-tumoral hypoxia through blocking tumor angiogenesis.
In turn, hypoxia induces EMT, forces cells to abandon their
native home, and reprograms the cancer stem cell niche thus
favoring cancer progression [66]. Hypoxia is crucial in CTC
formation, since it promotes cell invasion. Evidence has
been provided that both hypoxic and normoxic cancer cells
are able to intravasate [67]. Nevertheless, the survival of
CTCs is strongly influenced by hypoxia, which directly stimulates the malignant properties of cancer cells through the
expression of multiple genes associated with angiogenesis,
metabolic regulation, cell apoptosis, and EMT [68]. Godet
et al. demonstrated that cancer cells exposed to low oxygen
pressure, and then to the bloodstream, acquire a “hypoxic
memory or genetic signature” that is maintained even after
the cells are re-oxygenated. The authors demonstrated that
cancer cells exposed to physiological levels of hypoxia in the
primary tumor have a 4× greater probability of becoming a
Fig. 3 Hybrid parrots/naked
mole-rats metaphorically represent circulating tumor cells.
EMT and hypoxia features have
been investigated in CTCs isolated from metastatic colorectal
cancer patients in pre-treatment
(panel A) compared to treatment
failure (panel B). In panel B, a
strong upregulation of HIF-1
compared to baseline is shown.
Most CTCs maintained hybrid
EMT/hypoxic characteristics
13
viable CTC and that post-hypoxic cells have an enhanced
metastasis-initiating capability [69]. A study by Kallergi
et al. demonstrated that immunomagnetically isolated CTCs
co-expressed VEGF and HIF-1α, suggesting that the activation of pro-angiogenic pathways in CTCs could result in
evasion of apoptosis, enhancement of metastatic potential,
and resistance to endocrine therapy [70]. Accordingly, undifferentiated cells expressing HIF-1α and enriched in mesenchymal phenotypes have been observed at the invasive
edge of colorectal cancer [71]. Consistent with this hypothesis, our group recently observed that CTCs isolated from
colorectal cancer patients who rapidly progressed in course
of treatment with chemotherapy plus antiangiogenic drugs
co-express HIF-1α and vimentin in response to prolonged
drug exposure (unpublished data, Fig. 3). Both HIF-1α and
vimentin were found consistently upregulated in CTCs at
the time of treatment failure as compared to baseline. At a
metaphorical level, in response to anti-angiogenic and cytotoxic treatments, CTCs implement two different adaptation
strategies at the same time, i.e., hypoxia and hybrid E/M
phenotypes, becoming a sort of mythological creatures half
bird and half naked mole-rat.
Let’s come again to the above-stressed requirements for a
sensible use of intelligence metaphor: “adaptation to a changing environment,” “reaction to unexpected situations,” “capacity of problem solving,” and “ability to communicate.” Does
the metabolic adaptation of CTCs meet all these requirements?
The hypoxic signature of CTCs is certainly linked to a metabolic switch which represents a cell adaptation to a changing
microenvironment [72]. From this point of view, the change
in metabolism of the tumor cell during hypoxia represents a
further example of a reaction to an unexpected situation. It is
well established that hypoxic stress is a feature of most solid
tumors, arising from excessive oxygen consumption by growing tumor cells and the functionally inefficient tumor-associated
vasculature [73]. Hypoxia is lethal for many cancer cells that are
not able to rapidly adapt to the mutated conditions. Therefore,
hypoxia represents a problem that must be rapidly solved. The
solution, adopted only by the most “intelligent” cells, consists
in an adaptive response to hypoxia, involving HIF upregulation and the subsequent activation of cancer hallmarks such
Cancer and Metastasis Reviews
as angiogenesis, cell survival, proliferation, and metabolism
switch. Coming to intelligence as “ability to communicate,” HIF
has been reported to stimulate the production of CD47, a protein
that enables cancer cells to avoid destruction by immune cells.
Consistently, the analysis of circulating tumor cells isolated from
the blood of breast cancer patients revealed that CD47 expression identified a subpopulation of cells with the capability to
generate tumor xenografts in mice [74]. In the light of what has
been stated in this paragraph, we postulate that the adaptation
of CTCs to hypoxia is a further analogy of intelligent behavior,
being part of a complex adaptation mechanism including phenotypic, metabolic, and communicative modifications (Fig. 4).
Should we think of them as particularly intelligent?
1.4 Niche adaptations to stressors: Australian frogs
and circulating tumor cells as “niche organizers”
The niche-construction perspective within evolutionary biology
places emphasis on the changes that organisms bring about in
their selective environments to adapt to stressors [75]. Australian frogs, in order to adapt to draught, enclose themselves in a
transparent waterproof cocoon made from layers of shed skin
until the next wet period. In this mucus niche they can stay for
up to 7 months while waiting for rain. In the cancer ecosystem,
a pre-metastatic niche (PMN) is defined as an environment in
a secondary organ that can be conducive to the survival and
outgrowth of tumor cells before their arrival at these sites [76].
Thus, in contrast to the metastatic niche that is initiated and
shaped upon arrival of cancer cells, the PMN is deficient in
cancer cells but conducive to their tumor growth. Both tumorsecreted factors and tumor-shed extracellular vesicles (EVs)
promote the evolution of PMNs through a sequence of events,
starting from vascular leakiness to the alteration of local resident fibroblasts and the further recruitment of non-resident bone
marrow-derived cells (BMDCs), finally attracting circulating
tumor cells [77]. The establishment of PMNs facilitates metastasis by promoting CTC survival and outgrowth. A large body
of evidence demonstrate that only a small subset of circulating
tumor cells are able to form metastases [78]. Although little is
known about the underlying mechanisms of CTC colonization
in pre-metastatic niches, the specific niche microenvironment
is supposed to “educate” CTCs prior of their arrival in distant
organs, contributing to the survival of tumor cells before they
reach metastatic sites [79]. Tang et al. investigated the underlying mechanisms of hepatocellular carcinoma CTC colonization
in pre-metastatic niches, reporting that SDF-1 in the microenvironment induces the chemotaxis of circulating CXCR4-positive
CTCs to potential target organs [80]. Similarly to Australian
frogs that adapt to drought until the next rain, a subpopulation
of CTCs adapt to the PMN environment by acquiring enabling
Fig. 4 Metabolic adaptation as
an “intelligent” behavior of circulating cancer cells. Hypoxia is
a potent driver of EMT. Under
hypoxia, CTCs maintain EMT
features (1), reacting to this
unexpected situation through
HIF upregulation and the subsequent activation of cancer hallmarks such as metabolic switch,
survival, and metastasis (2).
HIF stimulates the production
of CD47, which enables cancer
cells to communicate with
innate immune system cells in
order to avoid destruction (3)
13
Cancer and Metastasis Reviews
characteristics, until their arrival in distant metastatic sites. This
subpopulation is mainly composed by cancer stem cells (CSCs)
that exhibit stem-like properties and are strongly committed to
efficiently colonize distant organs [81]. It has been recently suggested that CSCs and CTCs might reflect different functional
states of the same subpopulation of cancer cells, being CSCs the
only fraction of CTCs capable of giving rise to tumors in secondary recipients [82]. Notably, CSCs are characterized by functional plasticity for their ability to switch between mesenchymal-like and epithelial-like states; these observations led to the
hypothesis that EMT is not only associated to invasive phenotype, but may also induce stemness characteristics [83]. Furthermore, CSCs have been proven to be highly resistant to standard
anticancer treatments, making them a plausible cause of tumor
relapse [84]. Papadaki and colleagues analyzed CTCs from
breast cancer patients for CSC and EMT features, demonstrating
a link between the CSC+/partial EMT and reduced progressionfree survival (PFS). Interestingly, EMT-like CTCs displaying
CSC features were found only in patients non-responders to
chemotherapies [85]. Several lines of evidence obtained in
solid tumors support the presence of CSCs with tumor-initiating
capabilities within the blood [86, 87]. These cells are characterized by specific cell surface expression profiles, including
CD44, CD24, CD133, CD166, and ALDH1 [88]. Gradilone
et al., who investigated the expression of EMT and stemness
markers in breast cancer CTCs, reported a significant association
between EMT features and ALDH1 in CTCs from drug resistant
patients, underlining the urgent need for optimizing CTC detection methods through the combination of EMT markers with
CTC phenotype [89]. In colorectal cancer, CD44v6, the CD44
isoform mostly involved in cancer cell migration and invasion,
has been identified as a functional marker of CSCs [90–93].
Nicolazzo et al. [94] provided evidence that CD44v6-positive
CTCs predict treatment failure in patients with metastatic colorectal cancer undergoing first-line chemotherapy, suggesting
that CD44v6-positive CTCs reflect intrinsic drug resistance in
this cancer type. Consistently, we recently observed a strong
upregulation of CD44v6 in CTCs isolated from colorectal cancer patients at the time of treatment failure compared to baseline
(unpublished data, Fig. 5). Interestingly, all CTCs with stemness
features maintained EMT traits. Again, CTCs which survived
Fig. 5 Hybrid parrots/frogs as
a metaphor of circulating tumor
cells. EMT and stemness markers were assessed in CTCs isolated from metastatic colorectal
cancer patients in pre-treatment
(panel A) compared to treatment failure (panel B). In panel
B, a strong upregulation of
CD44v6 compared to baseline
is shown
13
treatments implemented two different adaptation strategies in the
meantime, becoming a half frog and half naked mole-rat, further
supporting that EMT-related plasticity is necessary for CTCs to
acquire stem-like features.
Let’s turn to clarify whether EMT+/CSCs can meet all the
four requirements of “intelligent” behavior, i.e., “adaptation
to a changing environment,” “reaction to unexpected situations,” and “capacity of problem solving.” Travelling in the
bloodstream is a difficult time for CTCs. They rapidly need to
adapt to various unexpected situations, specifically to all the
sources of genotoxic stress (mechanical stress, oxidative stress,
oncogene-induced replication stress). Evidence has been provided that CTCs experience a great amount of oxidative stress
in the bloodstream with increased metabolic demand of the
mitochondria, and that CTCs-specific elevated mitochondrial
energy production induces a stemness gene set as an adaptive
response [95]. Another difficult-to-solve problem is oncogeneinduced replication stress. It has been widely reported that
excess of MYC drives the cell into rapid cell cycle divisions,
exacerbating multiple sources of endogenous replication stress
[96]. MYC amplification is one solution to allow CTCs to survive endogenous replication stress during their journey from the
primary tumor to the distant metastatic site. Consistently, MYC
alterations have been described in 62% of CTCs+ patients. In
breast cancer, MYC-expressing cells acquire CSCs and EMT
features [97]. Clusters of cancer-associated fibroblasts (CAFs)
together with circulating CSCs have been reported in many
cancer types [98]. These complexes help CSCs to survive in
the blood and probably to colonize the pre-metastatic niche. In
mice, the co-injection of MCF7 cells and CAFs from a triple
negative breast cancer patient led to enrichment of CSCs-like
CTCs, triggering the presence of circulating CAFs/CSCs clusters [99]. Furthermore, a strict crosstalk exists between circulating CSCs and immune cells, which is mediated through
immune targets, as well as through extracellular vesicles (EVs)
that enable the transfer of large biomolecular cargos among different types of cells through the release of interleukins, matrix
metalloproteinases, and growth factors [100]. Furthermore, it
has been demonstrated that the expression of programmed cell
death ligand 1 (PD-L1), which allows circulating tumor cells to
elude immune attack, is often co-expressed with EMT/stemness
Cancer and Metastasis Reviews
features in CTCs, representing for these cells a possible molecular background for immune escape [38]. Altogether, the acquisition of stemness traits and PD-L1 overexpression by CTCs
further reinforces the analogy of a multifaceted “intelligent”
adaptation strategy (Fig. 6).
Crowding in large groups is one of the behavioral adaptations of penguins during storms to generate and share
body heat, ensuring survival in severe climate. Huddling
is a sort of social thermoregulation mechanism, since the
center of the huddle can reach temperatures of up to 37° Celsius [101]. Like penguins, CTCs build social relationships
through cluster formation, since individual adaptation is not
sufficient for a successful society [102]. Clusterization has
been reported as an adaptive mechanism that enhances CTC
survival in the bloodstream, and recent studies have suggested that CTC clusters, rather than being a simple aggregation of cancer cells, represent a highly specialized microenvironment [103]. A homotypic CTC cluster is defined as
the aggregation of two or more cancer cells through intercellular adhesion. Cancer cells can detach from the primary
site in clusters, as demonstrated by the evidence that collective migration is one of the mechanisms of invasion in
cancer-host interface in human pancreatic, colorectal, lung,
and breast adenocarcinomas [104]. Individual cancer cells
often clump together in the blood, since single cells are more
prone to apoptosis [105]. As compared to single CTCs, CTC
clusters have the physical advantage to resist to anoikis and
to be trapped in distant organs, thus being characterized by
an increased metastasis-seeding ability [106–108]. In breast,
lung, and prostate cancer, CTC clusters are reported to
metastasize at 20–100 times greater efficiency and patients
with higher numbers of CTC clusters have worse prognosis
in terms of progression-free survival and overall survival
compared to patients with single CTCs [109]. Evidence has
been provided that CTC clusters better undergo margination, rotation, and adherence to endothelium than individual
CTCs [110]. Several studies aimed to investigate the mechanisms of CTC clusters formation have shown that intratumor hypoxia results in cell-cell junction upregulation and
intravasation of CTC clusters; specifically, in breast cancer,
CTC clusters seem to derive from the hypoxic regions of the
primary tumor. Another crucial trigger for cluster formation is CD44, which leads to multicellular aggregation via
its target PAK2, suggesting that stemness might play a role
in cluster generation through the formation of homophylic
intercellular interactions [111]. Furthermore, CTC clusters
contain hybrid E/M phenotypes, expressing both epithelial
and mesenchymal markers. Combined stemness and EMT
features might contribute to resistance to anoikis, explaining
Fig. 6 Acquisition of stemness features as an “intelligent” behavior
of circulating cancer cells. Reactive oxygen species (ROS) are crucial in EMT engagement. CTCs acquire EMT plasticity when they
experience oxidative stress in the bloodstream with increased metabolic demand of the mitochondria (1). CTCs react to blood stressors
through MYC amplification, which allows CTCs to survive endogenous replication. Another adaptation consists in the acquisition of
stemness features, which allow CTC to prepare to colonize the premetastatic niche (2). A strict crosstalk exists between circulating CSCs
and immune cells, which is mediated through immune targets, as well
as through EVs that enable the transfer of large biomolecular cargos
among different types of cells through the release of interleukins,
matrix metalloproteinases, and growth factors, finally attracting CTCs
to colonize the pre-metastatic niche (3)
1.5 Collective adaptations to stressors: penguins
and circulating tumor cells as “social
relationship builders”
13
Cancer and Metastasis Reviews
the known pro-metastatic driven properties of clustered
tumor cells [112, 113]. A link between CTC clusters and
anticancer drug resistance has been previously described.
In ovarian cancer, the presence of CTC clusters correlates
with platinum resistance [114]. Furthermore, experiments
with clusters of tumor cells compartmentalized in microfluidic drops demonstrated that clusters are more resistant
to doxorubicin compared to their single-cell counterparts
[115]. The currently used platforms for CTC detection are
not ideal for cluster isolation, while size-exclusion assays
such as blood filtration represent an affordable approach
for CTC cluster isolation [116, 117]. Our group recently
described a new method for the simultaneous isolation of
CTC clusters and single CTCs from a single blood draw
through a sequential filtration, using adapted ScreenCell®
filters with increased pore size. We validated the assay in a
small population of patients with metastatic colorectal cancer [118]. Consistently with literature studies, CTC clusters
had more prominent hybrid-EMT features compared to single CTCs; furthermore, single CTCs significantly differ from
clusters in terms of HIF-1α expression, found constantly
expressed in clusters, but not in single CTCs. We further
investigated EMT, stemness, and hypoxic features in clusters
isolated from metastatic colorectal cancer patients at pretreatment and treatment failure (unpublished data, Fig. 7).
Although clusters were detected at both timepoints, vimentin, CD44v6, and HIF-1α were found strongly upregulated
at treatment failure compared to baseline.
Can clustering be viewed as an “intelligent” behavior
of cancer cells? Based on current knowledge, clusters are
able to outperform single CTCs in seeding metastasis with
an up to 100-fold higher efficiency. Thus, the “social”
Fig. 7 Circulating tumor clusters isolated from metastatic colorectal cancer patients remind of a penguin-like behavior. Clusters are detected at
both pre-treatment (panel A) and at treatment failure (panel B)
13
Cancer and Metastasis Reviews
behavior of CTCs can be viewed as part of CTCs “intelligent” strategy to carry out the metastatic program.
2 Conclusions: why can it be productive
to consider cancer as an intelligent
species?
Before going ahead and giving an answer to the most crucial question raised by this review, it is worth noting that
metaphors in science are the most powerful hypothesis
generators. Probably the most astonishing example of
this fact is the construction of periodic table of elements
by Dimitri Ivanovic Mendeleev around the half of nineteenth century. Mendeleev did not know anything about
the structure of atoms (that only after more than 50 years
was recognized to fit with its “octave”-based classification of elements), but he was a very good piano player.
Moreover, his most strict collaborator was Aleksandr
Porfirevic Borodin that, in addition to be a brilliant organic
chemistry, was one of the most renowned composers of
his times. Mendeleev had only knowledge of the relative
weight of 32 elements and of their “valence rules,” i.e.,
the relative proportion of mixing into a chemical reaction
so to obtain stable products. The musical background of
Mendeleev pushed him to adopt the metaphorical frame
of tonal music theory to order the known elements into
groups substituting elements to music tones and valence
rules to the interval ratios considered as “consonant” in the
well-tempered scale [119]. This choice not only allowed to
sketch a consistent ordination of the 32 elements known
at his times but allowed to predict the position in the system and the chemical properties of the elements that were
discovered in the future times. We do not think our review
work can be compared to the almost unique achievement
of Mendeleev; nevertheless, we wish to stress that, even if
not immediately evaluable in classical quantitative terms,
our concept of “cancer as an intelligent species” can be of
use for scientists to interpret many puzzling phenomena
of cancer development and progression. Going back to
the core of our work, it is worth noting that in the bloodstream, a highly heterogeneous community circulates,
since not all cells are equal [120]. This is a further demonstration of cancer biological intelligence, since an ecosystem where all individuals are identical is inexorably
doomed to extinction. The fate of most CTCs in the hostile blood environment is death. Conversely, the smartest
of the group quickly implement a set of different adaptation strategies, reminding a sort of mythological creatures
composed by parts of different animals. It is amazing how
CTCs are able to adopt more than one adaptation strategy
at the same time. We have described circulating tumor
cells capable of simultaneously acquiring characteristics
of phenotypic and metabolic plasticity, able to increase
their motility through EMT and to survive hypoxia through
metabolic switch (half bird, half naked mole-rat). Others are capable of acquiring phenotypic plasticity and an
extraordinary ability to adapt to specific niches to better metastasize (half bird, half Australian frog). Although
hybridization is not an adaptive solution to increase fitness in most animal species (while hybridization can be
a very adaptive trait in plants [121]), it provides a great
opportunity to cancer cells. Donkeys and horses can breed
to create mules, but hybrids are usually at risk of extinction, being sterile. Despite usually being an evolutionary
dead end, sometimes hybridization introduces beneficial
genes (adaptive introgression) [122]. CTCs seem more
prone than animal species to something which is in some
way similar to adaptive introgression. In conclusion, we
provided evidence that CTCs might fulfil all the strategic steps to fall into the broader definition of biological
intelligence. The required adaptations (either structural,
metabolic, and related to metastatic niche formation) and
“social” behavior are apparently recognized in circulating cancer cells as exemplified in the parallel behavior of
several real macroscopic animal models (Fig. 8).
Coming to the presumed biological intelligence of cancer, which eventually activates a program of self-destruction
causing the death of the host, one might argue that cancer
behaves like a kamikaze. Is committing suicide after this
enormous adaptation effort an intelligent behavior? Wouldn’t
it be more successful to establish a peaceful cohabitation
with its host? From a clinical perspective, establishing a
long-lasting coexistence between patient and tumor may
represent an intelligent therapeutic strategy [123]. The final
answer to these questions will progressively emerge from
the parallel evolution of tumors and anticancer treatments,
predictably involving on both sides new amazing adaptation
strategies.
2.1 Re‑interpreting cancer as an “intelligent”
species: hints from the genome architecture
theory
The so-called species problem, which refers to the lack of
agreement upon a clear definition of species, was recently
addressed in the context of the genome architecture theory,
proposed by Heng as a new tool to unify cancer with evolutionary biology strategy [5]. According to this theory, a species
is a population that shares the same genome architecture. Thus,
if we move from the concept of individual genes to that of the
whole genome as the primary unit of heritable information,
cancer represents a new species being cancer cells karyotypes
totally different from those of host cells. Without neglecting
the importance of individual genes in the evolutionary process, Heng postulates that the variation of single genes leads
13
Cancer and Metastasis Reviews
Fig. 8 A–E Real and imaginary animal models as a metaphoric representation of structural, metabolic, niche-related, and “social” modifications
of circulating cancer cells
to a slow and gradual microevolution (the Darwinian theory
of natural selection), while the variation of the genome leads
to macroevolution which represents a sort of “explosion” able
to suddenly generate new species. This “genome chaos” is the
evolutionary mechanism that guides cancer progression leading
to the generation of new species when the environment suddenly changes (as in course of anticancer treatments which act
collectively as stressors). Thus, genome chaos can be viewed
as an “intelligent” strategy to rapidly deliver macroevolutionary success by providing survivable karyotypes. This is exactly
what happens in massive extinction, when a specific ecosystem
suddenly is destroyed providing the opportunity for the less represented species to evolve, diversify, and become more prevalent. Although genome chaos will be lethal for some species,
the most resistant will then adapt through gene mutations and
epigenetic alterations with microevolution as the key feature
[124]. This theory is known as the two-phased cancer evolution.
According to Heng’s theory, even drug resistance is guided by
genomic chaos (therefore by stress-induced macroevolution,
not by microevolution) and generates new cellular species.
Elegant in vitro studies have demonstrated that many anticancer
drugs, independently from their mechanism of action, induce
genome chaos resulting in the appearance of resistant clones
13
with different structural and numerical karyotypes compared to
the parental cell line. In light of this, might cancer cell species
that circulate in the blood in course of treatment failure, which
in this review have been ironically designed as bizarre hybrid
animals, be the result of genomic chaos? To answer this question, while reading Heng’s genomic chaos theory, it occurred to
us that genomic chaos in response to various genotoxic stresses,
including anticancer treatments, may generate polyploid giant
cancer cells (PGCCs), characterized by multiple nuclei or a single giant nucleus containing multiple complete sets of chromosomes [125]. The mechanism leading to formation of PGCCs
may depend on endoreplication, mitotic slippage, cytokinesis
failure, or even cell fusion [126]. Although a circulating tumor
cell is classically defined as expressing epithelial markers such
as EPCAM and cytokeratin (CK) and lacking the leukocyte
marker CD45, the presence of CTCs with both epithelial and
leukocyte markers (dual-positive cells, DP cells, CK+/CD45+)
in the blood of cancer patients has recently been reported. In our
experience, such hybrid CTCs are frequently detected through
CellSearch platform in treatment-resistant patients (Fig. 9).
Evidence has been provided that circulating DP cells
are hybrids deriving from the fusion of tumor cells and
macrophages. In some tumor types, i.e., triple negative
Cancer and Metastasis Reviews
Fig. 9 Image galleries of dual
positive circulating tumor cells
isolated from treatment-resistant
breast cancer patients using the
CellSearch platform. Immunofluorescence staining for epithelial markers (CK), leukocyte
markers (CD45), and DAPI
breast cancer, such hybrids present aberrant genomes and
are associated with worse overall survival [127]. Indeed,
several studies have demonstrated that the fusion of cancer
cells with leukocytes produces hybrids with high metastatic competence by combining the epigenetic program
of the leukocyte with the uncontrolled cell division of the
cancer cell [128]. It is conceivable that a better characterization of DP circulating cancer cells might help to shed
light on genomic chaos induced by anticancer drugs and
unveil opportunities for therapeutic targeting. In conclusion, if cancer cells go through a rapid macroevolution
following drug exposure, we should re-think about drug
resistance as an active choice of cancer cells and not as the
passive phenomenon known as treatment selection according to Darwinian natural selection theory. Thus, if we can
think of drug resistance as the result of cancer evolvability
through macroevolution, genome chaos might be thought
as a very “intelligent” (in metaphoric sense) adaptive evolutionary strategy, allowing the formation of more aggressive cancers as a response to anticancer drugs. Such rapid,
adaptive, genome-based macroevolution of cancer allows
cancer cells to (1) change themselves to better fit a new
environment, (2) shape the environment to better fit their
needs, and (3) choose a new environment that better fits.
To support our metaphor about cancer intelligence, we
believe that the apparently eccentric view of cancer as an
intelligent system of collaborating and computing cells
(echoing the Frost [129] use of concepts borrowed by computation, game playing, and machine learning in defining
cancer intelligence) deserves a thorough attention allowing to overcome some too reductionist and fatally partial
views of cancer. Going back to the periodic table of elements, here we face an opposite difficulty with respect to
Mendeleev: while he had to cope with a too small set of
information (few chemical elements, minimal number of
features) in cancer research, we have a plethora of “pieces
of evidence” to be summarized into few basic concepts.
A recent paper [130] demonstrates that more than 87% of
human genes are somewhat related to cancer. These figures
are the signature of an entropic information catastrophe:
we can imagine an almost infinite number of “reliable”
mechanisms of cancer if we remain linked to the usual
quasi-deterministic pathway way of reasoning. This situation must be overcame by strong synthetic efforts at a more
systemic scale of analysis, and the “cancer as intelligent
species” hypothesis could be a promising candidate.
13
Cancer and Metastasis Reviews
Acknowledgements This paper was inspired by reading “Adaptive
Intelligence Surviving and Thriving in Times of Uncertainty” by Robert J. Sternberg. This work is dedicated to Claudio Apuzzo with whom
I shared and discussed images of his own cancer cells and who, even at
the end of his war against the hybrid monster, asked me to never give
up. We thank R. Rossi for manuscript editing.
9.
10.
Author contribution C.N. and F.F. performed the experiments, the literature search and data analysis. V. M drafted the work. P.G. ideated
and wrote the manuscript. P.G., A.G., and A.Z. critically revised the
work.
11.
Funding Open access funding provided by Università degli Studi di
Roma La Sapienza within the CRUI-CARE Agreement. This review
was partially supported by the Italian Association for Cancer Research
(AIRC) Grant No. 20744 awarded to A.Z.
12.
Declarations
13.
Conflict of interest The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format,
as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material
in this article are included in the article's Creative Commons licence,
unless indicated otherwise in a credit line to the material. If material
is not included in the article's Creative Commons licence and your
intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the
copyright holder. To view a copy of this licence, visit http://creat
ivecommons.org/licenses/by/4.0/.
14.
15.
16.
17.
References
1. Boveri, T. (1902). In W. Kabitzch (Ed.), Über mehrpolige
Mitosen als Mittel zur Analyse des Zellkerns. [Concerning
multipolar mitoses as a means of analysing the cell nucleus]
(Vol. 35). Verh. d. phys. med. Ges. zu WÜrzburg. N.F.
2. Duesberg, P., & Rasnick, D. (2000). Aneuploidy, the somatic
mutation that makes cancer a species of its own. Cell Motility
and the Cytoskeleton, 47(2), 81–107. https://doi.org/10.1002/
1097-0169(200010)47:2<81
3. Vincent, M. D. (2010). The animal within: Carcinogenesis and
the clonal evolution of cancer cells are speciation events sensu
stricto. Evolution, 64(4), 1173–1183. https://doi.org/10.1111/j.
1558-5646.2009.00942.x
4. Heng, H. H. (2015). Debating cancer: The paradox in cancer
research. World Scientific Publishing Co..
5. Heng, J., & Heng, H. H. (2022). Genome chaos, information
creation, and cancer emergence: Searching for new frameworks
on the 50th Anniversary of the “War on Cancer”. Genes, 13, 101.
https://doi.org/10.3390/genes13010101
6. Wilson, E. O. (1998). Consilience: The unity of knowledge.
Knopf.
7. Malan, G. (2016). Myth as metaphor. HTS Teologiese Studies/
Theological Studies, 72(4), a3260. https://doi.org/10.4102/hts.
v72i4.3260
8. Neisser, U., Boodoo, G., Bouchard, T. J., Jr., Boykin, A. W.,
Brody, N., Ceci, S. J., Halpern, D. F., Loehlin, J. C., Perloff, R.,
Sternberg, R. J., & Urbina, S. (1996). Intelligence: Knowns and
13
18.
19.
20.
21.
22.
23.
unknowns. American Psychologist, 51, 77–101. https://doi.org/
10.1037/0003-066X.51.2.77
Jerison, H. J., & Sternberg, R. J. (1982). The evolution of biological intelligence. In Handbook of human intelligence (pp.
723–791). Cambridge University Press https://books.google.it/
books?id=VG85AAAAIAAJ
Liu, J., & Nussinov, R. (2016). Allostery: An overview of its history, concepts, methods, and applications. PLoS Computational
Biology, 12(6). https://doi.org/10.1371/journal.pcbi.1004966
Sternberg, R. J. (2019). A theory of adaptive intelligence and its
relation to general intelligence. Journal of. Intelligence, 7(4).
https://doi.org/10.3390/jintelligence7040023
Ryding, S., Klaassen, M., Tattersall, G. J., Gardner, J. L., &
Symonds, M. R. E. (2021). Shape-shifting: Changing animal
morphologies as a response to climatic warming. Trends in Ecology & Evolution, 36(11), 1036–1048. https://doi.org/10.1016/j.
tree.2021.07.006
Grava, A., Grava, T., Didier, R., Lait, L. A., Dosso, J., Koran, E.,
et al. (2012). Interspecific dominance relationships and hybridization between black-capped and mountain chickadees. Behavioral Ecology, 23(3), 566–572. https://doi.org/10.1093/beheco/
arr229
Park, T. J., Smith, E. S. J., Reznick, J., Bennett, N. C., Applegate,
D. T., Larson, J., & Lewin, G. R. (2021). African naked molerats demonstrate extreme tolerance to hypoxia and hypercapnia.
Advances in Experimental Medicine and Biology, 1319, 255–269.
https://doi.org/10.1007/978-3-030-65943-1_9
Reynolds, S. J., Christian, K. A., & Tracy, C. R. (2010). The
cocoon of the fossorial frog Cyclorana australis functions primarily as a barrier to water exchange with the substrate. Physiological and Biochemical Zoology, 83(5), 877–884. https://doi.org/
10.1086/656218
Radchuk, V., Reed, T., Teplitsky, C., van de Pol, M., Charmantier, A.,
Hassall, C., & Kramer-Schadt, S. (2019). Adaptive responses of animals to climate change are most likely insufficient. Nature Communications, 10(1), 3109. https://doi.org/10.1038/s41467-019-10924-4
Bowden-Kerby, A. (2022). Coral-focused climate change adaptation and restoration based on accelerating natural processes:
Launching the “Reefs of Hope” paradigm. Oceans, 4(1), 13–26.
https://doi.org/10.3390/oceans4010002
Deary, I. J. (2012). Intelligence. Annual Review of Psychology, 63(1), 453–482. https:// doi. org/ 10. 1146/ annur
ev-psych-120710-100353
Ford, B. J. (2017). Cellular intelligence: Microphenomenology
and the realities of being. Progress in Biophysics and Molecular
Biology, 131, 273–287. https://doi.org/10.1016/j.pbiomolbio.
2017.08.012
Agozzino, L., Balázsi, G., Wang, J., & Dill, K. A. (2020). How
do cells adapt? Stories told in landscapes. Annual Review of
Chemical and Biomolecular Engineering, 11(1), 155–182.
https://doi.org/10.1146/annurev-chembioeng-011720-103410
Neinavaie, F., Ibrahim-Hashim, A., Kramer, A. M., Brown, J. S.,
& Richards, C. L. (2021). The genomic processes of biological
invasions: From invasive species to cancer metastases and back
again. Frontiers in Ecology and Evolution, 9, 681100. https://doi.
org/10.3389/fevo.2021.681100
Fluegen, G., Avivar-Valderas, A., Wang, Y., Padgen, M. R.,
Williams, J. K., Nobre, A. R., …, Aguirre-Ghiso, J. A. (2017).
Phenotypic heterogeneity of disseminated tumour cells is preset
by primary tumour hypoxic microenvironments. Nature Cell
Biology, 19(2), 120–132. https://doi.org/10.1038/ncb3465
Donato, C., Kunz, L., Castro-Giner, F., Paasinen-Sohns, A.,
Strittmatter, K., Szczerba, B. M., … Aceto, N. (2020). Hypoxia
triggers the intravasation of clustered circulating tumor cells.
Cell Reports, 32(10), 108105. https://doi.org/10.1016/j.celrep.
2020.108105
Cancer and Metastasis Reviews
24. Pantel, K., & Alix-Panabières, C. (2022). Crucial roles of circulating tumor cells in the metastatic cascade and tumor immune
escape: Biology and clinical translation. Journal for ImmunoTherapy of Cancer, 10(12), e005615. https://doi.org/10.1136/
jitc-2022-005615
25. Perea Paizal, J., Au, S. H., & Bakal, C. (2021). Squeezing
through the microcirculation: Survival adaptations of circulating tumour cells to seed metastasis. British Journal of Cancer,
124(1), 58–65. https://doi.org/10.1038/s41416-020-01176-x
26. Sauve, D., Friesen, V. L., & Charmantier, A. (2021). The effects
of weather on avian growth and implications for adaptation to
climate change. Frontiers in Ecology and Evolution, 9, 569741.
https://doi.org/10.3389/fevo.2021.569741
27. Jie, X.-X., Zhang, X.-Y., & Xu, C.-J. (2017). Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: Mechanisms and clinical applications. Oncotarget, 8(46),
81558–81571. https://doi.org/10.18632/oncotarget.18277
28. Nelson, C. M., Khauv, D., Bissell, M. J., & Radisky, D. C.
(2008). Change in cell shape is required for matrix metalloproteinase-induced epithelial-mesenchymal transition of mammary
epithelial cells. Journal of Cellular Biochemistry, 105(1), 25–33.
https://doi.org/10.1002/jcb.21821
29. Zeisberg, M., & Neilson, E. G. (2009). Biomarkers for epithelial-mesenchymal transitions. Journal of Clinical Investigation,
119(6), 1429–1437. https://doi.org/10.1172/JCI36183
30. Cao, Z., Livas, T., & Kyprianou, N. (2016). Anoikis and EMT:
Lethal “Liaisons” during cancer progression. Critical Reviews in
Oncogenesis, 21(3–4), 155–168. https://doi.org/10.1615/CritR
evOncog.2016016955
31. Kuburich, N. A., den Hollander, P., Pietz, J. T., & Mani, S. A.
(2022). Vimentin and cytokeratin: Good alone, bad together.
Seminars in Cancer Biology, 86, 816–826. https://doi.org/10.
1016/j.semcancer.2021.12.006
32. Song, K.-A., & Faber, A. C. (2019). Epithelial-to-mesenchymal
transition and drug resistance: Transitioning away from death.
Journal of Thoracic Disease, 11(6), E82–E85. https://doi.org/10.
21037/jtd.2019.06.11
33. Huang, Y., Hong, W., & Wei, X. (2022). The molecular mechanisms and therapeutic strategies of EMT in tumor progression
and metastasis. Journal of Hematology & Oncology, 15(1), 129.
https://doi.org/10.1186/s13045-022-01347-8
34. Wang, X., Zhao, S., Wang, Z., & Gao, T. (2022). Platelets
involved tumor cell EMT during circulation: Communications
and interventions. Cell Communication and Signaling, 20(1),
82. https://doi.org/10.1186/s12964-022-00887-3
35. Raimondi, C., Gradilone, A., Naso, G., Vincenzi, B., Petracca,
A., Nicolazzo, C., & Gazzaniga, P. (2011). Epithelial-mesenchymal transition and stemness features in circulating tumor
cells from breast cancer patients. Breast Cancer Research
and Treatment, 130(2), 449–455. https:// doi. org/ 10. 1007/
s10549-011-1373-x
36. De Las Rivas, J., Brozovic, A., Izraely, S., Casas-Pais, A., Witz,
I. P., & Figueroa, A. (2021). Cancer drug resistance induced by
EMT: Novel therapeutic strategies. Archives of Toxicology, 95(7),
2279–2297. https://doi.org/10.1007/s00204-021-03063-7
37. Yu, M., Bardia, A., Wittner, B. S., Stott, S. L., Smas, M. E.,
Ting, D. T., et al. (2013). Circulating breast tumor cells exhibit
dynamic changes in epithelial and mesenchymal composition.
Science, 339(6119), 580–584. https://doi.org/10.1126/science.
1228522
38. Raimondi, C., Carpino, G., Nicolazzo, C., Gradilone, A., Gianni,
W., Gelibter, A., et al. (2017). PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung
cancer patients: A molecular shield to evade immune system
? OncoImmunology, 6(12), e1315488. https://doi.org/10.1080/
2162402X.2017.1315488
39. Mego, M., Mani, S. A., Lee, B.-N., Li, C., Evans, K. W., Cohen,
E. N., et al. (2012). Expression of epithelial-mesenchymal transition-inducing transcription factors in primary breast cancer: The
effect of neoadjuvant therapy. International Journal of Cancer,
130(4), 808–816. https://doi.org/10.1002/ijc.26037
40. Oliveras-Ferraros, C., Corominas-Faja, B., Cufí, S., VazquezMartin, A., Martin-Castillo, B., Iglesias, J. M., & Menendez,
J. A. (2012). Epithelial-to-mesenchymal transition (EMT) confers primary resistance to trastuzumab (Herceptin). Cell Cycle,
11(21), 4020–4032. https://doi.org/10.4161/cc.22225
41. Zhu, X., Chen, L., Liu, L., & Niu, X. (2019). EMT-mediated
acquired EGFR-TKI resistance in NSCLC: Mechanisms and
strategies. Frontiers in Oncology, 9, 1044. https://doi.org/10.
3389/fonc.2019.01044
42. Saitoh, M. (2018). Involvement of partial EMT in cancer progression. The Journal of Biochemistry, 164(4), 257–264. https://doi.
org/10.1093/jb/mvy047
43. Lüönd, F., Sugiyama, N., Bill, R., Bornes, L., Hager, C., Tang, F.,
Santacroce, N., Beisel, C., Ivanek, R., Bürglin, T., Tiede, S., van
Rheenen, J., & Christofori, G. (2021). Distinct contributions of
partial and full EMT to breast cancer malignancy. Developmental
Cell, 56(23), 3203–3221.e11. https://doi.org/10.1016/j.devcel.
2021.11.006
44. Brabletz, S., Schuhwerk, H., Brabletz, T., & Stemmler, M. P.
(2021). Dynamic EMT: A multi-tool for tumor progression. The
EMBO Journal, 40(18). https://doi.org/10.15252/embj.20211
08647
45. Huang, R. Y.-J., Wong, M. K., Tan, T. Z., Kuay, K. T., Ng, A. H.
C., Chung, V. Y., Chu, Y.-S., Matsumura, N., Lai, H.-C., Lee, Y.
F., Sim, W.-J., Chai, C., Pietschmann, E., Mori, S., Low, J. J. H.,
Choolani, M., & Thiery, J. P. (2013). An EMT spectrum defines
an anoikis-resistant and spheroidogenic intermediate mesenchymal state that is sensitive to e-cadherin restoration by a src-kinase
inhibitor, saracatinib (AZD0530). Cell Death & Disease, 4(11),
e915–e915. https://doi.org/10.1038/cddis.2013.442
46. Paoli, P., Giannoni, E., & Chiarugi, P. (2013). Anoikis molecular pathways and its role in cancer progression. Biochimica et
Biophysica Acta (BBA) – Molecular Cell Research, 1833(12),
3481–3498. https://doi.org/10.1016/j.bbamcr.2013.06.026
47. de Sousa Mesquita, A. P., de Araújo Lopes, S., Pernambuco
Filho, P. C. A., Nader, H. B., & Lopes, C. C. (2017). Acquisition
of anoikis resistance promotes alterations in the Ras/ERK and
PI3K/Akt signaling pathways and matrix remodeling in endothelial cells. Apoptosis, 22(9), 1116–1137. https://doi.org/10.1007/
s10495-017-1392-0
48. Pereira-Veiga, T., Schneegans, S., Pantel, K., & Wikman, H.
(2022). Circulating tumor cell-blood cell crosstalk: Biology and
clinical relevance. Cell Reports, 40(9), 111298. https://doi.org/
10.1016/j.celrep.2022.111298
49. Lou, X.-L., Sun, J., Gong, S.-Q., Yu, X.-F., Gong, R., & Deng, H.
(2015). Interaction between circulating cancer cells and platelets:
Clinical implication. Chinese Journal of Cancer Research, 27(5).
50. Ward, M. P., Kane, E., Norris, L. A., Mohamed, B. M., Kelly,
T., Bates, M., Clarke, A., Brady, N., Martin, C. M., Brooks,
R. D., Brooks, D. A., Selemidis, S., Hanniffy, S., Dixon, E.
P., & O’Toole, S. A. (2021). Platelets, immune cells and the
coagulation cascade; Friend or foe of the circulating tumour
cell? Molecular Cancer, 20(1), 59. https:// doi. org/ 10. 1186/
s12943-021-01347-1
51. Szczerba, B. M., Castro-Giner, F., Vetter, M., Krol, I., Gkountela, S.,
Landin, J., Scheidmann, M. C., Donato, C., Scherrer, R., Singer,
J., Beisel, C., Kurzeder, C., Heinzelmann-Schwarz, V., Rochlitz,
C., Weber, W. P., Beerenwinkel, N., & Aceto, N. (2019). Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature, 566(7745), 553–557. https://doi.org/10.1038/
s41586-019-0915-y
13
Cancer and Metastasis Reviews
52. Eales, K. L., Hollinshead, K. E. R., & Tennant, D. A. (2016).
Hypoxia and metabolic adaptation of cancer cells. Oncogenesis,
5(1), e190–e190. https://doi.org/10.1038/oncsis.2015.50
53. Gorr, T. A., Wichmann, D., Hu, J., Hermes-Lima, M., Welker,
A. F., Terwilliger, N., Wren, J. F., Viney, M., Morris, S., Nilsson,
G. E., Deten, A., Soliz, J., & Gassmann, M. (2010). Hypoxia
tolerance in animals: Biology and application. Physiological and
Biochemical Zoology, 83(5), 733–752. https://doi.org/10.1086/
648581
54. Pamenter, M. E. (2022). Adaptations to a hypoxic lifestyle in
naked mole-rats. Journal of Experimental Biology, 225(4),
jeb196725. https://doi.org/10.1242/jeb.196725
55. Muz, B., de la Puente, P., Azab, F., & Azab, A. K. (2015). The
role of hypoxia in cancer progression, angiogenesis, metastasis,
and resistance to therapy. Hypoxia, 83. https://doi.org/10.2147/
HP.S93413
56. Infantino, V., Santarsiero, A., Convertini, P., Todisco, S., &
Iacobazzi, V. (2021). Cancer cell metabolism in hypoxia: Role
of HIF-1 as key regulator and therapeutic target. International
Journal of Molecular Sciences, 22(11), 5703. https://doi.org/10.
3390/ijms22115703
57. Ke, Q., & Costa, M. (2006). Hypoxia-inducible factor-1 (HIF-1).
Molecular Pharmacology, 70(5), 1469–1480. https://doi.org/10.
1124/mol.106.027029
58. Kierans, S. J., & Taylor, C. T. (2021). Regulation of glycolysis
by the hypoxia-inducible factor (HIF): Implications for cellular
physiology. The Journal of Physiology, 599(1), 23–37. https://
doi.org/10.1113/JP280572
59. Wei, X., Hou, Y., Long, M., Jiang, L., & Du, Y. (2022). Molecular mechanisms underlying the role of hypoxia-inducible factor-1 α in metabolic reprogramming in renal fibrosis. Frontiers in
Endocrinology, 13, 927329. https://doi.org/10.3389/fendo.2022.
927329
60. Pezzuto, A., & Carico, E. (2018). Role of HIF-1 in cancer progression: Novel insights. A Review. Current Molecular Medicine, 18(6), 343–351. https://doi.org/10.2174/156652401866618
1109121849
61. Rashid, M., Zadeh, L. R., Baradaran, B., Molavi, O., Ghesmati,
Z., Sabzichi, M., & Ramezani, F. (2021). Up-down regulation of
HIF-1α in cancer progression. Gene, 798, 145796. https://doi.
org/10.1016/j.gene.2021.145796
62. Wu, Q., You, L., Nepovimova, E., Heger, Z., Wu, W., Kuca, K., &
Adam, V. (2022). Hypoxia-inducible factors: Master regulators of
hypoxic tumor immune escape. Journal of Hematology & Oncology, 15(1), 77. https://doi.org/10.1186/s13045-022-01292-6
63. Zhang, W., Shi, X., Peng, Y., Wu, M., Zhang, P., Xie, R., Wu, Y.,
Yan, Q., Liu, S., & Wang, J. (2015). HIF-1α promotes epithelialmesenchymal transition and metastasis through direct regulation of ZEB1 in colorectal cancer. PLoS One, 10(6), e0129603.
https://doi.org/10.1371/journal.pone.0129603
64. Tam, S. Y., Wu, V. W. C., & Law, H. K. W. (2020). Hypoxiainduced epithelial-mesenchymal transition in cancers: HIF-1α
and beyond. Frontiers in Oncology, 10, 486. https://doi.org/10.
3389/fonc.2020.00486
65. Ueda, S., Saeki, T., Osaki, A., Yamane, T., & Kuji, I. (2017).
Bevacizumab induces acute hypoxia and cancer progression in
patients with refractory breast cancer: Multimodal functional
imaging and multiplex cytokine analysis. Clinical Cancer
Research, 23(19), 5769–5778. https:// doi. org/ 10. 1158/ 10780432.CCR-17-0874
66. Paredes, F., Williams, H. C., & San Martin, A. (2021). Metabolic
adaptation in hypoxia and cancer. Cancer letters, 502, 133–142.
https://doi.org/10.1016/j.canlet.2020.12.020
67. Noman, M. Z., Messai, Y., Muret, J., Hasmim, M., & Chouaib,
S. (2014). Crosstalk between CTC, immune system and hypoxic
13
68.
69.
70.
71.
72.
73.
74.
75.
76.
77.
78.
79.
80.
tumor microenvironment. Cancer Microenvironment, 7(3), 153–
160. https://doi.org/10.1007/s12307-014-0157-3
Wang, W.-C., Zhang, X.-F., Peng, J., Li, X.-F., Wang, A.-L., Bie,
Y.-Q., Shi, L.-H., Lin, M.-B., & Zhang, X.-F. (2018). Survival
mechanisms and influence factors of circulating tumor cells.
BioMed Research International, 2018, 1–9. https://doi.org/10.
1155/2018/6304701
Godet, I., Shin, Y. J., Ju, J. A., Ye, I. C., Wang, G., & Gilkes,
D. M. (2019). Fate-mapping post-hypoxic tumor cells reveals
a ROS-resistant phenotype that promotes metastasis. Nature
Communications, 10(1), 4862. https:// doi. org/ 10. 1038/
s41467-019-12412-1
Kallergi, G., Markomanolaki, H., Giannoukaraki, V., Papadaki,
M. A., Strati, A., Lianidou, E. S., Georgoulias, V., Mavroudis,
D., & Agelaki, S. (2009). Hypoxia-inducible factor-1α and vascular endothelial growth factor expression in circulating tumor
cells of breast cancer patients. Breast Cancer Research, 11(6),
R84. https://doi.org/10.1186/bcr2452
Imamura, T., Kikuchi, H., Herraiz, M.-T., Park, D.-Y., Mizukami,
Y., Mino-Kenduson, M., Lynch, M. P., Rueda, B. R., Benita, Y.,
Xavier, R. J., & Chung, D. C. (2009). HIF-1α and HIF-2α have
divergent roles in colon cancer. International Journal of Cancer,
124(4), 763–771. https://doi.org/10.1002/ijc.24032
Lee, P., Chandel, N. S., & Simon, M. C. (2020). Cellular adaptation to hypoxia through hypoxia inducible factors and beyond.
Nature Reviews Molecular Cell Biology, 21(5), 268–283. https://
doi.org/10.1038/s41580-020-0227-y
Chouaib, S., Noman, M. Z., Kosmatopoulos, K., & Curran, M.
A. (2017). Hypoxic stress: Obstacles and opportunities for innovative immunotherapy of cancer. Oncogene, 36(4), 439–445.
https://doi.org/10.1038/onc.2016.225
Rivera, A., Fu, X., Tao, L., & Zhang, X. (2015). Expression of
mouse CD47 on human cancer cells profoundly increases tumor
metastasis in murine models. BMC Cancer, 15(1), 964. https://
doi.org/10.1186/s12885-015-1980-8
Latorre, F., Deutschmann, I. M., Labarre, A., Obiol, A., Krabberød, A. K., Pelletier, E., et al. (2021). Niche adaptation promoted the evolutionary diversification of tiny ocean predators.
Proceedings of the National Academy of Sciences, 118(25),
e2020955118. https://doi.org/10.1073/pnas.2020955118
Wang, H., Pan, J., Barsky, L., Jacob, J. C., Zheng, Y., Gao, C.,
Wang, S., Zhu, W., Sun, H., Lu, L., Jia, H., Zhao, Y., Bruns,
C., Vago, R., Dong, Q., & Qin, L. (2021). Characteristics of
pre-metastatic niche: The landscape of molecular and cellular
pathways. Molecular Biomedicine, 2(1), 3. https://doi.org/10.
1186/s43556-020-00022-z
Liu, Z., Kong, Y., Dang, Q., Weng, S., Zheng, Y., Ren, Y., Lv,
J., Li, N., Han, Y., & Han, X. (2022). Liquid biopsy in pre-metastatic niche: From molecular mechanism to clinical application.
Frontiers in immunology, 13, 958360. https://doi.org/10.3389/
fimmu.2022.958360
Eslami-S, Z., Cortés-Hernández, L. E., Thomas, F., Pantel, K.,
& Alix-Panabières, C. (2022). Functional analysis of circulating tumour cells: The KEY to understand the biology of the
metastatic cascade. British Journal of Cancer, 127(5), 800–810.
https://doi.org/10.1038/s41416-022-01819-1
Raskov, H., Orhan, A., Salanti, A., & Gögenur, I. (2020). Premetastatic niches, exosomes and circulating tumor cells: Early
mechanisms of tumor dissemination and the relation to surgery.
International Journal of Cancer, 146(12), 3244–3255. https://
doi.org/10.1002/ijc.32820
Tang, Y., Lu, Y., Chen, Y., Luo, L., Cai, L., Peng, B., Huang,
W., Liao, H., Zhao, L., & Pan, M. (2019). Pre-metastatic niche
triggers SDF-1/CXCR4 axis and promotes organ colonisation
by hepatocellular circulating tumour cells via downregulation
Cancer and Metastasis Reviews
81.
82.
83.
84.
85.
86.
87.
88.
89.
90.
91.
92.
93.
of Prrx1. Journal of Experimental & Clinical Cancer Research,
38(1), 473. https://doi.org/10.1186/s13046-019-1475-6
Aramini, B., Masciale, V., Arienti, C., Dominici, M., Stella, F.,
Martinelli, G., & Fabbri, F. (2022). Cancer stem cells (CSCs),
circulating tumor cells (CTCs) and their interplay with cancer
associated fibroblasts (CAFs): A new world of targets and treatments. Cancers, 14(10), 2408. https:// doi. org/ 10. 3390/ cance
rs14102408
Papaccio, F. (2020). Circulating cancer stem cells: An interesting
niche to explore. Exploration of Targeted Anti-tumor Therapy,
1(4), 253–258. https://doi.org/10.37349/etat.2020.00016
Wang, S.-S., Jiang, J., Liang, X.-H., & Tang, Y.-L. (2015). Links
between cancer stem cells and epithelial-mesenchymal transition.
OncoTargets and therapy, 8, 2973–2980. https://doi.org/10.2147/
OTT.S91863
Phi, L. T. H., Sari, I. N., Yang, Y.-G., Lee, S.-H., Jun, N., Kim, K.
S., Lee, Y. K., & Kwon, H. Y. (2018). Cancer stem cells (CSCs)
in drug resistance and their therapeutic implications in cancer
treatment. Stem Cells International, 2018, 1–16. https://doi.org/
10.1155/2018/5416923
Papadaki, M. A., Stoupis, G., Theodoropoulos, P. A., Mavroudis,
D., Georgoulias, V., & Agelaki, S. (2019). Circulating tumor
cells with stemness and epithelial-to-mesenchymal transition
features are chemoresistant and predictive of poor outcome in
metastatic breast cancer. Molecular Cancer Therapeutics, 18(2),
437–447. https://doi.org/10.1158/1535-7163.MCT-18-0584
Reddy, K. B. (2020). Stem cells: Current status and therapeutic
implications. Genes, 11(11), 1372. https://doi.org/10.3390/genes
11111372
Yang, M.-H., Imrali, A., & Heeschen, C. (2015). Circulating
cancer stem cells: The importance to select. Chinese Journal of
Cancer Research, 27(5).
Aktas, B., Tewes, M., Fehm, T., Hauch, S., Kimmig, R., &
Kasimir-Bauer, S. (2009). Stem cell and epithelial-mesenchymal
transition markers are frequently overexpressed in circulating
tumor cells of metastatic breast cancer patients. Breast Cancer
Research, 11(4), R46. https://doi.org/10.1186/bcr2333
Gradilone, A., Naso, G., Raimondi, C., Cortesi, E., Gandini,
O., Vincenzi, B., Saltarelli, R., Chiapparino, E., Spremberg,
F., Cristofanilli, M., Frati, L., Aglianò, A. M., & Gazzaniga,
P. (2011). Circulating tumor cells (CTCs) in metastatic breast
cancer (MBC): Prognosis, drug resistance and phenotypic characterization. Annals of Oncology, 22(1), 86–92. https://doi.org/
10.1093/annonc/mdq323
Todaro, M., Gaggianesi, M., Catalano, V., Benfante, A., Iovino,
F., Biffoni, M., Apuzzo, T., Sperduti, I., Volpe, S., Cocorullo, G.,
Gulotta, G., Dieli, F., De Maria, R., & Stassi, G. (2014). CD44v6
is a marker of constitutive and reprogrammed cancer stem cells
driving colon cancer metastasis. Cell Stem Cell, 14(3), 342–356.
https://doi.org/10.1016/j.stem.2014.01.009
Ma, L., Dong, L., & Chang, P. (2019). CD44v6 engages in
colorectal cancer progression. Cell Death & Disease, 10(1), 30.
https://doi.org/10.1038/s41419-018-1265-7
Dinger, T. F., Chen, O., Dittfeld, C., Hetze, L., Hüther, M., Wondrak,
M., Löck, S., Eicheler, W., Breier, G., & Kunz-Schughart, L. A.
(2020). Microenvironmentally-driven plasticity of CD44 isoform
expression determines engraftment and stem-like phenotype in CRC
cell lines. Theranostics, 10(17), 7599–7621. https://doi.org/10.7150/
thno.39893
Ghatak, S., Hascall, V. C., Karamanos, N., Markwald, R. R., &
Misra, S. (2022). Chemotherapy induces feedback up-regulation
of CD44v6 in colorectal cancer initiating cells through β-catenin/
MDR1 signaling to sustain chemoresistance. Frontiers in Oncology, 12, 906260. https://doi.org/10.3389/fonc.2022.906260
94. Nicolazzo, C., Loreni, F., Caponnetto, S., Magri, V., Vestri,
A. R., Zamarchi, R., et al. (2020). Baseline CD44v6-positive
circulating tumor cells to predict first-line treatment failure in
patients with metastatic colorectal cancer. Oncotarget, 11(45),
4115–4122. https://doi.org/10.18632/oncotarget.27794
95. Dayem, A. A., Choi, H.-Y., Kim, J.-H., & Cho, S.-G. (2010).
Role of oxidative stress in stem, cancer, and cancer stem cells.
Cancers, 2(2), 859–884. https://doi.org/10.3390/cancers2020859
96. Curti, L., & Campaner, S. (2021). MYC-induced replicative
stress: A double-edged sword for cancer development and treatment. International Journal of Molecular Sciences, 22(12).
https://doi.org/10.3390/ijms22126168
97. Meškytė, E. M., Keskas, S., & Ciribilli, Y. (2020). MYC as a
multifaceted regulator of tumor microenvironment leading to
metastasis. International Journal of Molecular Sciences, 21(20).
https://doi.org/10.3390/ijms21207710
98. Hurtado, P., Martínez-Pena, I., & Piñeiro, R. (2020). Dangerous
liaisons: Circulating tumor cells (CTCs) and cancer-associated
fibroblasts (CAFs). Cancers, 12(10). https:// doi. org/ 10. 3390/
cancers12102861
99. Miller, P., Sharma, U., Medina-Saenz, K., Yeasky, T., PiconRuiz, M., Morata-Tarifa, C., Seagroves, T., Slingerland, J., Lippman, M., & El-Ashry, D. (2018). Abstract P2-01-10: Circulating CAF/CTC complexes and breast cancer metastasis. Cancer
Research, 78(4_Supplement), P2-01-10. https://doi.org/10.1158/
1538-7445.SABCS17-P2-01-10
100. Chen, P., Hsu, W.-H., Han, J., Xia, Y., & DePinho, R. A. (2021).
Cancer stemness meets immunity: From mechanism to therapy.
Cell Reports, 34(1), 108597. https://doi.org/10.1016/j.celrep.
2020.108597
101. Pirri, F., Ometto, L., Fuselli, S., Fernandes, F. A. N., Ancona,
L., Perta, N., Di Marino, D., Le Bohec, C., Zane, L., & Trucchi,
E. (2022). Selection-driven adaptation to the extreme Antarctic
environment in the Emperor penguin. Heredity, 129(6), 317–326.
https://doi.org/10.1038/s41437-022-00564-8
102. Amintas, S., Bedel, A., Moreau-Gaudry, F., Boutin, J., Buscail,
L., Merlio, J.-P., Vendrely, V., Dabernat, S., & Buscail, E. (2020).
Circulating tumor cell clusters: United we stand divided we fall.
International Journal of Molecular Sciences, 21(7). https://doi.
org/10.3390/ijms21072653
103. Fabisiewicz, A., & Grzybowska, E. (2017). CTC clusters in cancer progression and metastasis. Medical Oncology, 34(1), 12.
https://doi.org/10.1007/s12032-016-0875-0
104. Yang, Y., Zheng, H., Zhan, Y., & Fan, S. (2019). An emerging
tumor invasion mechanism about the collective cell migration.
American Journal of Translational Research, 11(9), 5301–5312.
105. Smietanka, U., Szostakowska-Rodzos, M., Tabor, S., Fabisiewicz, A., & Grzybowska, E. A. (2021). Clusters, assemblies and
aggregates of tumor cells in the blood of breast cancer patients;
Composition, mode of action, detection and impact on metastasis
and survival. International Journal of Translational Medicine,
1(1), 55–68. https://doi.org/10.3390/ijtm1010005
106. Aceto, N., Bardia, A., Miyamoto, D. T., Donaldson, M. C.,
Wittner, B. S., Spencer, J. A., Yu, M., Pely, A., Engstrom, A.,
Zhu, H., Brannigan, B. W., Kapur, R., Stott, S. L., Shioda, T.,
Ramaswamy, S., Ting, D. T., Lin, C. P., Toner, M., Haber, D. .
A., & Maheswaran, S. (2014). Circulating tumor cell clusters are
oligoclonal precursors of breast cancer metastasis. Cell, 158(5),
1110–1122. https://doi.org/10.1016/j.cell.2014.07.013
107. Castro-Giner, F., & Aceto, N. (2020). Tracking cancer progression: From circulating tumor cells to metastasis. Genome Medicine, 12(1), 31. https://doi.org/10.1186/s13073-020-00728-3
108. Schuster, E., Taftaf, R., Reduzzi, C., Albert, M. K., RomeroCalvo, I., & Liu, H. (2021). Better together: Circulating tumor
13
Cancer and Metastasis Reviews
109.
110.
111.
112.
113.
114.
115.
116.
117.
118.
cell clustering in metastatic cancer. Trends in Cancer, 7(11),
1020–1032. https://doi.org/10.1016/j.trecan.2021.07.001
Hong, Y., Fang, F., & Zhang, Q. (2016). Circulating tumor cell
clusters: What we know and what we expect (Review). International Journal of Oncology, 49(6), 2206–2216. https://doi.org/
10.3892/ijo.2016.3747
Kapeleris, J., Zou, H., Qi, Y., Gu, Y., Li, J., Schoning, J.,
Monteiro, M. J., & Gu, W. (2020). Cancer stemness contributes to cluster formation of colon cancer cells and high metastatic potentials. Clinical and Experimental Pharmacology
& Physiology, 47(5), 838–847. https://doi.org/10.1111/14401681.13247
Liu, X., Taftaf, R., Kawaguchi, M., Chang, Y.-F., Chen, W.,
Entenberg, D., Zhang, Y., Gerratana, L., Huang, S., Patel, D. B.,
Tsui, E., Adorno-Cruz, V., Chirieleison, S. M., Cao, Y., Harney,
A. S., Patel, S., Patsialou, A., Shen, Y., Avril, S., et al. (2019).
Homophilic CD44 interactions mediate tumor cell aggregation
and polyclonal metastasis in patient-derived breast cancer models. Cancer Discovery, 9(1), 96–113. https://doi.org/10.1158/
2159-8290.CD-18-0065
Genna, A., Vanwynsberghe, A. M., Villard, A. V., Pottier, C.,
Ancel, J., Polette, M., & Gilles, C. (2020). EMT-associated heterogeneity in circulating tumor cells: Sticky friends on the road
to metastasis. Cancers, 12(6). https://doi.org/10.3390/cancers120
61632
Giuliano, M., Shaikh, A., Lo, H. C., Arpino, G., De Placido,
S., Zhang, X. H., Cristofanilli, M., Schiff, R., & Trivedi, M. V.
(2018). Perspective on circulating tumor cell clusters: Why it
takes a village to metastasize. Cancer Research, 78(4), 845–852.
https://doi.org/10.1158/0008-5472.CAN-17-2748
Lee, M., Kim, E. J., Cho, Y., Kim, S., Chung, H. H., Park, N.
H., & Song, Y.-S. (2017). Predictive value of circulating tumor
cells (CTCs) captured by microfluidic device in patients with epithelial ovarian cancer. Gynecologic Oncology, 145(2), 361–365.
https://doi.org/10.1016/j.ygyno.2017.02.042
Housman, G., Byler, S., Heerboth, S., Lapinska, K., Longacre,
M., Snyder, N., & Sarkar, S. (2004). Drug resistance in cancer:
An overview. Cancers, 6, 1769–1792. https://doi.org/10.3390/
cancers6031769
Bithi, S. S., & Vanapalli, S. A. (2017). Microfluidic cell isolation
technology for drug testing of single tumor cells and their clusters.
Scientific Reports, 7, 41707. https://doi.org/10.1038/srep41707
Dolfus, C., Piton, N., Toure, E., & Sabourin, J.-C. (2015). Circulating tumor cell isolation: The assets of filtration methods with
polycarbonate track-etched filters. Chinese Journal of Cancer
Research = Chung-kuo yen cheng yen chiu, 27(5), 479–487.
https://doi.org/10.3978/j.issn.1000-9604.2015.09.01
Francescangeli, F., Magri, V., De Angelis, M. L., De Renzi, G.,
Gandini, O., Zeuner, A., Paola Gazzaniga Nicolazzo, C. (2021).
Sequential isolation and characterization of single CTCs and
large CTC clusters in metastatic colorectal cancer patients. Cancers, 13(24). https://doi.org/10.3390/cancers13246362
13
119. Emsley, J. (2020). Elements of chemistry: The periodic table: Its
story and its significance (p. 472). Oxford University Press.
120. Keller, L., & Pantel, K. (2019). Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nature
Reviews Cancer, 19(10), 553–567. https:// doi. org/ 10. 1038/
s41568-019-0180-2
121. Campbell, D. R., Galen, C., & Wu, C. A. (2005). Ecophysiology
of first and second generation hybrids in a natural plant hybrid
zone. Oecologia, 144, 214–225 http:// www. jstor. org/ stable/
20062322
122. Edelman, N. B., & Mallet, J. (2021). Prevalence and adaptive
impact of introgression. Annual Review of Genetics, 55, 265–
283. https://doi.org/10.1146/annurev-genet-021821-020805
123. Francescangeli, F., De Angelis, M. L., Rossi, R., Cuccu, A.,
Giuliani, A., De Maria, R., & Zeuner, A. (2023). Dormancy,
stemness, and therapy resistance: Interconnected players in
cancer evolution. Cancer Metastasis Reviews, 42(1), 197–215.
https://doi.org/10.1007/s10555-023-10092-4
124. Ye, J. C., Horne, S., Zhang, J. Z., Jackson, L., Heng, H., & H.
(2021). Therapy induced genome chaos: A novel mechanism of
rapid cancer drug resistance. Frontiers in Cell and Developmental Biology, 9, 676344. https://doi.org/10.3389/fcell.2021.676344
125. Zhou, X., Zhou, M., Zheng, M., Tian, S., Yang, X., Ning, Y., Li,
Y., & Zhang, S. (2022). Polyploid giant cancer cells and cancer
progression. Frontiers in Cell and Developmental Biology, 10,
1017588. https://doi.org/10.3389/fcell.2022.1017588
126. Reduzzi, C., Vismara, M., Gerratana, L., Silvestri, M., De Braud,
F., Raspagliesi, F., Verzoni, E., Di Cosimo, S., Locati, L. D.,
Cristofanilli, M., Daidone, M. G., & Cappelletti, V. (2020). The
curious phenomenon of dual-positive circulating cells: Longtime overlooked tumor cells. Seminars in Cancer Biology, 60,
344–350. https://doi.org/10.1016/j.semcancer.2019.10.008
127. Reduzzi, C., Gerratana, L., Zhang, Y., D'Amico, P., Shah, A. N.,
Davis, A. A., Manai, M., Silvestri, M., Zhang, Q., Donahue, J.,
& Cristofanilli, M. (2022). CK+/CD45+ (dual-positive) circulating cells are associated with prognosis in patients with advanced
breast cancer. Journal of Clinical Oncology, 40(16_suppl),
1093–1093.
128. Laberge, G. S., Duvall, E., Haedicke, K., & Pawelek, J. (2019).
Leukocyte-cancer cell fusion-Genesis of a deadly journey. Cells,
8(2), 170. https://doi.org/10.3390/cells8020170
129. Frost, J. (2020). Cancer’s intelligence. International Journal of
Unconventional Computing, 16, 41–78.
130. de Magalhães, J. P. (2022). Every gene can (and possibly will)
be associated with cancer. Trends in Genetics, 38(3), 216–217.
https://doi.org/10.1016/j.tig.2021.09.005
Publisher’s note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.