ArticlePDF AvailableLiterature Review

Abstract and Figures

Cancer and its morbidities, such as cancer cachexia, constitute a major public health problem. Although cancer cachexia has afflicted humanity for centuries, its underlying multifactorial and complex physiopathology mitigates the understanding of its mechanistic aspects. During the last decades we have witnessed a dramatic increase in the knowledge of cancer cachexia pathophysiology. Anorexia, muscle and adipose tissue wasting are the main features of cancer cachexia. These, apparently, independent symptoms have humoral factors secreted by the tumor as a common cause. Importantly, the hypothalamus has emerged as an organ that senses the peripheral signals emanated from the tumoral environment, and not only elicits anorexia but also contributes to the development of muscle and adipose tissue loss. Herein, we review the roles of factors secreted by the tumor and its effects on the hypothalamus, muscle and adipose tissue, as well as underscore key targets that are being exploited for cancer cachexia treatment.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
Molecular and neuroendocrine
mechanisms of cancer cachexia
Maria Carolina S Mendes, Gustavo D Pimentel, Felipe O Costa and Jose
´
B C Carvalheira
Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP),
MA: 13083-970 Campinas, Sao Paulo, Brazil
Correspondence
should be addressed
to J B C Carvalheira
Email
carvalheirajbc@uol.com.br
Abstract
Cancer and its morbidities, such as cancer cachexia, constitute a major public health problem.
Although cancer cachexia has afflicted humanity for centuries, its underlying multifactorial
and complex physiopathology has hindered the understanding of its mechanism. During the
last few decades we have witnessed a dramatic increase in the understanding of cancer
cachexia pathophysiology. Anorexia and muscle and adipose tissue wasting are the main
features of cancer cachexia. These apparently independent symptoms have humoral factors
secreted by the tumor as a common cause. Importantly, the hypothalamus has emerged as an
organ that senses the peripheral signals emanating from the tumoral environment, and not
only elicits anorexia but also contributes to the development of muscle and adipose tissue
loss. Herein, we review the roles of factors secreted by the tumor and its effects on the
hypothalamus, muscle and adipose tissue, as well as highlighting the key targets that are
being exploited for cancer cachexia treatment.
Key Words
" hypothalamus
" cancer
" muscle
" neuropeptides
" neuroendocrinology
Journal of Endocrinology
(2015) 226 , R29–R43
Introduction
The earliest report of significant weight loss dates back
to Hippocrates’ School of Medicine (about 460–377 BC).
Since that era, this syndrome has been recognized as a
condition associated with poor prognosis, justifying the
name cachexia, from the Greek kakos (i.e., bad) and hexis
(i.e., condition or appearance), or ‘bad condition’. It is
associated with many chronic or end-stage diseases such as
cancer, cardiac, respiratory, renal or hepatic failure and
infectious diseases, as well as aging (Doehner & Anker
2002). During human history, weight loss has always
been recognized as a marker in the perception of
control and damage in relation to health and disease.
Notably, a fit appearance is associated with willpower and
self-discipline, whilst the perception of potential harm
and loss of control is related to changing body states, such
as the development of obesity and especially cachexia
(Chamberlain 2004).
Patients’ and their families’ perception of muscle
wasting makes the disease visible and represents an
indication that death is closer (Hopkinson et al. 2006).
As cachexia goes on, wasting of skeletal muscle tissue
diminishes mobility and lethargy impairs concentration,
leading patients towards isolation and depression (Wata-
nabe & Bruera 1996, Stewart et al. 2006). Importantly,
cachexia not only affects the patient, but also their families,
caregivers, and healthcare professionals, who often experi-
ence emotions of fright and hopelessness as they try to
palliate symptoms by feeding the patients (Reid et al. 2009).
The emotional distress experienced by healthcare pro-
fessionals and nihilism regarding the effectiveness of
cachexia treatment frequently block conversation about
weight loss, which makes even the discussion of cachexia a
taboo (Booth et al. 1996, Parle et al. 1997, Churm et al.
2009). In this review, we will highlight the mechanistic
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R29–R43
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
foundation of cancer cachexia, the knowledge of which
has started to change the current nihilistic therapeutic
approach to this devastating condition.
Cancer cachexia
Cancer cachexia is defined as a multifactorial syndrome,
characterized by anorexia as well as diminished body
weight, loss of skeletal muscle, and atrophy of adipose
tissue (Fearon et al. 2011). Specifically, weight loss of
more than 5% in previously healthy individuals or more
than 2% in subjects with depletion of current body
weight (BMI less than 20 kg/m
2
) or in individuals with
reduced appendicular muscle index (males less than
7.26 kg/m
2
and females less than 5.45 kg/m
2
) constitute
the diagnosis of cancer cachexia (Fearon et al. 2011).
Recently, it has been recognized that weight loss alone is
insufficient to express the complexity of cachexia, and two
other clinical characteristics have been incorporated into
its definition: It cannot be fully reversed by conventional
nutritional support and it leads to functional impairment
(Muscaritoli et al. 2010, Fearon et al. 2011). Its incidence
varies according to tumor type, from 31% in patients with
good-risk non-Hodgkin’s lymphoma to 87% in those
with gastric cancer in some series (Dewys et al. 1980,
Teunissen et al. 2007). Importantly, since cachexia is
accompanied by the incapacity for improvement of
nutritional status through supplements, it leads to frailty
and ultimately accounts for approximately 20% of cancer
deaths (Dewys et al. 1980, Ross et al. 2004, Bachmann et al.
2008, Fearon et al. 2011, 2013). The cachexia-mediated
increased mortality is probably due to lower response to
chemotherapy and worse toxicity in anti-cancer
treatment, besides higher susceptibility to infections and
other clinical complications (Costa & Donaldson 1979,
Andreyev et al.1998, Nitenberg & Raynard 2000,
Arrieta et al. 2010).
It is well known that anorexia alone is not able to
cause cachexia. This is one of the main characteristics
that distinguishes cachexia from starvation. In the
former, both adipose tissue and skeletal muscle mass are
depleted, while muscle mass is preserved during starvation
(Fearon 2011). It is noteworthy that starvation in
cancer patients, may be associated with upper digestive
obstruction or fistula, particularly in head and neck,
esophageal, gastric and pancreatic cancer patients, or
peritoneal carcinomatosis-induced multi-level abdominal
obstruction (Dechaphunkul et al. 2013). However, the
great majority of advanced-cancer patients, mainly those
with lung, hepatic or bone metastasis and lung, cervical or
head and neck primary cancers, present a hypermetabolic
state that is characteristic of cachexia.
The physiopathology of cancer cachexia remains
unclear. Several cancer-related metabolic pathways induce
weight loss, muscle and adipose tissue wasting, anorexia,
anemia, and asthenia. The apparent causes of these
symptoms are energy imbalance (increased energy expen-
diture rate), negative protein balance (increased proteol-
ysis and decreased protein synthesis), and increased
lipolysis. Mechanistically, several factors such as increased
levels of hormones, cytokines and factors secreted by the
tumor as well as deregulation of control by the hypo-
thalamus of energy expenditure and hunger/satiety
promote cancer cachexia (Fig. 1).
In fact, cancer cachexia is characterized by maladap-
tive maintenance of inflammation. In contrast, acute
activation of the immune system in response to tissue
stress or infection serves as an adaptive response that is
essential to host survival (Ramos et al.2004). These
responses include fever, headache, changes in the sleep–
wake cycle, anorexia, fatigue, and nausea referred to as
‘sickness behavior’ (Hart 1988, Elmquist et al. 1997). The
organismal advantages of these actions are demonstrated
by their wide expression among vertebrates and also
partial expression in some invertebrates (Kluger 1991).
For instance, force-feeding acutely infected animals is
associated with higher mortality, signifying short-term
anorexia as a host defense mechanism in infection and
tissue injury (Murray & Murray 1979). Additionally,
somnolence and fatigue diminish energy expenditure
during periods of caloric intake restriction (Hart 1988,
Saper & Breder 1992, 1994).
Molecular mechanisms of skeletal muscle
wasting
Cachexia-induced muscle atrophy occurs as a result of
both reduced protein synthesis at initiation and elonga-
tion steps and increased protein degradation. Muscle
wasting is the main cause of poor prognosis and low
quality of life. Skeletal muscle protein degradation
is promoted by ubiquitin–proteasome and autophagy–
lysosomal pathways, as well as the calcium-dependent
enzymes (calpains), which can be activated by the
proteolysis-inducing factor (PIF), myostatin, activin A
(ActA), and cytokines (Matzuk et al. 1994, Tisdale 2009,
Zhou et al. 2010, Johns et al. 2013).
PIF, a glycoprotein first isolated from the MAC16
tumor, has been detected in the urine of cancer patients
with cachexia (Todorov et al. 1996, Cariuk et al. 1997).
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R30
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
Specifically, patients bearing a vast range of cancers, such
as pancreatic, breast, ovary, lung, and colon and rectum,
present increased circulating levels of PIF (Cariuk et al.
1997). Importantly, the isolation of this protein and
subsequent injection into mice induced severe and
prompt body-weight loss (Tisdale 2003). In striking
contrast, it has been reported that PIF is not related to
either survival or muscle wasting in patients with
advanced cancers (Wieland et al. 2007). Mechanistically,
PIF not only promotes protein degradation by increasing
mRNA levels of ubiquitin-carrier protein and proteasome
subunits (Tisdale 2003), but also inhibits protein synthesis
through activation of the RNA-dependent protein kinase
(PKR) (Eley & Tisdale 2007). The latter effect is dependent
on eukaryotic initiation factor 2 alpha-subunit (eIF2a)
phosphorylation, which suppresses protein synthesis by
the eIF2 complex (Eley & Tisdale 2007, Eley et al. 2010).
Interestingly, PKR also induces muscle protein
degradation by activating the transcription factor nuclear
factor kB(NF-kB). Nuclear accumulation of NF-kB
increases the expression of the muscle-specific ubiquitin
E3 ligases, and RING-finger protein 1 (MuRF1) as well as
some proteasome subunits upregulating the ubiquitin–
proteasome proteolytic mechanism and therefore
promoting skeletal muscle breakdown (Bodine et al.
2001, Argile
´
s et al. 2014). PIF also induces transitory
formation of reactive oxygen species (ROS) through
activation of NADPH oxidase by protein kinase C
(Fan et al. 1990, Smith et al. 2004). Since ROS induce
NF-kB nuclear translocation (Schreck et al. 1991), this
pathway also contributes to increasing the expression of
MuRF1 in skeletal muscle (Li et al. 2003, Cai et al. 2004,
Yu et al. 2008).
Myostatin and activins are members of the transform-
ing growth factor B family, which promote muscle wasting
in certain models of cachexia, including cancer cachexia
(Carlson et al. 1999, Ma et al. 2003, Zhou et al
. 2010, Chen
et al. 2014). Transgenic mice that lack myostatin, as well as
cattle with mutations that reduce the expression of
myostatin, show an increased muscle mass phenotype
(McPherron & Lee 1997, McPherron et al. 1997), whilst
recombinant viral overexpression of activins results in
muscle wasting and fibrosis (Chen et al . 2014). Myostatin
and activins share the same receptor, activin type 2
Figure 1
Tumor-secreted factors promote central- and peripheral-mediated cancer
cachexia. Tumor growth results in the secretion of pro-inflammatory
factors that promote cachexia by signaling anorexia, muscle wasting, and
white adipose tissue (WAT) atrophy. In par ticular, treatment with ghrelin
and parathyroid hormone-related protein (PTHrP) alleviates anorexia in
the hypothalamus. Tumors also secrete both the proteolysis-inducing factor
(PIF) and activin, whi ch leads to skeletal muscle degradation and
sarcopenia. Tumor-secreted zinc-alpha2-glycoprotein (ZAG) induces
lipid oxidation and WAT loss. IFN, interferon; IL, interleukin;
TNF, tumor necrosis factor.
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R31
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
receptor B (ActR2B), whose antagonism potently reverses
cancer-induced cachexia (Xia & Schneyer 2009, Zhou et al.
2010). Interestingly, circulating serum levels of ActA,
which has been demonstrated to be secreted by cancer
cells, are elevated in cancer cachectic patients (Zhou et al.
2010, Loumaye et al. 2015). Mechanistically, myostatin
and activins trigger skeletal muscle protein breakdown
by upregulating MuRF1 and MAFbx/Atrogin1, as well as
decreasing protein synthesis via inhibition of the Akt/
mTOR pathway (Chen et al. 2014, Gallot et al. 2014).
Activation of this pathway inhibits the activity of the
transcriptional factor Forkhead box O (FoxO), which
is a major regulator of MuRF1 and MAFbx/Atrogin1
expression. Accordingly, the use of a RNA oligonucleotide
to block FoxO1 or dominant-negative FoxO3 attenuates
loss of skeletal muscle mass in a model of cachexia by
suppressing MAFbx/Atrogin1 transcription (Sandri et al.
2004, 2006).
Increasing evidence indicates that cytokines play a
pivotal role in promoting skeletal muscle atrophy. It is
well established that tumor necrosis factor (TNF) is a key
cytokine that induces skeletal muscle wasting. For
instance, CHO cells that overexpress TNF promote
muscle wasting in mice (Oliff et al. 1987, Acharyya et al.
2004). In contrast, inhibition of TNF with a chimeric TNF
receptor prevented muscle wasting in mice bearing a
TNF-producing tumor (Teng et al. 1993). More recently,
TNF-induced atrophy was demonstrated to be mediated
by the induction of MAFbx/Atrogin1 in muscle by the
attenuation of FoxO activation (Wang et al. 2014) as well
as by increasing MuRF1 (Sishi & Engelbrecht 2011). TNF
also suppresses the PI3K/Akt pathway (Sishi & Engelbrecht
2011). Interestingly, inhibitor of nuclear factor kappa B
kinase subunit beta (IKKb) conditional knockout mice
present hyperphosphorylation of Akt. Conversely, Akt
inhibition leads to muscle atrophy, indicating the
existence of crosstalk between the IKKb/NF-kBand
PI3K/Akt pathways, which control muscle degradation
(Mourkioti et al. 2006). Recently, a new member of the
TNF superfamily has been described, TNF-like weak
inducer of apoptosis (TWEAK), which promotes cachexia
by a mechanism similar to that of TNF, i.e., by activating
NF-kB and promoting augmented expression of MuRF1,
which targets components of the thick filaments (Dogra
et al. 2007, Mittal et al. 2010, Kumar et al. 2012).
Increasing levels of interleukin 6 (IL6) also correlate
with development of cachexia. Accordingly, treatment
with an IL6 receptor antagonist, or MABs to murine IL6,
was able to suppress key cachexia parameters (Strassmann
et al. 1992, Enomoto et al. 2004, Zaki et al. 2004).
However, IL6 alone is not enough to promote cachexia
syndrome (Soda et al. 1994, 1995). Interestingly, increased
IL6 levels are correlated with poor prognosis in patients
with advanced cancer (Suh et al. 2013), and are associated
with increased weight loss, morbidity, and mortality in
patients with lung cancer (Bayliss et al. 2011). Despite the
absence of solid results in cancer cachectic patients,
interferon gamma MAB reversed wasting syndrome in a
cachexia animal model, indicating a role for this cytokine
in cachexia syndrome (Matthys et al. 1991).
Molecular mechanisms of adipose tissue loss
Although the mechanisms behind muscle wasting have
been extensively studied, much less is known about factors
that promote adipose tissue loss in cancer cachectic
patients. The fact that viable cancer cells do not induce
weight loss, particularly adipose tissue wasting, indicates
that tumor-secreted factors could be the cause of fat
atrophy (Costa & Holland 1962). The search for these
factors led to the discovery of a lipid-mobilizing factor,
which was purified from the urine of cachectic individuals
(Masuno et al. 1981, 1984, Taylor et al. 1992, McDevitt
et al. 1995).
Over the last decade, zinc-alpha2-glycoprotein (ZAG)
has been characterized as an adipokine, which induces
lipid mobilization and is upregulated in cancer cachexia
(Bing et al. 2004, 2010, Bao et al. 2005). Mechanistically,
the lipolytic effect of ZAG is mediated by activation of
B3-adrenoceptors (Russell et al. 2002), which, through
AMPc pathway activation in a GTP-dependent manner,
leads to hormone sensitive lipase (HSL) activation and
glycerol release (Hirai et al. 1998). Accordingly, both
genetically-obese (ob/ob) mice and outbred NMRI mice
treated with human ZAG display decreased body weight,
with pronounced carcass fat loss, without change in
body water or nonfat mass, and neither changes in
food nor water intake (Hirai et al. 1998, Russell et al.
2004). Moreover, mice bearing xenografts of a tumor cell
line that overexpress ZAG display dramatic weight loss
(Hale 2002). ZAG also induces lipid utilization, increasing
fat oxidation (Russell & Tisdale 2002, 2010), due to
upregulation of mitochondrial uncoupling protein 1
(UCP1) mRNA in brown adipose tissue (BAT) (Bing et al.
2002, Russell et al . 2004), mediated by ZAG binding and
activation of B3-adrenoreceptor in adipocytes (Russell
et al. 2002).
In addition to tumor-derived ZAG effects, inflam-
matory mediators, such as TNF, modulate white adipose
tissue (WAT) homeostasis. Importantly, TNF inhibits
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R32
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
lipoprotein lipase activity (Price et al. 1986), and increases
HSL mRNA expression (Tisdale 2004, Agustsson et al .
2007). Additionally, TNF has been shown to inhibit
glucose transport, by reducing glucose transporter 4
protein and mRNA levels, decreasing substrates for
lipogenesis (Hauner et al. 1995). TNF-induced lipolysis is
mediated by activation of MAPK kinase, ERK and elevation
of intracellular AMPc by decreasing the expression of
cyclic-nucleotide phosphodiesterase 3B (Zhang et al.
2002). MAPK and JNK activation lead to peroxisome
proliferator-activated receptor gamma (PPARY) phos-
phorylation, inhibiting pre-adipocyte differentiation
(Hu et al. 1996). It has also been observed that TNF
decreases the protein levels of perilipins A and B at the
surface of lipid droplets in 3T3L1 adipocytes, inducing
lipolysis. Furthermore, overexpression of perilipins by
adenovirus infection blocks this effect (Souza et al. 1998).
In cancer cachexia, TNF increases monocyte chemoat-
tractant protein 1 expression in adipocytes, attracting
monocytes to the adipose tissue, resulting in inflam-
mation (Machado et al. 2004). The infiltrating macro-
phages are responsible for increasing TNF production,
and also IL6 and IL1 beta, generating a vicious cycle of
macrophage recruitment and cytokine production.
Neuroendocrine regulation of food intake and
anorexia
The hypothalamus is the master key for the control of
energy homeostasis. Importantly, it is in this CNS area that
hundreds of signals converge, including hormones,
nutrients, and cytokines, to integrate the complex
energy expenditure/food intake balance physiology
(Schwartz et al. 2000, Laviano et al. 2008, 2012, Blanco
Martı
´
nez de Morentin et al. 2011, Pimentel et al. 2014).
The hypothalamus is subdivided into functional areas
that fine tune the energy balance by sending signals that
coordinately increase food intake and suppress energy
expenditure or vice versa. Historically, it was loss-of-
function experiments, performed in the 1930’s, that
provided the proof of concept that the CNS is crucial to
the regulation of energy balance. The results of these
initial studies revealed that different cerebral regions could
control energy balance, in particular it was verified that
CNS lesions performed in macaques and cats lead to
deregulation of food intake and loss of thermogenesis
control (Ranson et al. 1938). However, it was only in
the 1950’s that the hypothalamus was established as a
crucial organ for this control. Specifically, lesions in
the ventromedial region of the hypothalamus of rats
induce hyperphagia, while lateral hypothalamus lesions
promote anorexia (Anand & Brobeck 1951, Miller 1957,
Hervey 1959).
The hypothalamus is constituted by neurons that
coordinately secrete anorexigenic (cocaine- and
amphetamine-regulated transcript (CART) and pro-opio-
melanocortin (POMC)) or orexigenic (agouti-related
protein (AgRP) and neuropeptide Y (NPY)) NPs to control
food intake. These NPs are produced mainly in the arcuate
(ARC) nucleus and paraventricular nucleus (PVN), but also
in the ventromedial hypothalamus (VMH) (Schwartz et al.
2000, Lage et al. 2008, Pimentel et al. 2013). The VMH
contains neurons that promote increased energy expendi-
ture (Schwartz et al. 2000, Blanco Martı
´
nez de Morentin
et al. 2011, Pimentel et al. 2013, Martı
´
nez et al. 2014).
Consistent with a VMH tonic pro-anorexigenic effect,
VMH-specific injection of colchicine (a neuronal blocker)
into anorectic rats increased food intake (
Varma et al. 2000,
Laviano et al. 2002). Moreover, certain areas of the brain,
such as the nucleus of the solitary tract (NST) have also been
implicated in the control of appetite. Accordingly, there is
an increase in NST neuron c-Fos activity after i.c.v. IL1B
injection (DeBoer et al. 2009).
Several lines of evidence indicate that the melano-
cortin system has a key role in hypothalamus dysfunction
in cancer cachexia. This system is mainly composed
of POMC neurons that secrete aMSH and exert their
anorexigenic effects on neurons that contain the melano-
cortin 4 receptor (MC4R; Balthasar et al. 2005, Cone 2005,
Silva et al.2014). It is noteworthy that mouse
neuronal cells express both POMC and CART in the
same neurons, while CART is not found in perikarya and
axons of human POMC neurons (Menyhe
´
rt et al. 2007).
Interestingly, MC4R-, but not MC3R-knockout mice, are
resistant to cachexia (Marks et al. 2001, 2003). Accor-
dingly, the administration of MC4R antagonists directly
to the hypothalamus ameliorates cancer-associated
and chronic-kidney-disease-associated cachexia and
attenuates the anorexigenic actions of the sphingosine 1
phosphate (Wisse et al. 2001, Markison et al. 2005, Cheung
et al. 2007, DeBoer et al. 2008, Silva et al. 2014).
MC4R is also expressed in orexigenic neurons and
these neurons are inhibited by a MSH decreasing
NPY/AgRP release (Laviano et al. 2008). Injection of a
melanocortin receptor antagonist attenuates radiation-
mediated anorexia and cachexia, when compared with
non-irradiated mice, in an AgRP-dependent manner
(Joppa et al. 2007). Interestingly, treatment with megestrol
acetate (MA), a drug approved by the FDA for cancer
cachexia, alleviates anorexia due to increased
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R33
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
hypothalamic NPY expression (McCarthy et al. 1994),
which is decreased in anorectic cancer patients (Jatoi et al.
2001). Taken together, these ndings indicate that
decreased activity of NPY/AgRP neurons occurs synergis-
tically to the hyperstimulation of POMC neuronal cells
and that the melanocortin system is critical for neuro-
endocrine-axis-mediated cancer cachexia.
In addition to the melanocortin system, other
neuronal circuits have been found to be dysfunctional in
cancer cachexia. Among these, hypothalamic serotoni-
nergic and dopaminergic systems are the most studied.
Consistent with an anorexigenic effect of the serotoniner-
gic system, 5HT1B-receptor is upregulated in PVN and
supraoptic nuclei of tumor-bearing rats (Makarenko et al.
2005) and VMH-specific serotoninergic system blockade
ameliorates appetite in anorexic rats (Laviano et al. 1996).
On the other hand, consistent with a dual effect of the
dopaminergic system in cancer cachexia, VMH-specific
dopamine 1 receptor antagonist leads to decreased
appetite and, in contrast, dopamine 2 receptor antagonist
administration increases food intake in tumor-bearing
rodents (Sato et al. 2001). Much less is known about the
glutamatergic neural circuit in the genesis of cancer
cachexia, but the increased activity of this system is
associated with anorexia. Consistent with this, a reduction
of vagal/glutamatergic neurotransmission with metabo-
tropic glutamate receptor antagonist (I(C)-AP3) alleviates
inflammation-LPS-driven anorexia, cachexia and febrile
states (Weiland et al. 2006).
Cancer cachexia molecular signals that
modulate the hypothalamus
It is beyond the scope of this review to report on the
innumerous signals that control energy homeostasis,
but these associated with cancer cachexia will be covered.
It is well established that pro-inflammatory cytokines
released from tumors promote cancer progression and
anorexia (Laviano et al. 2003, Seruga et al. 2008).
The results of initial studies have revealed that VMH-
specific injection of IL1 receptor antagonist attenuates
anorexia in tumor-bearing rats (Laviano et al. 1995, 2000).
Moreover, s.c. injection of the TNF inhibitor improved
food intake, with increased meal number and size in
anorectic rats (Torelli et al. 1999). Accordingly, tumor-
bearing rodents and cancer patients display higher IL1B
and TNF levels in cerebrospinal fluid (CSF; Opara et al.
1995a,b, Protas et al. 2011).
Mechanistically, cytokines induce anorexia by activat-
ing neuronal cells expressing POMC in the ARC nucleus of
the hypothalamus, which increases the central melanocor-
tin system timbre (Lawrence & Rothwell 2001, Reyes &
Sawchenko 2002, Scarlett et al.2007). Consistent with this
model, the use of a selective antagonist of MC4R was enough
to attenuate the anorexigenic effects of IL1B (Joppa et al.
2005). These data indicate that cytokines are CSF soluble
factors critical to hypothalamus-mediated anorexia.
In addition to pro-inflammatory cytokines, other
molecules have been implicated in cancer cachexia, such as
ghrelin and parathyroid hormone-related protein (PTHrP).
Although cachectic patients present high levels of
circulating ghrelin (Shimizu et al. 2003, Garcia et al. 2005),
treatment with ghrelin (s.c.) improves food consumption
in both rodents (DeBoer et al. 2007, Lage et al. 2008,
Fujitsuka et al. 2011) and cancer patients (Neary et al. 2004).
These findings indicate that hyperghrelinemia is a com-
pensatory mechanism that fails to overcome the cancer-
cachexia-induced decreased ghrelin signaling in the hypo-
thalamus (
Fujitsuka et al. 2011). The orexigenic ghrelin
effects are mediated by the hypothalamus, where this
hormone suppresses the expression of IL1R and POMC, and
increases AgRP and NPY expression (DeBoer et al. 2007).
Ghrelin-mediated attenuation of cachexia is reproduced in
different models, interestingly in fasting, denervation and
chronic-kidney-disease-mediated cachexia, ghrelin treat-
ment attenuated muscle protein degradation due, at least in
part, to the inhibition of actinomyosin cleavage (DeBoer
et al. 2008, Porporato et al. 2013).
The results of recent studies have indicated that
tumors release PTHrP, which not only decreases food
intake but also promotes muscle wasting (Asakawa et al.
2010, Kir et al. 2014). The results of these studies indicate
that blocking PTHrP may be an effective strategy for
treating cancer cachexia. Mechanistically, PTHrP activates
hypothalamic urocortins 2/3 via vagal afferent pathways
and inhibition of gastric emptying (Asakawa et al. 2010).
Importantly, PTPHrP neutralization is enough to suppress
B-adrenergic timbre, which attenuates energy expenditure
and muscle mass loss in anorectic mice (Kir et al. 2014).
Although the intracellular mechanisms that promote
hypothalamic-hormone-mediated anorexia are still
unclear, the activation of hypothalamic AMP-activated
protein kinase (AMPK) is a crucial event. AMPK is a key
mediator of energy balance that modulates food intake
and energy expenditure (Blanco Martı
´
nez de Morentin
et al. 2011, Hardie 2015). The results of recent studies
indicate that AMPK senses a multitude of nutritional and
hormonal signals such as berberine, omega 3 fatty acids,
glucose, alpha lipoic acid and leucine, insulin, leptin,
thyroid hormones, and inflammatory mediators (Kahn
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R34
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
et al. 2005, Ropelle et al. 2007, 2008a,b, Lage et al. 2008,
Steinberg et al. 2009, Lo
´
pez et al. 2010, Pimentel et al. 2013,
Santos et al. 2013, Zhang et al. 2014). Likewise, activation
of AMPK not only blunts cancer-induced reduction of food
intake, but also attenuates inflammation and prolongs the
survival of tumor-bearing rats (Ropelle et al. 2007).
Neuroendocrine regulation of
cachexia-induced thermogenesis and
skeletal muscle sarcopenia
The hypothalamus not only promotes anorexia but also
contributes to the development of other cancer cachexia
symptoms, such as increased thermogenesis and skeletal
muscle sarcopenia (Fig. 2). Interestingly, cancer-associated
cachexia increases energy expenditure, an effect mainly
mediated by the BAT and coordinated by the hypothalamus
(Brooks et al.1981, Bianchi et al.1989, Tsoli et al.2012,
Kir et al. 2014). This organ senses the increased
levels of TNF, the tyrotropin-releasing hormone, and
the corticotropin-releasing hormone to promote heat
production via a B3-adrenergic neuronal circuit (Arruda
et al.2011).
Recently, cachexia has been found to be associated
with the conversion of white adipose cells into beige
cells, a process described as ‘browning’ (Kir et al. 2014,
Nedergaard & Cannon 2014, Petruzzelli et al. 2014). Beige
cells display abundant levels of UCP1, which reduces the
mitochondrial electrochemical gradient to promote
thermogenesis. Mechanistically, it has been suggested
that cancer cachexia-induced browning is also mediated
by an increase in B-adrenergic tonus (Cao et al. 2011, Kir
et al. 2014, Petruzzelli et al. 2014). Unfortunately, it is
not known whether the CNS is implicated in WAT
browning regulation during cancer cachexia. Since several
obesity studies have identified the hypothalamus as an
important regulator of browning (Cao et al . 2011, Baboota
et al. 2014, Beiroa et al. 2014, Owen et al. 2014, Ruan et al.
2014, Dodd et al. 2015), future studies to explore the
role of the hypothalamus in cachexia-induced browning
are encouraged.
Although the influence of the hypothalamus on the
modutation of lean body mass is clear, the mechanisms
are only partially elucidated (Marks et al. 2001, 2003,
Wisse et al. 2001,
Cheung et al. 2008, Braun et al. 2011).
The hypothalamic–pituitary–adrenal axis is an important
Figure 2
The hypothalamus is at the crossroads of cancer cachexia’s main features.
Pro-anorexigenic factors are integrated in discrete nuclei of the
hypothalamus. The ventromedial nucleus (VMH) promotes heat production
in brown adipose tissue (BAT) and may mediate white adipose browning
via the B3 adrenergic system. The paraventricular nucleus (PVN) and
arcuate (ARC) nucleus are the major integrating cente rs of food intake,
modulating the timbre of serotonin (5HT) expression and melanocortin 4
receptor (MC4R) respectively. Interestingly, pro-opiomelanocortin leads to
skeletal muscle break down and sarcopenia. 3V, third ventricle.
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R35
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
axis that links the CNS to the muscle catabolic program.
Interestingly, brain–IL1B injection leads to muscle wasting
and increases in markers of muscle protein breakdown,
such as MURF and Atrogin1. In accordance with the
existance of an adrenal-mediated effect, adrenalectomy
suppressed IL1B-induced muscle atrophy, whilst gluco-
corticoid treatment was enough to promote muscle
atrophy (Braun et al. 2011). Interestingly, in spite of
muscle wasting induced by cancer, uremia, or LPS, as well
as IL1B-induced anorexia is suppressed by MC4R blockade
(Marks et al. 2001, 2003, Wisse et al. 2001, Cheung et al.
2008, Whitaker & Reyes 2008), MC4R-knockout animals
are not saved from body lean mass loss after central
infusion of IL1B (Braun et al. 2011), these findings indicate
that different neuronal circuits are involved in the CNS
modulation of muscle catabolic programs and that the
hypothalamus is crucial for induction and maintenance of
the main symptoms of cancer cachexia.
Treatment of cancer cachexia
Initial efforts
Although a number of nutritional supplements and drugs,
such as Cannabis (Strasser et al. 2006), eicosapentaenoic
acid (Beck et al. 1991, Barber et al. 1999, Mantovani et al.
2008) and branched-chain amino acids (Eley et al. 2007)
have shown promising results in pre-clinical studies, the
results of phase III clinical trials have failed to demonstrate
a substantial effect of these drugs and nutritional
supplements as treatments for cancer cachexia.
Currently, the only FDA-approved drug for the
treatment of cancer cachexia is medroxyprogesterone.
Medroxyprogesterone acetate and MA are both synthetic
progestins currently used to improve appetite and
promote weight gain in cancer cachexia (Tchekmedyian
et al. 1992). In accordance, the results of recent meta-
analysis indicated that MA is associated with a small effect
on weight gain and increase in appetite (Ruiz et al. 2013).
Although the mechanism of action is unknown, these
drugs reduce pro-inflammatory cytokines and increase
NPY levels in the hypothalamus (Mantovani et al. 2001).
Corticosteroids are alternative orexigenic agents for the
treatment of cancer cachexia (Popiela et al. 1989, Shih &
Jackson 2007). Importantly, dexamethasone treatment
resulted in similar-magnitude effects on weight gain and
increased appetite when compared with MA; however,
this approach was associated with an increased drug
discontinuation rate because of increased collateral effects
(Loprinzi et al. 1999).
New perspectives for the treatment of cancer cachexia
Triggered by better knowledge of the molecular
mechanisms of cachexia, we are observing an increasing
number of cancer cachexia clinical trials. One of the most
promising approaches for cancer cachexia is ghrelin
treatment. A proof of concept study of ghrelin infusion
revealed that this resulted in an increase of energy intake
and in pleasantness of the meal in patients with advanced
incurable cancer in a dose-dependent manner (Neary et al.
2004, Strasser et al. 2008, Hiura et al. 2012). More recently,
an oral mimetic of ghrelin (anamorelin) has been tested
and promising results were achieved with 16 cachectic
patients with different types of tumors (Garcia et al. 2013).
Numerous clinical trials to evaluate beneficial effects of
ghrelin and anamorelin in the treatment of cancer
cachexia are active (NCT0933361, NCT00681486,
NCT00225745, and NCT01505764). Although the use of
ghrelin in these patients appears to be safe, more studies
are necessary to confirm its efficacy and safety.
Despite the proven importance of TNF in the
pathogenesis of cancer cachexia, treatment with inflixi-
mab (a MAB to TNF) did not result in improvement in
cachexia cases (Jatoi
et al. 2001, 2010, Wiedenmann et al.
2008). In contrast, cancer cachexia treatment with
thalidomide, a drug with potent anti-inflammatory effects
(Moreira et al. 1993, Fujita et al. 2001, Keifer et al. 2001,
Richardson et al. 2002) presented encouraging preliminary
results (Davis et al. 2012), but we still do not have
sufficient data to recommend this drug in clinical practice
(Reid et al. 2012).
Cancer cachexia promotes insulin resistance, which
not only blunts muscle glucose uptake and liver glucose
production, but also inhibits protein anabolism, contri-
buting to muscle atrophy (Yoshikawa et al. 2001, Winter
et al. 2012). Metformin, the most widely used agent for the
treatment of type 2 diabetes, increases food intake and
prolongs survival in cachectic rats bearing Walker256
tumors (Ropelle et al. 2007). Interestingly, the results of
a clinical trial in individuals with prostate cancer
without cancer cachexia indicated that the association
of metformin, exercise, and low-glycemic-index diet
improved body weight (Nobes et al. 2012). Another insulin
sensitizer, rosiglitazone, a PPAR agonist that improves
insulin sensitivity, prevented weight loss, and helped
avoid muscle protein degradation in an experimental
colon cancer model of cachexia. These effects were
paralleled by a decrease in Atrogin1 and MuRF1 expression
(Asp et al. 2010). Interestingly, emerging evidence has
indicated that insulin resistance-mediated blunted protein
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R36
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
anabolism is not refractory to post-prandial physiological
amino-acid infusion, indicating conventional nutritional
support to be a promising approach for overcoming
anabolic resistance (Winter et al. 2012). As such, insulin
sensitizers are good candidates for the therapeutic
treatment of cancer cachexia, but clinical studies to
confirm experimental data are necessary.
The use of an ActR2B decoy receptor (sActR2B)
prevents muscle wasting and inhibits muscle loss in
different animal models of cachexia (Zhou et al. 2010).
Since the levels of activins are increased in cancer cachectic
patients (Loumaye et al. 2015), a promising approach for
cancer cachexia treatment may be the blockade of ActR2B.
Conclusion
Although cancer cachexia has been a major burden on
our society for centuries, it is only in recent decades that
there has been unprecedented progress in the under-
standing of its molecular basis. A broad concept that
has emerged is that the hypothalamus is a key center for
the control of anorexia and fat loss in cancer cachexia.
Additionally, the results of animal studies have revealed
numerous factors produced by the tumor that act in
muscle, promoting its wasting. Although the potential
therapeutic implications have not yet been fully exploited
in humans, this collective work has already demonstrated
that targeting the hypothalamus and tumor-secreted
factors are attractive therapeutic approaches for alleviating
cancer cachexia.
Declaration of interest
The authors declare that there is no conflict of interest that could be
perceived as prejudicing the impartiality of this review.
Funding
J B C C was supported by grants from the Conselho Nacional de
Dese nvolvimento Cientı
´
fico e Tecnolo
´
gico (CNPq; 306821/2010-9) and
the Fundac¸a
˜
o de Amparo a
`
Pesquisa de Sa
˜
o Paulo (2013/07607-8). G D P
was supported by grants from the Fundac¸a
˜
o de Amparo a
`
Pesquisa de
Sa
˜
o Paulo (2014/22347-5).
Author contribution statement
M C S M, G D P, and F O C wrote the initial drafts of the manuscript and
J B C C revised the manuscript.
Acknowledgements
We thank Nicola Conran for reviewing the English.
References
Acharyya S, Ladner KJ, Nelsen LL, Damrauer J, Reiser PJ, Swoap S &
Guttridge DC 2004 Cancer cachexia is regulated by selective targeting
of skeletal muscle gene products. Journal of Clinical Investigation 114
370–378. (doi:10.1172/JCI200420174)
Agustsson T, Ryde
´
n M, Hoffstedt J, van Harmelen V, Dicker A,
Laurencikiene J, Isaksson B, Permert J & Arner P 2007 Mechanism of
increased lipolysis in cancer cachexia. Cancer Research 67 5531–5537.
(doi:10.1158/0008-5472.CAN-06-4585)
Anand BK & Brobeck JR 1951 Hypothalamic control of food intake in rats
and cats. Yale Journal of Biology and Medicine 24 123–140.
Andreyev HJ, Norman AR, Oates J & Cunningham D 1998 Why do patients
with weight loss have a worse outcome when undergoing chemo-
therapy for gastrointestinal malignancies? European Journal of Cancer 34
503–509. (doi:10.1016/S0959-8049(97)10090-9)
Argile
´
s JM, Busquets S, Stemmler B & Lo
´
pez-Soriano FJ 2014 Cancer
cachexia: understanding the molecular basis. Nature Reviews. Cancer 14
754–762. (doi:10.1038/nrc3829)
Arrieta O, Michel Ortega RM, Villanueva-Rodrı
´
guez G, Serna-Thome
´
MG,
Flores-Estrada D, Diaz-Romero C, Rodrı
´
guez CM, Martı
´
nez L & Sa
´
nchez-
Lara K 2010 Association of nutritional status and serum albumin levels
with development of toxicity in patients with advanced non-small cell
lung cancer treated with paclitaxel–cisplatin chemotherapy: a
prospective study. BMC Cancer 10 50. (doi:10.1186/1471-2407-10-50)
Arruda AP, Milanski M, Coope A, Torsoni AS, Ropelle E, Carvalho DP,
Carvalheira JB & Velloso LA 2011 Low-grade hypothalamic inflam-
mation leads to defective thermogenesis, insulin resistance, and
impaired insulin secretion. Endocrinology 152 1314–1326. (doi:10.1210/
en.2010-0659)
Asakawa A, Fujimiya M, Niijima A, Fujino K, Kodama N, Sato Y, Kato I,
Nanba H, Laviano A, Meguid MM et al. 2010 Parathyroid hormone-
related protein has an anorexigenic activity via activation of
hypothalamic urocortins 2 and 3. Psychoneuroendocrinology 35
1178–1186. (doi:10.1016/j.psyneuen.2010.02.003)
Asp ML, Tian M, Wendel AA & Belury MA 2010 Evidence for the
contribution of insulin resistance to the development of cachexia in
tumor-bearing mice. International Journal of Cancer 126 756–763.
(doi:10.1002/ijc.24784)
Baboota RK, Murtaza N, Jagtap S, Singh DP, Karmase A, Kaur J, Bhutani KK,
Boparai RK, Premkumar LS, Kondepudi KK et al. 2014 Capsaicin-
induced transcriptional changes in hypothalamus and alterations in
gut microbial count in high fat diet fed mice. Journal of Nutritional
Biochemistry 25 893–902. (doi:10.1016/j.jnutbio.2014.04.004)
Bachmann J, Heiligensetzer M, Krakowski-Roosen H, Bu
¨
chler MW, Friess H
& Martignoni ME 2008 Cachexia worsens prognosis in patients with
resectable pancreatic cancer. Journal of Gastrointestinal Surgery 12
1193–1201. (doi:10.1007/s11605-008-0505-z)
Balthasar N, Dalgaard LT, Lee CE, Yu J, Funahashi H, Williams T, Ferreira M,
Tang V, McGovern RA, Kenny CD et al. 2005 Divergence of
melanocortin pathways in the control of food intake and energy
expenditure. Cell 123 493–505. (doi:10.1016/j.cell.2005.08.035)
Bao Y, Bing C, Hunter L, Jenkins JR, Wabitsch M & Trayhurn P 2005
Zinc-a
2
-glycoprotein, a lipid mobilizing factor, is expressed and
secreted by human (SGBS) adipocytes. FEBS Letters 579 41–47.
(doi:10.1016/j.febslet.2004.11.042)
Barber MD, Ross JA, Voss AC, Tisdale MJ & Fearon KC 1999 The effect of an
oral nutritional supplement enriched with fish oil on weight-loss in
patients with pancreatic cancer. British Journal of Cancer 81 80–86.
(doi:10.1038/sj.bjc.6690654)
Bayliss TJ, Smith JT, Schuster M, Dragnev KH & Rigas JR 2011 A humanized
anti-IL-6 antibody (ALD518) in non-small cell lung cancer. Expert
Opinion on Biological Therapy 11 1663–1668. (doi:10.1517/14712598.
2011.627850)
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R37
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
Beck SA, Smith KL & Tisdale MJ 1991 Anticachectic and antitumor effect of
eicosapentaenoic acid and its effect on protein turnover. Cancer
Research 51 6089–6093.
Beiroa D, Imbernon M, Gallego R, Senra A, Herranz D, VillarroyaF, Serrano M,
Fernø J, Salvador J, Escalada J et al. 2014 GLP-1 agonism stimulates
brown adipose tissue thermogenesis and browning through hypo-
thalamic AMPK. Diabetes 63 3346–3358. (doi:10.2337/db14-0302)
Bianchi A, Bruce J, Cooper AL, Childs C, Kohli M, Morris ID, Morris-Jones P
& Rothwell NJ 1989 Increased brown adipose tissue activity in children
with malignant disease. Hormone and Metabolic Research 21 640–641.
(doi:10.1055/s-2007-1009308)
Bing C, Russell ST, Beckett EE, Collins P, Taylor S, Barraclough R, Tisdale MJ
& Williams G 2002 Expression of uncoupling proteins-1, -2 and -3
mRNA is induced by an adenocarcinoma-derived lipid-mobilizing
factor. British Journal of Cancer 86 612–618. (doi:10.1038/sj.
bjc.6600101)
Bing C, Bao Y, Jenkins J, Sanders P, Manieri M, Cinti S, Tisdale MJ &
Trayhurn P 2004 Zinc-a2-glycoprotein, a lipid mobilizing factor,
is expressed in adipocytes and is up-regulated in mice with
cancer cachexia. PNAS 101 2500–2505. (doi:10.1073/pnas.
0308647100)
Bing C, Mracek T, Gao D & Trayhurn P 2010 Zinc-a2-glycoprotein: an
adipokine modulator of body fat mass? International Journal of Obesity
34 1559–1565. (doi:10.1038/ijo.2010.105)
Blanco Martı
´
nez de Morentin P, Gonza
´
lez CR, Saha AK, Martins L,
Die
´
guez C, Vidal-Puig A, Tena-Sempere M & Lo
´
pez M 2011 Hypo-
thalamic AMP-activated protein kinase as a mediator of whole body
energy balance. Reviews in Endocr ine & Metabolic Disorders 12 127–140.
(doi:10.1007/s11154-011-9165-5)
Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA,
Poueymirou WT, Panaro FJ, Na E, Dharmarajan K et al. 2001
Identification of ubiquitin ligases required for skeletal muscle atrophy.
Science 294 1704–1708. (doi:10.1126/science.1065874)
Booth K, Maguire PM, Butterworth T & Hillier VF 1996 Perceived
professional support and the use of blocking behaviours by hospice
nurses. Journal of Advanced Nursing 24 522–527. (doi:10.1046/j.1365-
2648.1996.22012.x)
Braun TP, Zhu X, Szumowski M, Scott GD, Grossberg AJ, Levasseur PR,
Graham K, Khan S, Damaraju S, Colmers WF et al. 2011 Central nervous
system inflammation induces muscle atrophy via activation of the
hypothalamic–pituitary–adrenal axis. Journal of Experimental Medicine
208 2449–2463. (doi:10.1084/jem.20111020)
Brooks SL, Neville AM, Rothwell NJ, Stock MJ & Wilson S 1981 Sympathetic
activation of brown-adipose-tissue thermogenesis in cachexia.
Bioscience Reports 1 509–517. (doi:10.1007/BF01121584)
Cai D, Frantz JD, Tawa NE, Melendez PA, Oh BC, Lidov HG, Hasselgren PO,
Frontera WR, Lee J, Glass DJ et al. 2004 IKKb/NF-kB activation causes
severe muscle wasting in mice. Cell 119 285–298. (doi:10.1016/j.cell.
2004.09.027)
Cao L, Choi EY, Liu X, Martin A, Wang C, Xu X & During MJ 2011 White to
brown fat phenotypic switch induced by genetic and environmental
activation of a hypothalamic–adipocyte axis. Cell Metabolism 14
324–338. (doi:10.1016/j.cmet.2011.06.020)
Cariuk P, Lorite MJ, Todorov PT, Field WN, Wigmore SJ & Tisdale MJ 1997
Induction of cachexia in mice by a product isolated from the urine
of cachectic cancer patients. British Journal of Cancer 76 606–613.
(doi:10.1038/bjc.1997.433)
Carlson CJ, Booth FW & Gordon SE 1999 Skeletal muscle myostatin mRNA
expression is fiber-type specific and increases during hindlimb
unloading. American Journal of Physiology 277 R601–R606.
Chamberlain K 2004 Food and health: expanding the agenda for health
psychology. Journal of Health Psychology 9 467–481. (doi:10.1177/
1359105304044030)
Chen JL, Walton KL, Winbanks CE, Murphy KT, Thomson RE, Makanji Y,
Qian H, Lynch GS, Harrison CA & Gregorevic P 2014 Elevated
expression of activins promotes muscle wasting and cachexia. FASEB
Journal 28 1711–1723. (doi:10.1096/fj.13-245894)
Cheung WW, Kuo HJ, Markison S, Chen C, Foster AC, Marks DL & Mak RH
2007 Peripheral administration of the melanocortin-4 receptor
antagonist NBI-12i ameliorates uremia-associated cachexia in mice.
Journal of the American Society of Nephrology 18 2517–2524. (doi:10.1681/
ASN.2006091024)
Cheung WW, Rosengren S, Boyle DL & Mak RH 2008 Modulation
of melanocortin signaling ameliorates uremic cachexia. Kidney
International 74 180–186. (doi:10.1038/ki.2008.150)
Churm D, Andrew IM, Holden K, Hildreth AJ & Hawkins C 2009 A
questionnaire study of the approach to the anorexia–cachexia
syndrome in patients with cancer by staff in a district general hospital.
Supportive Care in Cancer 17 503–507. (doi:10.1007/s00520-008-0486-1)
Cone RD 2005 Anatomy and regulation of the central melanocortin
system. Nature Neuroscience 8 571–578. (doi:10.1038/nn1455)
Costa G & Donaldson SS 1979 Current concepts in cancer: effects of cancer
and cancer treatment on the nutrition of the host. New England
Journal of Medicine 300 1471–1474. (doi:10.1056/
NEJM197906283002606
)
Costa G & Holland JF 1962 Effects of Krebs-2 carcinoma on the lipide
metabolism of male Swiss mice. Cancer Research 22 1081–1083.
Davis M, Lasheen W, Walsh D, Mahmoud F, Bicanovsky L & Lagman R
2012 A phase II dose titration study of thalidomide for cancer-
associated anorexia. Journal of Pain and Symptom Management 43 78–86.
(doi:10.1016/j.jpainsymman.2011.03.007)
DeBoer MD, Zhu XX, Levasseur P, Meguid MM, Suzuki S, Inui A, Taylor JE,
Halem HA, Dong JZ, Datta R et al. 2007 Ghrelin treatment causes
increased food intake and retention of lean body mass in a rat model
of cancer cachexia. En docrinology 148 3004–3012. (doi:10.1210/
en.2007-0016)
DeBoer MD, Zhu X, Levasseur PR, Inui A, Hu Z, Han G, Mitch WE, Taylor JE,
Halem HA, Dong JZ et al. 2008 Ghrelin treatment of chronic kidney
disease: improvements in lean body mass and cytokine profile.
Endocrinology 149 827–835. (doi:10.1210/en.2007-1046)
DeBoer MD, Scarlett JM, Levasseur PR, Grant WF & Marks DL 2009
Administration of IL-1b to the 4th ventricle causes anorexia that is
blocked by agouti-related peptide and that coincides with activation of
tyrosine-hydroxylase neurons in the nucleus of the solitary tract.
Peptides 30 210–218. (doi:10.1016/j.peptides.2008.10.019)
Dechaphunkul T, Martin L, Alberda C, Olson K, Baracos V & Gramlich L
2013 Malnutrition assessment in patients with cancers of the head and
neck: a call to action and consensus. Critical Reviews in Oncology/He-
matology 88 459–476. (doi:10.1016/j.critrevonc.2013.06.003)
Dewys WD, Begg C, Lavin PT, Band PR, Bennett JM, Bertino JR, Cohen MH,
Douglass HO, Engstrom PF, Ezdinli EZ et al. 1980 Prognostic effect of
weight loss prior to chemotherapy in cancer patients. Eastern
Cooperative Oncology Group. American Journal of Medicine 69 491–497.
(doi:10.1016/S0149-2918(05)80001-3)
Dodd GT, Decherf S, Loh K, Simonds SE, Wiede F, Balland E, Merry TL,
Mu
¨
nzberg H, Zhang ZY, Kahn BB et al. 2015 Leptin and insulin act on
POMC neurons to promote the browning of white fat. Cell 160 88–104.
(doi:10.1016/j.cell.2014.12.022)
Doehner W & Anker SD 2002 Cardiac cachexia in early literature: a review
of research prior to Medline. International Journal of Cardiology 85 7–14.
(doi:10.1016/S0167-5273(02)00230-9)
Dogra C, Changotra H, Wedhas N, Qin X, Wergedal JE & Kumar A 2007
TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal
muscle-wasting cytokine. FASEB Journal 21 1857–1869. (doi:10.1096/fj.
06-7537com)
Eley HL & Tisdale MJ 2007 Skeletal muscle atrophy, a link between
depression of protein synthesis and increase in degradation.
Journal of
Biological Chemistry 282 7087–7097. (doi:10.1074/jbc.M610378200)
Eley HL, Russell ST & Tisdale MJ 2007 Effect of branched-chain amino acids
on muscle atrophy in cancer cachexia. Biochemical Journal 407
113–120. (doi:10.1042/BJ20070651)
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R38
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
Eley HL, Russell ST & Tisdale MJ 2010 Mechanism of activation of dsRNA-
dependent protein kinase (PKR) in muscle atrophy. Cellular Signalling
22 783–790. (doi:10.1016/j.cellsig.2010.01.002)
Elmquist JK, Scammell TE & Saper CB 1997 Mechanisms of CNS response
to systemic immune challenge: the febrile response. Trends in
Neurosciences 20 565–570. (doi:10.1016/S0166-2236(97)01138-7)
Enomoto A, Rho MC, Fukami A, Hiraku O, Komiyama K & Hayashi M 2004
Suppression of cancer cachexia by 20S,21-epoxy-resibufogenin-3-
acetate–a novel nonpeptide IL-6 receptor antagonist. Biochemical
and Biophysical Research Communications 323 1096–1102. (doi:10.1016/
j.bbrc.2004.08.196)
Fan XT, Huang XP, Da Silva C & Castagna M 1990 Arachidonic acid and
related methyl ester mediate protein kinase C activation in intact
platelets through the arachidonate metabolism pathways. Biochemical
and Biophysical Research Communications 169 933–940. (doi:10.1016/
0006-291X(90)91983-Y)
Fearon KC 2011 Cancer cachexia and fat–muscle physiology. New England
Journal of Medicine 365 565–567. (doi:10.1056/NEJMcibr1106880)
Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, Jatoi A,
Loprinzi C, MacDonald N, Mantovani G et al. 2011 Definition and
classification of cancer cachexia: an international consensus. Lancet.
Oncology 12 489–495. (doi:10.1016/S1470-2045(10)70218-7)
Fearon K, Arends J & Baracos V 2013 Understanding the mechanisms and
treatment options in cancer cachexia. Nature Reviews. Clinical Oncology
10 90–99. (doi:10.1038/nrclinonc.2012.209)
Fujita J, Mestre JR, Zeldis JB, Subbaramaiah K & Dannenberg AJ 2001
Thalidomide and its analogues inhibit lipopolysaccharide-mediated
induction of cyclooxygenase-2. Clinical Cancer Research 7 3349–3355.
Fujitsuka N, Asakawa A, Uezono Y, Minami K, Yamaguchi T, Niijima A,
Yada T, Maejima Y, Sedbazar U, Sakai T et al. 2011 Potentiation of
ghrelin signaling attenuates cancer anorexia–cachexia and prolongs
survival. Translational Psychiatry 1 e23. (doi:10.1038/tp.2011.25)
Gallot YS, Durieux AC, Castells J, Desgeorges MM, Vernus B, Plantureux L,
Re
´
mond D, Jahnke VE, Lefai E, Dardevet D et al. 2014 Myostatin gene
inactivation prevents skeletal muscle wasting in cancer. Cancer Research
74 7344–7356. (doi:10.1158/0008-5472.CAN-14-0057)
Garcia JM, Garcia-Touza M, Hijazi RA, Taffet G, Epner D, Mann D, Smith RG,
Cunningham GR & Marcelli M 2005 Active ghrelin levels and active to
total ghrelin ratio in cancer-induced cachexia. Journal of Clinical
Endocrinology and Metabolism 90 2920–2926. (doi:10.1210/jc.2004-1788)
Garcia JM, Friend J & Allen S 2013 Therapeutic potential of anamorelin, a
novel, oral ghrelin mimetic, in patients with cancer-related cachexia:
a multicenter, randomized, double-blind, crossover, pilot study.
Supportive Care in Cancer 21 129–137. (doi:10.1007/s00520-012-1500-1)
Hale LP 2002 Zinc a-2-glycoprotein regulates melanin production by
normal and malignant melanocytes. Journal of Investigative Dermatology
119 464–470. (doi:10.1046/j.1523-1747.2002.01813.x)
Hardie DG 2015 AMPK: positive and negative regulation, and its role in
whole-body energy homeostasis. Current Opinion in Cell Biology 33 1–7.
(doi:10.1016/j.ceb.2014.09.004)
Hart BL 1988 Biological basis of the behavior of sick animals. Neuroscience
and Biobehavioral Reviews 12 123–137. (doi:10.1016/S0149-
7634(88)80004-6)
Hauner H, Petruschke T, Russ M, Ro
¨
hrig K & Eckel J 1995 Effects of tumour
necrosis factor alpha (TNFa) on glucose transport and lipid metabolism
of newly-differentiated human fat cells in cell culture. Diabetologia 38
764–771. (doi:10.1007/s001250050350)
Hervey GR 1959 The effects of lesions in the hypothalamus in parabiotic
rats. Journal of Physiology 145 336–352. (doi:10.1113/jphysiol.1959.
sp006145)
Hirai K, Hussey HJ, Barber MD, Price SA & Tisdale MJ 1998 Biological
evaluation of a lipid-mobilizing factor isolated from the urine of cancer
patients. Cancer Research 58 2359–2365.
Hiura Y, Takiguchi S, Yamamoto K, Takahashi T, Kurokawa Y, Yamasaki M,
Nakajima K, Miyata H, Fujiwara Y, Mori M et al. 2012 Effects of ghrelin
administration during chemotherapy with advanced esophageal cancer
patients: a prospective, randomized, placebo-controlled phase 2 study.
Cancer 118 4785–4794. (doi:10.1002/cncr.27430)
Hopkinson JB, Wright DN, McDonald JW & Corner JL 2006 The prevalence
of concern about weight loss and change in eating habits in people with
advanced cancer. Journal of Pain and Symptom Management 32 322–331.
(doi:10.1016/j.jpainsymman.2006.05.012)
Hu E, Kim JB, Sarraf P & Spiegelman BM 1996 Inhibition of adipogenesis
through MAP kinase-mediated phosphorylation of PPARg. Science 274
2100–2103. (doi:10.1126/science.274.5295.2100)
Jatoi A, Loprinzi CL, Sloan JA, Klee GG & Windschitl HE 2001
Neuropeptide Y, leptin, and cholecystokinin 8 in patients with
advanced cancer and anorexia: a North Central Cancer Treatment
Group exploratory investigation. Cancer 92 629–633. (doi:10.1002/
1097-0142(20010801)92:3!629::AID-CNCR1363O3.0.CO;2-M)
Jatoi A, Ritter HL, Dueck A, Nguyen PL, Nikcevich DA, Luyun RF, Mattar BI
& Loprinzi CL 2010 A placebo-controlled, double-blind trial of
infliximab for cancer-associated weight loss in elderly and/or poor
performance non-small cell lung cancer patients (N01C9). Lung Cancer
68 234–239. (doi:10.1016/j.lungcan.2009.06.020)
Johns N, Stephens NA & Fearon KC 2013 Muscle wasting in cancer.
International Journal of Biochemistry & Cell Biology 45 2215–2229.
(doi:10.1016/j.biocel.2013.05.032)
Joppa MA, Ling N, Chen C, Gogas KR, Foster AC & Markison S 2005 Central
administration of peptide and small molecule MC4 receptor antagon-
ists induce hyperphagia in mice and attenuate cytokine-induced
anorexia. Peptides 26 2294–2301. (doi:10.1016/j.peptides.2005.03.002)
Joppa MA, Gogas KR, Foster AC & Markison S 2007 Central infusion of the
melanocortin receptor antagonist agouti-related peptide (AgRP(83–
132)) prevents cachexia-related symptoms induced by radiation and
colon-26 tumors in mice. Peptides 28 636–642. (doi:10.1016/j.peptides.
2006.11.021)
Kahn BB, Alquier T, Carling D & Hardie DG 2005 AMP-activated protein
kinase: ancient energy gauge provides clues to modern understanding of
metabolism. Cell Metabolism 1 15–25. (doi:10.1016/j.cmet.2004.12.003)
Keifer JA, Guttridge DC, Ashburner BP & Baldwin AS 2001 Inhibition of NF-
kB activity by thalidomide through suppression of IkB kinase activity.
Journal of Biological Chemistry 276 22382–22387. (doi:10.1074/jbc.
M100938200)
Kir S, White JP, Kleiner S, Kazak L, Cohen P, Baracos VE & Spiegelman BM
2014 Tumour-derived PTH-related protein triggers adipose tissue
browning and cancer cachexia. Nature 513 100–104. (doi:10.1038/
nature13528)
Kluger MJ 1991 Fever: role of pyrogens and cryogens. Physiological Reviews
71 93–127.
Kumar A, Bhatnagar S & Paul PK 2012 TWEAK and TRAF6 regulate skeletal
muscle atrophy. Current Opinion in Clinical Nutrition and Metabolic Care
15 233–239. (doi:10.1097/MCO.0b013e328351c3fc)
Lage R, Die
´
guez C, Vidal-Puig A & Lo
´
pez M 2008 AMPK: a metabolic gauge
regulating whole-body energy homeostasis. Trends in Molecular Medicine
14 539–549. (doi:10.1016/j.molmed.2008.09.007)
Laviano A, Renvyle T, Meguid MM, Yang ZJ, Cangiano C & Rossi Fanelli F
1995 Relationship between interleukin-1 and cancer anorexia. Nutrition
11 680–683.
Laviano A, Cangiano C, Preziosa I, Meguid MM, Muscaritoli M,
Conversano L, Cascino A, Torelli GF, Cherubini S & Rossi Fanelli F 1996
Serotoninergic block in the ventromedial nucleus of hypothalamus
improves food intake in anorectic tumor bearing rats. Advances in
Experimental Medicine and Biology 398 551–553. (doi:10.1007/978-1-
4613-0381-7_88)
Laviano A, Gleason JR, Meguid MM, Yang ZJ, Cangiano C & Rossi Fanelli F
2000 Effects of intra-VMN mianserin and IL-1ra on meal number in
anorectic tumor-bearing rats. Journal of Investigative Medicine
48 40–48.
Laviano A, Meguid MM, Gleason JR & Rossi-Fanelli F 2002 VMN/LHA
functional inhibition in tumor-bearing rats suggests hypothalamic
involvement in cancer anorexia. Nutritional Neuroscience 5 443–456.
(doi:10.1080/1028415021000039202)
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R39
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
Laviano A, Meguid MM & Rossi-Fanelli F 2003 Cancer anorexia: clinical
implications, pathogenesis, and therapeutic strategies. Lancet. Oncology
4 686–694. (doi:10.1016/S1470-2045(03)01247-6)
Laviano A, Inui A, Marks DL, Meguid MM, Pichard C, Rossi Fanelli F &
Seelaender M 2008 Neural control of the anorexia–cachexia syndrome.
American Journal of Physiology. Endocrinology and Metabolism 295
E1000–E1008. (doi:10.1152/ajpendo.90252.2008)
Laviano A, Seelaender M, Rianda S, Silverio R & Rossi Fanelli F 2012
Neuroinflammation: a contributing factor to the pathogenesis of
cancer cachexia. Critical Reviews in Oncogenesis 17 247–251.
(doi:10.1615/CritRevOncog.v17.i3.20)
Lawrence CB & Rothwell NJ 2001 Anorexic but not pyrogenic actions of
interleukin-1 are modulated by central melanocortin-3/4 receptors in
the rat. Journal of Neuroendocrinology 13 490–495. (doi:10.1046/j.1365-
2826.2001.00660.x)
Li YP, Chen Y, Li AS & Reid MB 2003 Hydrogen peroxide stimulates
ubiquitin-conjugating activity and expression of genes for specific E2 and
E3 proteins in skeletal muscle myotubes. American Journal of Physiology.
Cell Physiology 285 C806–C812. (doi:10.1152/ajpcell.00129.2003)
Lo
´
pez M, Varela L, Va
´
zquez MJ, Rodrı
´
guez-Cuenca S, Gonza
´
lez CR,
Velagapudi VR, Morgan DA, Schoenmakers E, Agassandian K, Lage R
et al. 2010 Hypothalamic AMPK and fatty acid metabolism mediate
thyroid regulation of energy balance. Nature Medicine 16 1001–1008.
(doi:10.1038/nm.2207)
Loprinzi CL, Kugler JW, Sloan JA, Mailliard JA, Krook JE, Wilwerding MB,
Rowland KM, Camoriano JK, Novotny PJ & Christensen BJ 1999
Randomized comparison of megestrol acetate versus dexamethasone
versus fluoxymesterone for the treatment of cancer anorexia/cachexia.
Journal of Clinical Oncology 17 3299–3306.
Loumaye A, de Barsy M, Nachit M, Lause P, Frateur L, van Maanen A,
Trefois P, Gruson D & Thissen JP 2015 Role of activin A and myostatin
in human cancer cachexia. Journal of Clinical Endocrinology and
Metabolism 100 2030–2038. (doi:10.1210/jc.2014-4318)
Ma K, Mallidis C, Bhasin S, Mahabadi V, Artaza J, Gonzalez-Cadavid N,
Arias J & Salehian B 2003 Glucocorticoid-induced skeletal muscle
atrophy is associated with upregulation of myostatin gene expression.
American Journal of Physiology. Endocrinology and Metabolism 285
E363–E371. (doi:10.1152/ajpendo.00487.2002)
Machado AP, Costa Rosa LF & Seelaender MC 2004 Adipose tissue in
Walker 256 tumour-induced cachexia: possible association between
decreased leptin concentration and mononuclear cell infiltration. Cell
and Tissue Research 318 503–514. (doi:10.1007/s00441-004-0987-2)
Makarenko IG, Meguid MM, Gatto L, Goncalves CG, Ramos EJ, Chen C &
Ugrumov MV 2005 Hypothalamic 5-HT
1B
-receptor changes in
anorectic tumor bearing rats. Neuroscience Letters 376 71–75.
(doi:10.1016/j.neulet.2004.11.026)
Mantovani G, Maccio
`
A, Massa E & Madeddu C 2001 Managing cancer-
related anorexia/cachexia. Drugs 61 499–514. (doi:10.2165/00003495-
200161040-00004)
Mantovani G, Maccio
`
A, Madeddu C, Gramignano G, Serpe R, Massa E,
Dessı
`
M, Tanca FM, Sanna E, Deiana L et al. 2008 Randomized phase III
clinical trial of five different arms of treatment for patients with cancer
cachexia: interim results. Nutrition 24 305–313. (doi:10.1016/j.nut.
2007.12.010)
Markison S, Foster AC, Chen C, Brookhart GB, Hesse A, Hoare SR, Fleck BA,
Brown BT & Marks DL 2005 The regulation of feeding and metabolic
rate and the prevention of murine cancer cachexia with a
small-molecule melanocortin-4 receptor antagonist. Endocrinology
146 2766–2773. (doi:10.1210/en.2005-0142)
Marks DL, Ling N & Cone RD 2001 Role of the central melanocortin system
in cachexia. Cancer Research 61 1432–1438.
Marks DL, Butler AA, Turner R, Brookhart G & Cone RD 2003 Differential
role of melanocortin receptor subtypes in cachexia. Endocrinology 144
1513–1523. (doi:10.1210/en.2002-221099)
Martı
´
nez D, Pentinat T, Ribo
´
S, Daviaud C, Bloks VW, Cebria
`
J,
Villalmanzo N, Kalko SG, Ramo
´
n-Krauel M, Dı
´
az R et al. 2014 In utero
undernutrition in male mice programs liver lipid metabolism in the
second-generation offspring involving altered Lxra DNA methylation.
Cell Metabolism 19 941–951. (doi:10.1016/j.cmet.2014.03.026)
Masuno H, Yamasaki N & Okuda H 1981 Purification and characterization
of a lipolytic factor (toxohormone-L) from cell-free fluid of ascites
sarcoma 180. Cancer Research 41 284–288.
Masuno H, Yoshimura H, Ogawa N & Okuda H 1984 Isolation of a lipolytic
factor (toxohormone-L) from ascites fluid of patients with hepatoma
and its effect on feeding behavior. European Journal of Cancer & Clinical
Oncology 20 1177–1185. (doi:10.1016/0277-5379(84)90127-5)
Matthys P, Heremans H, Opdenakker G & Billiau A 1991 Anti-interferon-g
antibody treatment, growth of Lewis lung tumours in mice and
tumour-associated cachexia. European Journal of Cancer 27 182–187.
(
doi:10.1016/0277-5379(91)90483-T)
Matzuk MM, Finegold MJ, Mather JP, Krummen L, Lu H & Bradley A 1994
Development of cancer cachexia-like syndrome and adrenal tumors in
inhibin-deficient mice. PNAS 91 8817–8821. (doi:10.1073/pnas.91.19.
8817)
McCarthy HD, Crowder RE, Dryden S & Williams G 1994 Megestrol acetate
stimulates food and water intake in the rat: effects on regional
hypothalamic neuropeptide Y concentrations. European Journal of
Pharmacology 265 99–102. (doi:10.1016/0014-2999(94)90229-1)
McDevitt TM, Todorov PT, Beck SA, Khan SH & Tisdale MJ 1995
Purification and characterization of a lipid-mobilizing factor associated
with cachexia-inducing tumors in mice and humans. Cancer Research
55 1458–1463.
McPherron AC & Lee SJ 1997 Double muscling in cattle due to mutations in
the myostatin gene. PNAS 94 12457–12461. (doi:10.1073/pnas.94.23.
12457)
McPherron AC, Lawler AM & Lee SJ 1997 Regulation of skeletal muscle
mass in mice by a new TGF-b superfamily member. Nature 387 83–90.
(doi:10.1038/387083a0)
Menyhe
´
rt J, Wittmann G, Lechan RM, Keller E, Liposits Z & Fekete C 2007
Cocaine- and amphetamine-regulated transcript (CART) is colocalized
with the orexigenic neuropeptide Y and agouti-related protein and
absent from the anorexigenic a-melanocyte-stimulating hormone
neurons in the infundibular nucleus of the human hypothalamus.
Endocrinology 148 4276–4281. (doi:10.1210/en.2007-0390)
Miller NE 1957 Experiments on motivation. Studies combining
psychological, physiological, and pharmacological techniques.
Science 126 1271–1278. (doi:10.1126/science.126.3286.1271)
Mittal A, Bhatnagar S, Kumar A, Lach-Trifilieff E, Wauters S, Li H,
Makonchuk DY & Glass DJ 2010 The TWEAK–Fn14 system is a critical
regulator of denervation-induced skeletal muscle atrophy in mice.
Journal of Cell Biology 188 833–849. (doi:10.1083/jcb.200909117)
Moreira AL, Sampaio EP, Zmuidzinas A, Frindt P, Smith KA & Kaplan G
1993 Thalidomide exerts its inhibitory action on tumor necrosis factor
a by enhancing mRNA degradation. Journal of Experimental Medicine
177 1675–1680. (doi:10.1084/jem.177.6.1675)
Mourkioti F, Kratsios P, Luedde T, Song YH, Delafontaine P, Adami R,
Parente V, Bottinelli R, Pasparakis M & Rosenthal N 2006 Targeted
ablation of IKK2 improves skeletal muscle strength, maintains mass,
and promotes regeneration. Journal of Clinical Investigation 116
2945–2954. (doi:10.1172/JCI28721)
Murray MJ & Murray AB 1979 Anorexia of infection as a mechanism of host
defense. American Journal of Clinical Nutrition 32 593–596.
Muscaritoli M, Anker SD, Argile
´
s J, Aversa Z, Bauer JM, Biolo G, Boirie Y,
Bosaeus I, Cederholm T, Costelli P et al. 2010 Consensus definition of
sarcopenia, cachexia and pre-cachexia: joint document elaborated by
Special Interest Groups (SIG) “cachexia–anorexia in chronic wasting
diseases” and “nutrition in geriatrics”.
Clinical Nutrition 29 154–159.
(doi:10.1016/j.clnu.2009.12.004)
Neary NM, Small CJ, Wren AM, Lee JL, Druce MR, Palmieri C, Frost GS,
Ghatei MA, Coombes RC & Bloom SR 2004 Ghrelin increases energy
intake in cancer patients with impaired appetite: acute, randomized,
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R40
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
placebo-controlled trial. Journal of Clinical Endocrinology and Metabolism
89 2832–2836. (doi:10.1210/jc.2003-031768)
Nedergaard J & Cannon B 2014 The browning of white adipose tissue: some
burning issues. Cell Metabolism 20 396–407. (doi:10.1016/j.cmet.2014.
07.005)
Nitenberg G & Raynard B 2000 Nutritional support of the cancer patient:
issues and dilemmas. Critical Reviews in Oncology/Hematology 34
137–168. (doi:10.1016/S1040-8428(00)00048-2)
Nobes JP, Langley SE, Klopper T, Russell-Jones D & Laing RW 2012 A
prospective, randomized pilot study evaluating the effects of metfor-
min and lifestyle intervention on patients with prostate cancer
receiving androgen deprivation therapy. BJU International 109
1495–1502. (doi:10.1111/j.1464-410X.2011.10555.x)
Oliff A, Defeo-Jones D, Boyer M, Martinez D, Kiefer D, Vuocolo G, Wolfe A &
Socher SH 1987 Tumors secreting human TNF/cachectin induce cachexia
in mice. Cell 50 555–563. (doi:10.1016/0092-8674(87)90028-6)
Opara EI, Laviano A & Meguid MM 1995a Correlation between food intake
and cerebrospinal fluid interleukin 1a in anorectic tumor-bearing rats.
Nutrition 11 678–679.
Opara EI, Laviano A, Meguid MM & Yang ZJ 1995b Correlation between
food intake and CSF IL-1a in anorectic tumor bearing rats. Neuroreport 6
750–752. (doi:10.1097/00001756-199503270-00011)
Owen BM, Ding X, Morgan DA, Coate KC, Bookout AL, Rahmouni K,
Kliewer SA & Mangelsdorf DJ 2014 FGF21 acts centrally to induce
sympathetic nerve activity, energy expenditure, and weight loss. Cell
Metabolism 20 670–677. (doi:10.1016/j.cmet.2014.07.012)
Parle M, Maguire P & Heaven C 1997 The development of a training model
to improve health professionals’ skills, self-efficacy and outcome
expectancies when communicating with cancer patients. Social Science
& M edicine 44 231–240. (doi:10.1016/S0277-9536(96)00148-7)
Petruzzelli M, Schweiger M, Schreiber R, Campos-Olivas R, Tsoli M, Allen J,
Swarbrick M, Rose-John S, Rincon M, Robertson G et al. 2014 A switch
from white to brown fat increases energy expenditure in cancer-
associated cachexia. Cell Metabolism 20 433–447. (doi:10.1016/j.cmet.
2014.06.011)
Pimentel GD, Ropelle ER, Rocha GZ & Carvalheira JB 2013 The role of
neuronal AMPK as a mediator of nutritional regulation of food intake
and energy homeostasis. Metabolism 62 171–178. (doi:10.1016/j.
metabol.2012.07.001)
Pimentel GD, Ganeshan K & Carvalheira JB 2014 Hypothalamic inflam-
mation and the central nervous system control of energy homeostasis.
Molecular and Cellular Endocrinology 397
15–22. (doi:10.1016/j.mce.
2014.06.005)
Popiela T, Lucchi R & Giongo F 1989 Methylprednisolone as palliative
therapy for female terminal cancer patients. The Methylprednisolone
Female Preterminal Cancer Study Group. European Journal of Cancer &
Clinical Oncology 25 1823–1829. (doi:10.1016/0277-5379(89)90354-4)
Porporato PE, Filigheddu N, Reano S, Ferrara M, Angelino E, Gnocchi VF,
Prodam F, Ronchi G, Fagoonee S, Fornaro M et al. 2013 Acylated and
unacylated ghrelin impair skeletal muscle atrophy in mice. Journal of
Clinical Investigation 123 611–622. (doi:10.1172/JCI39920)
Price SR, Olivecrona T & Pekala PH 1986 Regulation of lipoprotein lipase
synthesis by recombinant tumor necrosis factor – the primary
regulatory role of the hormone in 3T3-L1 adipocytes. Archives of
Biochemistry and Biophysics 251 738–746. (doi:10.1016/0003-
9861(86)90384-X)
Protas PT, Holownia A, Muszynska-Roslan K, Wielgat P, Krawczuk-Rybak M
& Braszko JJ 2011 Cerebrospinal fluid IL-6, TNF-a and MCP-1 in
children with acute lymphoblastic leukaemia during chemotherapy.
Neuropediatrics 42 254–256. (doi:10.1055/s-0031-1295477)
Ramos EJ, Suzuki S, Marks D, Inui A, Asakawa A & Meguid MM 2004 Cancer
anorexia–cachexia syndrome: cytokines and neuropeptides. Current
Opinion in Clinical Nutrition and Metabolic Care 7 427–434. (doi:10.1097/
01.mco.0000134363.53782.cb)
Ranson SW, Fisher C & Ingram WR 1938 Adiposity and diabetes mellitus in
a monkey with hypothalamic lesions. Endocrinology 23 7. (doi:10.1210/
endo-23-2-175)
Reid J, McKenna H, Fitzsimons D & McCance T 2009 Fighting over food:
patient and family understanding of cancer cachexia. Oncology Nursing
Forum 36 439–445. (doi:10.1188/09.ONF.439-445)
Reid J, Mills M, Cantwell M, Cardwell CR, Murray LJ & Donnelly M 2012
Thalidomide for managing cancer cachexia. In Cochrane Database
of Systematic Reviews CD008664. (doi:10.1002/14651858.
CD008664.pub2)
Reyes TM & Sawchenko PE 2002 Involvement of the arcuate nucleus of
the hypothalamus in interleukin-1-induced anorexia. Journal of
Neuroscience 22 5091–5099.
Richardson P, Hideshima T & Anderson K 2002 Thalidomide: emerging
role in cancer medicine. Annual Review of Medicine 53 629–657.
(doi:10.1146/annurev.med.53.082901.104043)
Ropelle ER, Pauli JR, Zecchin KG, Ueno M, de Souza CT, Morari J, Faria MC,
Velloso LA, Saad MJ & Carvalheira JB 2007 A central role for neuronal
adenosine 5
0
-monophosphate-activated protein kinase in cancer-induced
anorexia. Endocrinology 148 5220–5229. (doi:10.1210/en.2007-0381)
Ropelle ER, Fernandes MF, Flores MB, Ueno M, Rocco S, Marin R, Cintra DE,
Velloso LA, Franchini KG, Saad MJ et al. 2008a Central exercise action
increases the AMPK and mTOR response to leptin. PLoS ONE 3 e3856.
(doi:10.1371/journal.pone.0003856)
Ropelle ER, Pauli JR, Fernandes MF, Rocco SA, Marin RM, Morari J,
Souza KK, Dias MM, Gomes-Marcondes MC, Gontijo JA et al. 2008b
A central role for neuronal AMP-activated protein kinase (AMPK) and
mammalian target of rapamycin (mTOR) in high-protein diet-induced
weight loss. Diabetes 57 594–605. (doi:10.2337/db07-0573)
Ross PJ, Ashley S, Norton A, Priest K, Waters JS, Eisen T, Smith IE & O’Brien
ME 2004 Do patients with weight loss have a worse outcome when
undergoing chemotherapy for lung cancers? British Journal of Cancer 90
1905–1911. (doi:10.1038/sj.bjc.6601781)
Ruan HB, Dietrich MO, Liu ZW, Zimmer MR, Li MD, Singh JP, Zhang K,
Yin R, Wu J, Horvath TL et al. 2014 O-GlcNA transferase enables
AgRP neurons to suppress browning of white fat. Cell 159 306–317.
(doi:10.1016/j.cell.2014.09.010)
Ruiz GV, Lo
´
pez-Briz E, Carbonell SR, Gonzalvez PJL & Bort-Marti S 2013
Megestrol acetate for treatment of anorexia–cachexia syndrome.
Cochrane Database of Systematic Revi ews CD004310. (doi:10.1002/
14651858.CD004310.pub3)
Russell ST & Tisdale MJ 2002 Effect of a tumour-derived lipid-mobilising
factor on glucose and lipid metabolism in vivo. British Journal of Cancer
87 580–584. (doi:10.1038/sj.bjc.6600493)
Russell ST & Tisdale MJ 2010 Antidiabetic properties of zinc-a
2
-glyco-
protein in ob/ob mice. Endocrinology 151 948–957. (doi:10.1210/en.
2009-0827)
Russell ST, Hirai K & Tisdale MJ 2002 Role of b3-adrenergic receptors in the
action of a tumour lipid mobilizing factor. British Journal of Cancer 86
424–428. (doi:10.1038/sj.bjc.6600086)
Russell ST, Zimmerman TP, Domin BA & Tisdale MJ 2004 Induction of
lipolysis in vitro and loss of body fat in vivo by zinc-a
2
-glycoprotein.
Biochimica et Biophysica Acta 1636 59–68. (doi:10.1016/j.bbalip.2003.
12.004)
Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K,
Schiaffino S, Lecker SH & Goldberg AL 2004 Foxo transcription factors
induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal
muscle atrophy. Cell 117 399–412. (doi:10.1016/S0092-
8674(04)00400-3)
Sandri M, Lin J, Handschin C, Yang W, Arany ZP, Lecker SH, Goldberg AL &
Spiegelman BM 2006 PGC-1a protects skeletal muscle from atrophy by
suppressing FoxO3 action and atrophy-specific gene transcription.
PNAS 103 16260–16265. (doi:10.1073/pnas.0607795103)
Santos GA, Moura RF, Vitorino DC, Roman EA, Torsoni AS, Velloso LA &
Torsoni MA 2013 Hypothalamic AMPK activation blocks
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R41
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
lipopolysaccharide inhibition of glucose production in mice liver.
Molecular and Cellular Endocrinology 381 88–96. (doi:10.1016/j.mce.
2013.07.018)
Saper CB & Breder CD 1992 Endogenous pyrogens in the CNS: role in
the febrile response. Progress in Brain Research 93 419–428 (discussion
428–429). (doi:10.1016/S0079-6123(08)64587-2)
Saper CB & Breder CD 1994 The neurologic basis of fever. New England
Journal of Medicine 330 1880–1886. (doi:10.1056/
NEJM199406303302609)
Sato T, Meguid MM, Fetissov SO, Chen C & Zhang L 2001 Hypothalamic
dopaminergic receptor expressions in anorexia of tumor-bearing rats.
American Journal of Physiology. Regulatory, Integrative and Comparative
Physiology 281 R1907–R1916.
Scarlett JM, Jobst EE, Enriori PJ, Bowe DD, Batra AK, Grant WF, Cowley MA
& Marks DL 2007 Regulation of central melanocortin signaling
by interleukin-1b. Endocrinology 148 4217–4225. (doi:10.1210/en.
2007-0017)
Schreck R, Rieber P & Baeuerle PA 1991 Reactive oxygen intermediates as
apparently widely used messengers in the activation of the NF-kB
transcription factor and HIV-1. EMBO Journal 10 2247–2258.
Schwartz MW, Woods SC, Porte D, Seeley RJ & Baskin DG 2000
Central nervous system control of food intake. Nature 404 661–671.
(doi:10.1038/35007534)
Seruga B, Zhang H, Bernstein LJ & Tannock IF 2008 Cytokines and their
relationship to the symptoms and outcome of cancer. Nature Reviews.
Cancer 8 887–899. (doi:10.1038/nrc2507)
Shih A & Jackson KC 2007 Role of corticosteroids in palliative care.
Journal of Pain & Palliative Care Pharmacotherapy 21 69–76.
(doi:10.1080/J354v21n04_14)
Shimizu Y, Nagaya N, Isobe T, Imazu M, Okumura H, Hosoda H, Kojima M,
Kangawa K & Kohno N 2003 Increased plasma ghrelin level in lung
cancer cachexia. Clinical Cancer Research 9 774–778.
Silva VR, Micheletti TO, Pimentel GD, Katashima CK, Lenhare L, Morari J,
Mendes MC, Razolli DS, Rocha GZ, de Souza CT et al. 2014
Hypothalamic S1P/S1PR1 axis controls energy homeostasis. Nature
Communications 5 4859. (doi:10.1038/ncomms5859)
Sishi BJ & Engelbrecht AM 2011 Tumor necrosis factor alpha (TNF-a)
inactivates the PI3-kinase/PKB pathway and induces atrophy and
apoptosis in L6 myotubes. Cytokine 54 173–184. (doi:10.1016/j.cyto.
2011.01.009)
Smith HJ, Wyke SM & Tisdale MJ 2004 Role of protein kinase C and NF-k
B
in proteolysis-inducing factor-induced proteasome expression in C
2
C
12
myotubes. British Journal of Cancer 90 1850–1857. (doi:10.1038/sj.bjc.
6601767)
Soda K, Kawakami M, Kashii A & Miyata M 1994 Characterization of mice
bearing subclones of colon 26 adenocarcinoma disqualifies interleukin-
6 as the sole inducer of cachexia. Japanese Journal of Cancer Research 85
1124–1130. (doi:10.1111/j.1349-7006.1994.tb02917.x)
Soda K, Kawakami M, Kashii A & Miyata M 1995 Manifestations of cancer
cachexia induced by colon 26 adenocarcinoma are not fully ascribable
to interleukin-6. International Journal of Cancer 62 332–336.
(doi:10.1002/ijc.2910620317)
Souza SC, de Vargas LM, Yamamoto MT, Lien P, Franciosa MD, Moss LG &
Greenberg AS 1998 Overexpression of perilipin A and B blocks the
ability of tumor necrosis factor a to increase lipolysis in 3T3-L1
adipocytes. Journal of Biological Chemistry 273 24665–24669.
(doi:10.1074/jbc.273.38.24665)
Steinberg GR, Watt MJ & Febbraio MA 2009 Cytokine regulation of AMPK
signalling. Frontiers in Bioscience 14 1902–1916. (doi:10.2741/3350)
Stewart GD, Skipworth RJ & Fearon KC 2006 Cancer cachexia and fatigue.
Clinical Medicine 6 140–143. (doi:10.7861/clinmedicine.6-2-140)
Strasser F, Luftner D, Possinger K, Ernst G, Ruhstaller T, Meissner W, Ko YD,
Schnelle M, Reif M, Cerny T et al. 2006 Comparison of orally
administered Cannabis extract and delta-9-tetrahydrocannabinol in
treating patients with cancer-related anorexia–cachexia syndrome: a
multicenter, phase III, randomized, double-blind, placebo-controlled
clinical trial from the Cannabis-In-Cachexia-Study-Group. Journal of
Clinical Oncology 24 3394–3400. (doi:10.1200/JCO.2005.05.1847)
Strasser F, Lutz TA, Maeder MT, Thuerlimann B, Bueche D, Tscho
¨
pM,
Kaufmann K, Holst B, Bra
¨
ndle M, von Moos R et al. 2008 Safety,
tolerability and pharmacokinetics of intravenous ghrelin for cancer-
related anorexia/cachexia: a randomised, placebo-controlled, double-
blind, double-crossover study. British Journal of Cancer 98 300–308.
(doi:10.1038/sj.bjc.6604148)
Strassmann G, Fong M, Kenney JS & Jacob CO 1992 Evidence for the
involvement of interleukin 6 in experimental cancer cachexia.
Journal of Clinical Investigation 89 1681–1684. (doi:10.1172/JCI115767)
Suh SY, Choi YS, Yeom CH, Kwak SM, Yoon HM, Kim DG, Koh SJ, Park J,
Lee MA, Lee YJ et al. 2013 Interleukin-6 but not tumour necrosis factor-
alpha predicts survival in patients with advanced cancer. Supportive
Care in Cancer 21 3071–3077. (doi:10.1007/s00520-013-1878-4
)
Taylor DD, Gercel-Taylor C, Jenis LG & Devereux DF 1992 Identification of
a human tumor-derived lipolysis-promoting factor. Cancer Research 52
829–834.
Tchekmedyian NS, Hickman M, Siau J, Greco FA, Keller J, Browder H &
Aisner J 1992 Megestrol acetate in cancer anorexia and weight loss.
Cancer 69 1268–1274. (doi:10.1002/cncr.2820690532)
Teng MN, Turksen K, Jacobs CA, Fuchs E & Schreiber H 1993 Prevention
of runting and cachexia by a chimeric TNF receptor-Fc protein.
Clinical Immunology and Immunopathology 69 215–222. (doi:10.1006/
clin.1993.1172)
Teunissen SC, Wesker W, Kruitwagen C, de Haes HC, Voest EE & de Graeff A
2007 Symptom prevalence in patients with incurable cancer: a
systematic review. Journal of Pain and Symptom Management 34 94–104.
(doi:10.1016/j.jpainsymman.2006.10.015)
Tisdale MJ 2003 The ‘cancer cachectic factor’. Supportive Care in Cancer 11
73–78. (doi:10.1007/s00520-002-0408-6)
Tisdale MJ 2004 Cancer cachexia. Langenbeck’s Archives of Surger y 389
299–305. (doi:10.1007/s00423-004-0486-7)
Tisdale MJ 2009 Mechanisms of cancer cachexia. Physiological Reviews 89
381–410. (doi:10.1152/physrev.00016.2008)
Todorov P, Cariuk P, McDevitt T, Coles B, Fearon K & Tisdale M 1996
Characterization of a cancer cachectic factor. Nature 379 739–742.
(doi:10.1038/379739a0)
Torelli GF, Meguid MM, Moldawer LL, Edwards CK, Kim HJ, Carter JL,
Laviano A & Rossi Fanelli F 1999 Use of recombinant human soluble
TNF receptor in anorectic tumor-bearing rats. American Journal of
Physiology 277 R850–R855.
Tsoli M, Moore M, Burg D, Painter A, Taylor R, Lockie SH, Turner N,
Warren A, Cooney G, Oldfield B et al. 2012 Activation of thermogenesis
in brown adipose tissue and dysregulated lipid metabolism associated
with cancer cachexia in mice. Cancer Research 72 4372–4382.
(doi:10.1158/0008-5472.CAN-11-3536)
Varma M, Laviano A, Meguid MM, Gleason JR, Yang ZJ & Oler A 2000
Comparison of early feeding pattern dynamics in female and male rats
after reversible ventromedial nucleus of hypothalamus block. Journal of
Investigative Medicine 48 417–426.
Wang DT, Yin Y, Yang YJ, Lv PJ, Shi Y, Lu L & Wei LB 2014 Resveratrol
prevents TNF-a-induced muscle atrophy via regulation of Akt/mTOR/
FoxO1 signaling in C2C12 myotubes. International Immunoph armacol-
ogy 19 206–213. (doi:10.1016/j.intimp.2014.02.002)
Watanabe S & Bruera E 1996 Anorexia and cachexia, asthenia, and
lethargy. Hematology/Oncology Clinics of North America 10 189–206.
(doi:10.1016/S0889-8588(05)70334-8)
Weiland TJ, Anthony-Harvey-Beavis D, Voudouris NJ & Kent S 2006
Metabotropic glutamate receptors mediate lipopolysaccharide-induced
fever and sickness behavior.
Brain, Behavior, and Immunity 20 233–245.
(doi:10.1016/j.bbi.2005.08.007)
Whitaker KW & Reyes TM 2008 Central blockade of melanocortin receptors
attenuates the metabolic and locomotor responses to peripheral
interleukin-1b administration. Neu ropharmacology 54 509–520.
(doi:10.1016/j.neuropharm.2007.10.014)
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R42
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
Wiedenmann B, Malfertheiner P, Friess H, Ritch P, Arseneau J,
Mantovani G, Caprioni F, Van Cutsem E, Richel D, DeWitte M et al.
2008 A multicenter, phase II study of infliximab plus gemcitabine in
pancreatic cancer cachexia. Journal of Supportive Oncology 6 18–25.
Wieland BM, Stewart GD, Skipworth RJ, Sangster K, Fearon KC, Ross JA,
Reiman TJ, Easaw J, Mourtzakis M, Kumar V et al. 2007 Is there a
human homologue to the murine proteolysis-inducing factor?
Clinical Cancer Research 13 4984–4992. (doi:10.1158/1078-0432.CCR-
07-0946)
Winter A, MacAdams J & Chevalier S 2012 Normal protein anabolic
response to hyperaminoacidemia in insulin-resistant patients with
lung cancer cachexia. Clinical Nutrition 31 765–773. (doi:10.1016/
j.clnu.2012.05.003)
Wisse BE, Frayo RS, Schwartz MW & Cummings DE 2001 Reversal of cancer
anorexia by blockade of central melanocortin receptors in rats.
Endocrinology 142 3292–3301. (doi:10.1210/endo.142.8.8324)
Xia Y & Schneyer AL 2009 The biology of activin: recent advances in
structure, regulation and function. Journal of Endocrinology 202 1–12.
(doi:10.1677/JOE-08-0549)
YoshikawaT, NoguchiY, Doi C,MakinoT & Nomura K 2001 Insulinresistance
in patients with cancer: relationships with tumor site, tumor stage,
body-weight loss, acute-phase response, and energy expenditure.Nutrition
17 590–593. (doi:10.1016/S0899-9007(01)00561-5)
Yu Z, Li P, Zhang M, Hannink M, Stamler JS & Yan Z 2008 Fiber type-specific
nitric oxide protects oxidative myofibers against cachectic stimuli. PLoS
ONE 3 e2086. (doi:10.1371/journal.pone.0002086)
Zaki MH, Nemeth JA & Trikha M 2004 CNTO 328, a monoclonal antibody
to IL-6, inhibits human tumor-induced cachexia in nude mice.
International Journal of Cancer 111 592–595. (doi:10.1002/ijc.20270)
Zhang HH, Halbleib M, Ahmad F, Manganiello VC & Greenberg AS 2002
Tumor necrosis factor-a stimulates lipolysis in differentiated human
adipocytes through activation of extracellular signal-related kinase and
elevation of intracellular cAMP. Diabetes 51 2929–2935. (doi:10.2337/
diabetes.51.10.2929)
Zhang Z, Zhang H, Li B, Meng X, Wang J, Zhang Y, Yao S, Ma Q, Jin L,
Yang J et al. 2014 Berberine activates thermogenesis in white and
brown adipose tissue. Nature Communications 5 5493. (doi:10.1038/
ncomms6493)
Zhou X, Wang JL, Lu J, Song Y, Kwak KS, Jiao Q, Rosenfeld R, Chen Q,
Boone T, Simonet WS et al. 2010 Reversal of cancer cachexia and muscle
wasting by ActRIIB antagonism leads to prolonged survival. Cell 142
531–543. (doi:10.1016/j.cell.2010.07.011)
Received in final form 12 June 2015
Accepted 22 June 2015
Accepted Preprint published online 25 June 2015
Journal of Endocrinology
Review
M C S MENDES and others Mechanisms of cancer cachexia
226:3 R43
http://joe.endocrinology-journals.org Ñ 2015 Society for Endocrinology
DOI: 10.1530/JOE-15-0170 Printed in Great Britain
Published by Bioscientifica Ltd.
... The decrease in muscle mass is a standard feature, while weight loss and fat mass reduction are commonly seen in cachexia but not typically found in sarcopenia. In cachexia, we can always find the specific disease that led to this condition, whereas sarcopenia is a multifactorial condition that can often occur without a triggering illness [42]. ...
... Chronic low-grade inflammation contributes to a loss of muscle mass, strength, and function; therefore, high levels of cytokines like IL-8 and IL-2R are often elevated in sarcopenic patients [42,46]. ...
Article
Full-text available
Simple Summary Sarcopenia is a geriatric syndrome characterized by progressive and generalized loss of muscle strength and muscle mass. It associated with reduced physical function and negative outcomes such as falls, hospitalization, loss of autonomy, disability, and mortality. Sarcopenia has bought value in the cancer management, given its impact on patients’ treatments and prognosis. In hematologic diseases, sarcopenia predicts toxicity, treatment’s response, influences overall survival and non-cancer-related risk of death. Diagnosing and properly framing sarcopenia is of great importance in designing the patient appropriate treatment and to perform a personalized or “tailor made” approach and supportive care. Early recognize sarcopenia allows to reverse the muscle loss process and to avoid negative impacts of sarcopenia syndrome on the patients’ trajectory. Abstract Sarcopenia is a geriatric syndrome characterized by a progressive loss of systemic muscle mass and decreased muscle strength or physical function. Several conditions have a role in its pathogenesis, significantly impacting adverse outcomes such as falls, functional decline, frailty, disability, multiple hospitalizations, and mortality. In the oncological setting, sarcopenia is associated with an increased risk of treatment toxicity, postoperative complications, and a higher mortality rate related to other causes (e.g., pneumonia). In the hematological field, even more so, sarcopenia predicts toxicity and response to treatments. In patients with hematologic malignancy, low muscle mass is associated with adverse outcomes and is a predictor of overall survival and non-relapse mortality. Therefore, it is essential to correctly recognize sarcopenia, evaluate the risk factors and their impact on the patient’s trajectory, and effectively treat sarcopenia. Sarcopenia is a reversible condition. The most effective intervention for reversing it is physical exercise combined with nutrition. The objective of clinical assessment focused on sarcopenia is to be able to carry out a “tailor-made treatment”.
... Among the reasons for muscle loss in oncological patients, some important processes have been recognized: 1) a greater splanchnic sequestration of amino acids, in particular of EAAs, compared to physiological condition, in order to support the liver activity during the dramatic inflammatory response [61]. In cancer patients, EAA muscle release is strongly activated to sustain the increased demand of systemic requirement; 2) an increased energy demand that over-regulates protein breakdown so as to supply amino acid substrates for glucose and glycogen synthesis, causing further skeletal muscle mass loss [64]; 3) the effects of antitumor treatments and the metabolic alterations due to the illness which can negatively affect the absorption, digestion or use of EAAs, modifying their physiological plasma profiles [63]; 4) the anabolic responsiveness to amino acid signalling is significantly reduced (i.e., anabolic resistance). ...
... In fact, cancer patients display symptoms of anorexia, due to cancer cachexia, which induce the weight loss as a consequence of altered neurohormonal signals of appetite control [27]. The hypothalamus coordinates the neurons responsible to secrete anorexigenic (cocaine-and amphetamine-regulated transcript (CART) and pro-opio-melanocortin (POMC)) or orexigenic (agouti-related protein (AgRP) and neuropeptide Y (NPY)) to control food intake; however, the dysfunctional upregulating of the POMC neurons secretion results in a low food intake [9,28]. ...
Article
Full-text available
Background: Malnutrition and metabolic alterations of cancer cachexia are often associated with negative weight loss and muscle mass wasting. In this sense, protein supplementation can be a strategy to help counteract the loss and/or maintenance of mass in these patients. The aim of this study was to evaluate the effect of leucine supplementation on body composition in outpatients with gastrointestinal tract cancer. Methods: It was a randomized, blinded, controlled, parallel trial, performed in male patients with a cancer diagnosis of the gastrointestinal tract and appendix organs undergoing chemotherapy. All the patients were allocated to one of the protocol groups: L-leucine supplement or the control group, during 8 weeks of intervention. We evaluated the body composition through bioelectrical impedance analysis, the cancer cachexia classification, and the diet intake before and after the intervention protocol. The intention-to-treat approach was performed to predict the missing values for all patients who provide any observation data. Results: The patients were an average age of 65.11 ± 7.50 years old. In the body composition analysis with patients who finished all the supplementation, we observed a significant gain in body weight (61.79.9 ± 9.02 versus 64.06 ± 9.45, p = 0.01), ASMM (7.64 ± 1.24 versus 7.81 ± 1.20, p = 0.02) in the Leucine group, whereas patients in the control did not present significant variation in these parameters. There was no significant intergroup difference. While in the analysis included the patients with intention-to-treat, we found a significant increase in body weight (p = 0.01), BMI (p = 0.01), FFM (p = 0.03), and ASMM (p = 0.01) in the Leucine group. No significant intergroup differences. These results also similar among cachectic patients. Conclusion: A balanced diet enriched with free-Leucine supplementation was able to promotes gains in body weight and lean mass in older men diagnosticated with gastrointestinal and appendix organs of digestion cancer after 8 weeks. However, the fact that most men are non-cachectic or pre-cachectic is not clear if the increase in muscle mass was due to a high intake of leucine, since no difference between groups was detected. Moreover, we know that benefits on body composition are due to adequate calorie and macronutrients consumption and that balanced feeding according to nutrition Guidelines seems crucial and must be advised during the oncological treatment.
... Biomarkers released by tumor cells or the immune system contribute to the development of anorexia. TNF (Tumor necrosis factor), IL-1, and IL-6 are the most common biomarkers that reduce appetite (Mendes et al., 2015;Teixeira et al., 2019). Additionally, these cytokines may indicate skeletal muscle inflammation in cancer patients. ...
Article
The number of people being diagnosed with cancer is increasing rapidly worldwide. Cancer remains the greatest cause of death worldwide. Whey has attracted much attention in recent years as a natural source because of its significant applications to health benefits. Nowadays, whey is mostly utilised as an energy source for sports beverages and medicinal purposes in several nations. Whey components are becoming more popular among consumers for their nutraceutical properties, making them an appealing topic for cancer research which is a global public health issue. Researchers use whey protein in cancer prevention and therapy. Several in vitro and in vivo studies have shown whey protein consumption to elicit anti-cancer effects. Medical science now overwhelmingly favours whey protein's therapeutic significance, particularly in cancer cachexia treatment. Furthermore, whey protein supplementation is a viable, practicable, and cost-effective strategy for cancer cachexia syndrome treatment. This is particularly true considering whey protein's high leucine content and intrinsic capacity to alter IGF-I concentrations. More research is required to assess the usefulness of whey protein supplementation as an adjuvant in cancer therapy. Bearing in mind the nutraceutical significance of whey-derived proteins, this review paper highlights its emerging bioactive role to prevent cancer and improve human health.
... Chronic inflammation induces myeloidderived suppressor cells, which suppress the activity of CD8+ T-cells and activate immunosuppressive regulatory T-cells [25][26][27]. Additionally, the inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), IL-1, and IL-8, affect skeletal muscle, leading to cancer cachexia [28]. TNF-α activates the nuclear factor kappa-light-chain-enhancer of activated B cell pathway and ubiquitin-mediated proteasome catabolism of muscle protein [29]. ...
Article
Full-text available
Simple Summary Although body composition-related biomarkers are associated with the prognosis of patients with cancer, whether they are associated with the therapeutic effects of immune checkpoint inhibitors remains unclear. We found that among body composition-related biomarkers, sarcopenia based on skeletal muscle mass was strongly associated with the treatment efficacy of immune checkpoint inhibitors in patients with metastatic renal cell carcinoma. However, body composition-related biomarkers based on subcutaneous or visceral fat were not associated with treatment efficacy. The skeletal muscle releases myokines to activate the immune system. Therapeutic interventions for sarcopenia may not only improve patients’ quality of life but also improve the therapeutic effects of immune checkpoint inhibitors and prolong the prognosis of patients. Abstract Introduction: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic renal cell carcinoma (mRCC); however, validating body composition-related biomarkers for their efficacy remains incomplete. We evaluated the association between body composition-related markers and the prognosis of patients with mRCC who received ICI-based first-line therapies. Patients and Methods: We retrospectively investigated 60 patients with mRCC who underwent ICI-based therapy as their first-line treatment between 2019 and 2023. Body composition variables, including skeletal muscle, subcutaneous fat, and visceral fat indices, were calculated using baseline computed tomography scans. Sarcopenia was defined according to sex-specific cut-off values of the skeletal mass index. The associations between body composition indices and objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) were evaluated. Results: Patients with sarcopenia had lower ORR and DCR than those without sarcopenia (33.3% vs. 61.1%, p = 0.0436 and 52.4% vs. 94.4%, p = 0.0024, respectively). Patients with sarcopenia had a significantly shorter median PFS (14 months vs. not reached, p = 0.0020) and OS (21 months vs. not reached, p = 0.0023) than patients without sarcopenia did. Sarcopenia was a significant predictor of PFS (hazard ratio [HR], 4.31; 95% confidence interval [CI], 1.65–14.8; p = 0.0018) and OS (HR, 5.44; 95% CI, 1.83–23.4; p = 0.0013) along with poor IMDC risk. No association was found between the subcutaneous, visceral, and total fat indices and the therapeutic effect of ICI-based therapy. Conclusions: Sarcopenia was associated with a lower response and shorter survival rates in patients with mRCC who received first-line ICI-based therapy.
Preprint
Full-text available
Background Taste disorders are common in patients with cancer undergoing systemic therapy, persist during treatment and are associated with reduced food intake, increasing the risk of malnutrition. Cachectic syndrome, which is common in these patients and characterized by marked weight loss, anorexia, asthenia and anemia, is linked to the presence and growth of the tumor and leads to systemic inflammation. Synsepalum dulcificum is a plant whose berries contain miraculin, a glycoprotein that transforms sour tastes into sweet ones and could serve to ameliorate taste disorders in patients with cancer. Objective To evaluate the effect of the regular intake of Dried Miracle Berries (DMB), a novel food containing miraculin, on several biomarkers of inflammation and cachexia in malnourished patients with cancer and taste disorders receiving systemic antineoplastic therapy. Materials and methods Triple-blind, randomized, placebo-controlled clinical trial. Thirty-one patients with cancer of various etiologies receiving chemotherapy were enrolled in a pilot study and divided into three groups. The first group received a tablet containing 150 mg of DMB (standard dose); the high-dose group received a tablet of 300 mg of DMB, and the third group received a tablet with 300 mg of the placebo for three months before each main meal. Plasma levels of several molecules associated with inflammation and cancer cachexia were measured using the X-MAP Luminex multiplexing platform. Results The three groups showed a decrease in the plasma levels of IL-6, IL-1β, TNF-α, and PIF throughout the intervention, although the percentage change from baseline was greater in patients receiving a standard dose of DMB. In contrast, the CNTF concentration only decreased in the DMB standard-dose group. This group also presented the greatest reduction in the IL-6/ IL-10 ratio, while IL-15 and IL-10 increased in the groups treated with DMB but not in the placebo. Regardless of DMB consumption, sTNFR-II tended to decrease with treatment in patients who responsed well to the antineoplastic treatment. We did not find significant correlations between cytokines and sensory variables or dietary and nutritional status. Conclusions The regular consumption of a standard dose of the food supplement DMB containing miraculin along with a systemic antineoplastic treatment can contribute to reducing biomarkers of inflammation and cachexia in malnourished patients with cancer exhibiting taste disorders.
Article
Full-text available
Taste disorders are common among cancer patients undergoing chemotherapy, with a prevalence ranging from 20% to 86%, persisting throughout treatment. This condition leads to reduced food consumption, increasing the risk of malnutrition. Malnutrition is associated not only with worse treatment efficacy and poor disease prognosis but also with reduced functional status and quality of life. The fruit of Synsepalum dulcificum (Daniell), commonly known as miracle berry or miracle fruit, contains miraculin, a taste-modifying protein with profound effects on taste perception. The CLINMIR Protocol is a triple-blind, randomized, placebo-controlled clinical trial designed to evaluate the regular consumption of a food supplement containing a miraculin-based novel food, dried miracle berry (DMB), on the taste perception (measured through electrogustometry) and nutritional status (evaluated through the GLIM Criteria) of malnourished cancer patients under active antineoplastic treatment. To this end, a pilot study was designed with 30 randomized patients divided into three study arms (150 mg DMB + 150 mg freeze-dried strawberries, 300 mg DMB, or placebo) for three months. Throughout the five main visits, an exhaustive assessment of different parameters susceptible to improvement through regular consumption of the miraculin-based food supplement will be conducted, including electrical and chemical taste perception, smell perception, nutritional and morphofunctional assessment, diet, quality of life, the fatty acid profile of erythrocytes, levels of inflammatory and cancer-associated cytokines, oxidative stress, antioxidant defense system, plasma metabolomics, and saliva and stool microbiota. The primary anticipated result is that malnourished cancer patients with taste distortion who consume the miraculin-based food supplement will report an improvement in food taste perception. This improvement translates into increased food intake, thereby ameliorating their nutritional status and mitigating associated risks. Additionally, the study aims to pinpoint the optimal dosage that provides maximal benefits. The protocol adheres to the SPIRIT 2013 Statement, which provides evidence-based recommendations and is widely endorsed as an international standard for trial protocols. The clinical trial protocol has been registered at the platform for Clinical Trials (NCT05486260).
Article
Objective The objective of this paper was to provide an understanding of cachexia in relation to oral squamous cell carcinoma relevant to oral health care. The paper is a scoping review of aspects of the clinical presentation, aetiology and management of cachexia in relation to oral health and oral health care. Methods A combined search of MEDLINE and EMBASE databases (via OVID) was conducted using the terms ([Head and Neck] OR [Oral Squamous Cell Carcinoma]) AND (Cachexia). Duplicates were removed and results were subsequently limited to studies published between 2000 and 2023, humans and English language. After screening and full‐text assessment a total number of 87 studies were included in the review. Results It is evident that cachexia is a not uncommon feature of patients with advanced malignancy of the head and neck driven by a multitude of mechanisms, induced by the tumour itself, that lead to reduced nutritional intake, increased metabolism and loss of adipose and skeletal tissue. Conclusion While a variety of nutritional, physical, psychological and pharmacological interventions may improve quality and duration of life, ultimately the diagnosis of cachexia in relation to head and neck cancer remains an indicator of poor life expectancy.
Article
Background: Cancer-cachexia is a complex syndrome secondary to physiological mechanisms related to classical hormone and immune alterations, where contributions of neuro-endocrine involvement have been less evaluated. Therefore, the aim of our study was to explore relationships between PTHrP and whole body metabolism in patients with progressive pancreatic carcinoma; relevant to "fat tissue browning". Methods: Patient serum samples and clinical information were retrieved from earlier translational projects (1995-2005), at Sahlgrenska University Hospital in Gothenburg. Blood PTHrP levels were determined at Harvard medical School (2014). Patient data included: medical history, clinical laboratory tests, food diaries, resting metabolic expenditure, body composition, exercise capacity, Health-Related Quality of Life (SF-36) and mental disorders (HAD-scales). Results: Serum PTHrP was detectable in 17 % of all samples without significance to tumor stage. PTHrP-negativity at inclusion remained during follow-up. Mean PTHrP concentration was 262±274 pg/ml, without sex difference and elevation over time. PTHrP-positive and negative patients experienced similar body weight loss (%) at inclusion, with a trend to deviate at follow ups (16.8±8.2% vs. 13.1±8.2%, p<0.06), where PTHrP concentrations showed correlations to weight loss, handgrip strength and Karnofsky performance, without difference in exercise capacity. PTHrP-positivity was related to increased whole body fat oxidation (p<0.006-0.01) and reduced carbohydrate oxidation (p<0.01-0.03), independently of peripheral lipolysis. Metabolic alterations in PTHrP-positive patients were related to reduced Health Related Quality of life (SF: p<0.08, MH: p<0.02), and increased anxiety and depression (HAD 1-7: p<0.004; HAD 8-14: p<0.008). Conclusion: Serum PTHrP positivity in patients with pancreatic carcinoma was related to altered whole body oxidative metabolism; perhaps induced by "browning" of fat cells?
Article
Full-text available
Context: Cachexia is a multifactorial syndrome, characterized by loss of skeletal muscle mass and not fully reversible by nutritional support. Recent animal observations suggest that production of Activin A (ActA) and Myostatin (Mstn) by some tumors might contribute to cancer cachexia. Objective: Our goal was to investigate the role of ActA and Mstn in the development of the human cancer cachexia. Design/Setting: The ACTICA study is a cross-sectional study, which prospectively enrolled patients from a tertiary-care center between January 2012 and March 2014. Subjects/Outcome Measures: 152 patients with colorectal or lung cancer had clinical, nutritional and functional assessment. Body composition was measured by CT-scan, anthropometry and bioimpedance. Plasma concentrations of ActA, Mstn and Follistatin were determined. Results: Cachexia was associated with reduced lean and fat mass (p<0.01 and p<0.001), reduced physical function, lower quality of life and increased symptoms (QLQC30; p<0.001). Anorexia (SNAQ score <14) was more common in cachectic patients (CC) than in non-cachectic patients (CNC) (p<0.001). ActA concentrations in CC patients were higher than in CNC patients (+40%; p<0.001) and were correlated positively with weight loss (R=0.323; p<0.001) and negatively with SNAQ score (R=-0.225; p<0.01). In contrast, Mstn concentrations were decreased in CC patients compared to CNC patients (-35%; p<0.001). Conclusions: These results demonstrate an association between circulating concentrations of ActA and the presence of the anorexia/cachexia syndrome in cancer patients. Given the known muscle atrophic effects of ActA, our study suggests that increased circulating concentrations of ActA may contribute to the development of cachexia in cancer patients.
Article
Full-text available
Cachexia is a muscle-wasting syndrome that contributes significantly to morbidity and mortality of many patients with advanced cancers. However, little is understood about how the severe loss of skeletal muscle characterizing this condition occurs. In the current study, we tested the hypothesis that the muscle protein myostatin is involved in mediating the pathogenesis of cachexia-induced muscle wasting in tumor-bearing mice. Myostatin gene inactivation prevented the severe loss of skeletal muscle mass induced in mice engrafted with Lewis lung carcinoma (LLC) cells or in ApcMin/+ mice, an established model of colorectal cancer and cachexia. Mechanistically, myostatin loss attenuated the activation of muscle fiber proteolytic pathways by inhibiting the expression of atrophy-related genes, MuRF1 and MAFbx/Atrogin-1, along with autophagy-related genes. Notably, myostatin loss also impeded the growth of LLC tumors, the number and the size of intestinal polyps in ApcMin/+ mice, thus strongly increasing survival in both models. Gene expression analysis in the LLC model showed this phenotype to be associated with reduced expression of genes involved in tumor metabolism, activin signaling, and apoptosis. Taken together, our results reveal an essential role for myostatin in the pathogenesis of cancer cachexia and link this condition to tumor growth, with implications for furthering understanding of cancer as a systemic disease.
Article
Full-text available
Obesity develops when energy intake exceeds energy expenditure. Promoting brown adipose tissue formation and function increases energy expenditure and hence may counteract obesity. Berberine (BBR) is a compound derived from the Chinese medicinal plant Coptis chinensis. Here we show that BBR increases energy expenditure, limits weight gain, improves cold tolerance and enhances brown adipose tissue (BAT) activity in obese db/db mice. BBR markedly induces the development of brown-like adipocytes in inguinal, but not epididymal adipose depots. BBR also increases expression of UCP1 and other thermogenic genes in white and BAT and primary adipocytes via a mechanism involving AMPK and PGC-1α. BBR treatment also inhibits AMPK activity in the hypothalamus, but genetic activation of AMPK in the ventromedial nucleus of the hypothalamus does not prevent BBR-induced weight loss and activation of the thermogenic programme. Our findings establish a role for BBR in regulating organismal energy balance, which may have potential therapeutic implications for the treatment of obesity.
Article
The primary task of white adipose tissue (WAT) is the storage of lipids. However, "beige" adipocytes also exist in WAT. Beige adipocytes burn fat and dissipate the energy as heat, but their abundance is diminished in obesity. Stimulating beige adipocyte development, or WAT browning, increases energy expenditure and holds potential for combating metabolic disease and obesity. Here, we report that insulin and leptin act together on hypothalamic neurons to promote WAT browning and weight loss. Deletion of the phosphatases PTP1B and TCPTP enhanced insulin and leptin signaling in proopiomelanocortin neurons and prevented diet-induced obesity by increasing WAT browning and energy expenditure. The coinfusion of insulin plus leptin into the CNS or the activation of proopiomelanocortin neurons also increased WAT browning and decreased adiposity. Our findings identify a homeostatic mechanism for coordinating the status of energy stores, as relayed by insulin and leptin, with the central control of WAT browning. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Induction of beige cells causes the browning of white fat and improves energy metabolism. However, the central mechanism that controls adipose tissue browning and its physiological relevance are largely unknown. Here, we demonstrate that fasting and chemical-genetic activation of orexigenic AgRP neurons in the hypothalamus suppress the browning of white fat. O-linked β-N-acetylglucosamine (O-GlcNAc) modification of cytoplasmic and nuclear proteins regulates fundamental cellular processes. The levels of O-GlcNAc transferase (OGT) and O-GlcNAc modification are enriched in AgRP neurons and are elevated by fasting. Genetic ablation of OGT in AgRP neurons inhibits neuronal excitability through the voltage-dependent potassium channel, promotes white adipose tissue browning, and protects mice against diet-induced obesity and insulin resistance. These data reveal adipose tissue browning as a highly dynamic physiological process under central control, in which O-GlcNAc signaling in AgRP neurons is essential for suppressing thermogenesis to conserve energy in response to fasting. PAPERCLIP:
Article
Cancer cachexia is a devastating, multifactorial and often irreversible syndrome that affects around 50-80% of cancer patients, depending on the tumour type, and that leads to substantial weight loss, primarily from loss of skeletal muscle and body fat. Since cachexia may account for up to 20% of cancer deaths, understanding the underlying molecular mechanisms is essential. The occurrence of cachexia in cancer patients is dependent on the patient response to tumour progression, including the activation of the inflammatory response and energetic inefficiency involving the mitochondria. Interestingly, crosstalk between different cell types ultimately seems to result in muscle wasting. Some of the recent progress in understanding the molecular mechanisms of cachexia may lead to new therapeutic approaches.
Article
The AMP-activated protein kinase (AMPK) is a sensor of energy status that, when activated by metabolic stress, maintains cellular energy homeostasis by switching on catabolic pathways and switching off ATP-consuming processes. Recent results suggest that activation of AMPK by the upstream kinase LKB1 in response to nutrient lack occurs at the surface of the lysosome. AMPK is also crucial in regulation of whole body energy balance, particularly by mediating effects of hormones acting on the hypothalamus. Recent crystal structures of complete AMPK heterotrimers have illuminated its complex mechanisms of activation, involving both allosteric activation and increased net phosphorylation mediated by effects on phosphorylation and dephosphorylation. Finally, AMPK is negatively regulated by phosphorylation of the ‘ST loop’ within the catalytic subunit.
Article
Sphingosine 1-phosphate receptor 1 ( S1PR1) is a G-protein-coupled receptor for sphingosine-1-phosphate ( S1P) that has a role in many physiological and pathophysiological processes. Here we show that the S1P/ S1PR1 signalling pathway in hypothalamic neurons regulates energy homeostasis in rodents. We demonstrate that S1PR1 protein is highly enriched in hypothalamic POMC neurons of rats. Intracerebroventricular injections of the bioactive lipid, S1P, reduce food consumption and increase rat energy expenditure through persistent activation of STAT3 and the melanocortin system. Similarly, the selective disruption of hypothalamic S1PR1 increases food intake and reduces the respiratory exchange ratio. We further show that STAT3 controls S1PR1 expression in neurons via a positive feedback mechanism. Interestingly, several models of obesity and cancer anorexia display an imbalance of hypothalamic S1P/ S1PR1/ STAT3 axis, whereas pharmacological intervention ameliorates these phenotypes. Taken together, our data demonstrate that the neuronal S1P/ S1PR1/ STAT3 signalling axis plays a critical role in the control of energy homeostasis in rats.