You are on page 1of 14

Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Contents lists available at ScienceDirect

Critical Reviews in Oncology / Hematology


journal homepage: www.elsevier.com/locate/critrevonc

Cancer cachexia: Diagnosis, assessment, and treatment T


a,b,c b,c d e
Mohammadamin Sadeghi , Mahsa Keshavarz-Fathi , Vickie Baracos , Jann Arends ,

Maryam Mahmoudib,f,g, Nima Rezaeih,i,j,
a
Molecular Medicine Interest Group (MMIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
b
Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
c
School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
d
Department of Oncology, Division of Palliative Care Medicine, University of Alberta, Edmonton, AB, Canada
e
Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
f
Dietitians and Nutrition Experts Team (DiNET), Universal Scientific Education and Research Network (USERN), Tehran, Iran
g
Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
h
Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
i
Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
j
Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK

A R T I C LE I N FO A B S T R A C T

Keywords: Cancer cachexia is a multi-factorial syndrome, which negatively affects quality of life, responsiveness to che-
Cancer cachexia motherapy, and survival in advanced cancer patients. Our understanding of cachexia has grown greatly in recent
Nutrition years and the roles of many tumor-derived and host-derived compounds have been elucidated as mediators of
Palliative medicine cancer cachexia. However, cancer cachexia remains an unmet medical need and attempts towards a standard
Biomarker
treatment guideline have been unsuccessful. This review covers the diagnosis, assessment, and treatment of
Pharmacologic treatment
Nutritional treatment
cancer cachexia; the elements impeding the formulation of a standard management guideline; and future di-
Exercise rections of research for the improvement and standardization of current treatment procedures.
Psychological treatment

1. Introduction indirectly to the death of a significant proportion of cancer patients.


Moreover, cachexia negatively affects quality of life, responsiveness to
Cachexia is a multifactorial and multi-organ syndrome that is one of chemotherapy, and survival (Bachmann et al., 2008; Dewys et al., 1980;
the main causes of morbidity and mortality in late stages of chronic Penet and Bhujwalla, 2015). In 2011, an international consensus
conditions such as AIDS, chronic obstructive pulmonary disease statement defined cancer cachexia as:” a multifactorial syndrome
(COPD), congestive heart failure, multiple sclerosis, tuberculosis, and characterized by ongoing loss of skeletal muscle mass (with or without
cancer. More than 50 percent of cancer patients suffer from cachexia at loss of fat mass) that cannot be fully reversed by conventional nutri-
death, with the distribution varying by tumor type; the incidence is tional support and leads to progressive functional impairment (Fearon
highest in patients with gastric and pancreatic cancer (∼80%) while et al., 2011).” The main clinical presentation of cachexia in cancer
patients with breast cancer and leukemia demonstrate the lowest fre- patients is involuntary weight loss. Anorexia, systemic inflammation,
quency (∼40%) of affliction with cachexia (Argiles et al., 2005; insulin resistance, and increased resting energy expenditure (REE) are
Muscaritoli et al., 2006; Tisdale, 2005). Furthermore, one estimate of also usually associated with the condition.
the role of cachexia as a major contributor to cancer deaths stands at The wide variety of symptoms observed in cachexia is mediated
almost 20 percent (Warren, 1932). Other studies, however, have found through a spectrum of both tumor-derived and host-derived factors.
this proportion to be lower (Sesterhenn et al., 2012; Ambrus et al., Zinc alpha 2-glycoprotein (ZAG) is a well-recognized procachectic
1975; Inagaki et al., 1974), whereas some have considered this estimate factor secreted by tumor cells (Wyke and Tisdale, 2005; Felix et al.,
conservative (Langer et al., 2001)(Sesterhenn et al., 2012) achieved this 2011; Hirai et al., 1998). Moreover, cytokines secreted by tumor cells
estimate even though they had studied head and neck cancer patients may complement the host inflammatory response to the tumor (Tan
who are known to be at an especially high risk for malnutrition (Gorenc et al., 2014; Argiles et al., 2009). Pro-inflammatory cytokines (IL-1, IL-
et al., 2015). Nevertheless, cachexia can contribute directly or 6, TNF-α, IFN-γ) act through both centrally-mediated and peripheral


Corresponding author at: Research Center for Immunodeficiencies, Children’s Medical Center, Dr Qarib St, Keshavarz Blvd, 14194 Tehran, Iran.
E-mail address: rezaei_nima@tums.ac.ir (N. Rezaei).

https://doi.org/10.1016/j.critrevonc.2018.05.006
Received 30 May 2017; Received in revised form 16 April 2018; Accepted 9 May 2018
1040-8428/ © 2018 Elsevier B.V. All rights reserved.
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

pathways. These factors, either independently (Faggioni et al., 1997) or Table 1


through dysregulation of the leptin response pathway (Janik et al., Consensus-based definitions of cancer cachexia.
1997; Grunfeld et al., 1996), cause the persistent stimulation of anor- Number Study Criteria
exigenic pathways (Grossberg et al., 2010). These cytokines also lead to
the many metabolic changes observed in cachexia. These changes 1 Fearon et al. ≥ 2 out of next 3 criteria:
Weight loss (≥ 10%)
characterize the state of hypercatabolism (lipolysis (Petruzzelli et al.,
Low food intake (≤1500 kcal/d)
2014; Kir et al., 2014), muscle degradation (Fearon et al., 2013), and Systemic inflammation (C-reactive protein [CRP] ≥
acute phase response (Fearon et al., 1999)) observed in cachectic pa- 10 mg/L)
**
tients. In addition to the numerous molecular mediators of cachexia, 2 Evans et al. Weight loss > 5% over past 12 months and underlying
mechanical or digestive factors have been identified (Fearon et al., chronic disease
Or
2013; Tuca et al., 2013). Tumor burden or chemotherapy may lead to
BMI < 20
nausea, dysphagia, mucositis, pancreatic insufficiency, and malab- And
sorption (Deutsch and Kolhouse, 2004), resulting in reduced food in- 3 out of next 5 criteria:
take and subsequently weight loss (Wigmore et al., 1997). Abnormal Biochemistry
CRP > 5 mg/L
Although the aforementioned consensus-definition for cachexia has
HB < 12 g/dL
been validated (Blum et al., 2014), diagnosis remains a challenge due to Albumin < 3.2 g/dL
several confounding factors such as cachexia-like complications of Fatigue
cancer therapy and sarcopenic obesity (Fearon et al., 2013). Even Anorexia
though our knowledge of cancer cachexia has increased considerably Decreased Muscle Strength
Lean Muscle Depletion
during the last decade, cachexia remains an unmet medical need. The
3 EPCRC Weight loss > 5% over past 6 months without
condition is rarely recognized in clinical settings as weight loss is not starvation
routinely assessed (Churm et al., 2009) and treatment is usually in- And/or
adequate (Sun et al., 2015; Spiro et al., 2006). Furthermore, hetero- Weight loss > 2% and BMI < 20
geneity in the clinical presentation of cachexia (Fearon et al., 2012), And/or
Weight loss > 2% and sarcopenia
difficulties in consistent diagnosis of the disease at early stages (pre-
cachexia) (Blum et al., 2014; Argiles et al., 2014), the advanced age of ** This definition is not specific for cancer cachexia and has been proposed
many patients (Dodson et al., 2011), and the complexity of the multi- for cachexia associated with all types of underlying chronic disease.
modal approach needed to manage cachexia (Fearon, 2008) have
hampered efforts towards a standardized and effective treatment. In criteria does not directly measure systemic inflammation (assessed by
this review, we focus on the clinical management of cancer cachexia CRP levels) as a diagnostic factor due to the lack of contemporary data
(diagnosis, assessment, and treatment) and the latest advances in this sets for blood CRP levels in cancer cachectic patients (Fearon et al.,
field. We will also propose future directions towards the development 2011). The importance of contemporary data sets was highlighted in a
of an effective and standard guideline for the treatment of cachexia. study seeking to formulate a robust risk assessment method based on
cancer-associated weight loss percentage in 2015. In this study, the
2. Diagnosis demographic shift in cancer patients towards obesity and overweight in
recent times required the analysis of contemporary patient groups for
Recent advances in the field of cancer-associated cachexia have truly valid results. In the opinion of the consortium, the risk assessment
elucidated the pathophysiology of the disease to a great extent. As a method proposed in the 2015 study supersedes the weight criteria
result, a general consensus has formed within circles of health care originally proposed in the 2011 definition. In addition, this study de-
professionals and experts in cancer cachexia on the main symptoms of monstrated that the prognostic value of percent weight loss is limited
the condition, namely: weight loss, loss of appetite, failure to thrive, when considered independently from BMI (Martin et al., 2015). Finally,
and muscle wasting (Muscaritoli et al., 2016). However, the hetero- in comparing these three definitions, it must be noted that unlike the
geneity in the clinical presentation of the condition has impeded efforts first two, the EPCRC definition has been validated in a large patient
towards a single unifying definition of cachexia (Fearon et al., 2012). group in an international multicenter study (Blum et al., 2014).
Nevertheless, several attempts towards this goal have been made Although few in number, several studies have sought to evaluate
(Fearon et al., 2011; Bozzetti and Mariani, 2009; Fearon et al., 2006a; these proposed frameworks. In a study conducted by Vanhoutte et al.,
Evans et al., 2008; Muscaritoli et al., 2010; Argiles et al., 2011)(Fearon the third definition was shown to give a higher prevalence of cachexia
et al. (2006a))(Evans et al. (2008)), and an international panel of ex- in comparison to the second one. However, the difference of survival
perts under the auspices of the European Palliative Care Research rates between cachectic and non-cachectic patients was more sig-
Collaborative (EPCRC) (Fearon et al., 2011). In the end, a definition nificant when using the second definition, leading the authors to con-
proposed by the SCRINIO working group is also mentioned (Bozzetti clude that it may identify more severe cases of the disease (Vanhoutte
and Mariani, 2009). The main diagnostic criteria of these three fra- et al., 2016). In another study, the generic framework was similarly
meworks are listed in Table 1. reported to better differentiate between survival rates. However, unlike
It is important to note the basic differences between these three the Vanhoutte et al. study, more cachectic patients were identified
frameworks. The second framework has been proposed for cachexia using the general framework (van der Meij et al., 2013). A similar lack
associated with all types of underlying chronic disease (generic fra- of concordance between different definitions was observed in compar-
mework) while the other two frameworks have been formed specifically isons of the first and third definitions (Wesseltoft-Rao et al., 2015;
for cancer-associated cachexia (cancer-specific framework). Moreover, Thoresen et al., 2013; Wallengren et al., 2013). Although there are
compared to the other two definitions, the second definition puts less many drawbacks to comparing data accrued based on different sets of
emphasis on body weight loss. Instead, through the use of additional criteria, the observed contradiction of data between the studies may be
criteria, it tries to take into account other metabolic and nutritional attributed to the heterogeneity of patient groups, small sample sizes,
aspects of the disease (Vanhoutte et al., 2016). Furthermore, the Evans and long time-spans between measurement and assessment of different
et al. framework does not consider sarcopenia to be a diagnostic factor, features (Wesseltoft-Rao et al., 2015). Further studies addressing these
which is in stark contrast with the EPCRC framework. issues are warranted.
On the other hand, even though the authors of the EPCRC definition The validity of these definitions may also be questioned more
acknowledge blood CRP levels as an indication of catabolic drive, their

92
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

directly. For example, both the EPCRC and Evans et al. frameworks use 3.1. Nutritional screening and assessment
a single cut-off point of 20 for BMI (Fearon et al., 2011; Evans et al.,
2008). However, the recent European Society of Clinical Nutrition Insufficient dietary intake is a commonly observed phenomenon in
(ESPEN) statement advises the use of a higher cut-off point of 22 in the advanced cancer patients. Both independently and in association with
elderly (Cederholm et al., 2015). Given the importance of BMI in the cachexia, it is a main cause of morbidity and reduction of responsive-
risk assessment of cancer patients, this might affect analyses carried out ness towards treatment (Senesse et al., 2008). For example, pre-
on patients from different age groups. Another issue may be that the operative nutritional status has been shown to affect postoperative
EPCRC definition groups some cachectic patients as having refractory survival in lung cancer patients (Bo et al., 2015). Conversely, tumors,
disease based on, among other criteria, short life expectancy (Fearon especially of the head and neck and gastrointestinal tract, may directly
et al., 2011). However, many studies have reported estimations of life impair ingestion of food. Additionally, anti-cancer therapies including
expectancy to be inaccurate (Wilson et al., 2005; White et al., 2016; surgery, radiotherapy, and chemotherapy could impair nutrition intake
Mackillop and Quirt, 1997; Chow et al., 2005). This is important since through various unfavorable GI abnormalities (Nicolini et al., 2013). As
such categorization affects the treatment approach taken towards these a result, a vicious cycle forms, with malnutrition as a result of in-
patients. Furthermore, one study scrutinized the diagnostic value of sufficient dietary intake resulting in increasing therapy toxicity and the
including additional criteria to weight loss, the main criterion of the unmanaged tumor causing even more severe malnutrition. The effects
second framework. No correlation was observed between overall sur- of malnutrition are not merely physiological; with psychosocial effects
vival and these “minor criteria” (Letilovic and Vrhovac, 2013). Another and reduction in quality of life (QoL) commonly manifested (Isenring
study concluded that the differences observed in cachectic patients with et al., 2004; McGrath, 2002). Thus, it is clear that routine nutritional
different underlying chronic diseases might make the use of the generic screening and assessment of cancer patients could lead to the identifi-
definition for all types of cachexia irrelevant (Letilovic et al., 2013). cation of cancer patients at earlier stages of malnutrition. This could
Finally, the concept of pre-cachexia has been the subject of recent have considerable benefits as it would lead to the earlier commence-
discussion. Several studies have identified an association between the ment of therapy (Arends et al., 2017). Because routine assessment by
pre-cachectic status and better treatment response (Martin et al., 2015; nutritionists would not be timely and cost-effective, several nutritional
van der Meij et al., 2013; Wesseltoft-Rao et al., 2015). These studies, screening and assessment tools have been proposed instead (Table 2).
however, are not consistent in terms of the significance of the difference As it is evident from the table, the available nutritional screens are
in survival between pre-cachectic and non-cachectic patients (Martin far from perfect. Notably, many more studies in cohorts of patients with
et al., 2015; Wesseltoft-Rao et al., 2015). On the other hand, the con- advanced malignancies are required to fully validate these tests and to
cept of a pre-cachectic stage has been discredited because of either its clarify the contradictory results. As for the MNA, some studies have
very low prevalence (Blauwhoff-Buskermolen et al., 2014) or the lack of validated the prognostic value of the test, even suggesting MNA as a
sufficient diagnostic criteria to confidently denote a patient as pre-ca- better indicator than weight loss history (Gioulbasanis et al., 2011a;
chectic (Blum et al., 2014). Indeed, controlled clinical trials are needed Gioulbasanis et al., 2011b). However, other studies have questioned its
to objectively validate the presence of any added benefit from treat- validity mainly due to its low specificity (Neelemaat et al., 2011). Si-
ment in the pre-cachectic status. milarly, MUST has been validated in some studies (Velasco et al., 2011)
In the end, the definition proposed by the SCRINIO working group while discredited in others due to its low specificity and sensitivity
based on 1307 oncology patients in a multicenter study also merits (Neelemaat et al., 2011; Bauer and Capra, 2003). The NRS-2002’s uti-
discussion. This cancer-specific definition identifies and classifies ca- lity as a nutritional screening tool has also been questioned (Thoresen
chectic patients based on combinations of body weight loss (< 10%, et al., 2013). Furthermore, none of the nutritional screening tools are
precachexia; ≥10%, cachexia) and the presence/absence of at least 1 able to detect sarcopenic obesity as they do not assess muscularity
symptom of anorexia, fatigue, or early satiation. Unlike the three con- (Blum and Strasser, 2011). It must be noted, however, that comparison
sensus-based definitions, these criteria focus mainly on features avail- of different nutritional screening tools developed for different target
able upon clinical examination without considering systemic in- populations and different purposes (e.g., diagnostic vs prognostic) may
flammation and body composition and muscularity as important be flawed. This is especially true if data on disease outcomes without
diagnostic factors. As a result, this definition can be utilized by any nutritional intervention is used to assess the tools (Elia and Stratton,
clinician in any clinical setting (Bozzetti and Mariani, 2009). Im- 2012). Nevertheless, the importance of screening for the identification
portantly, the patient groups identified by this definition and the of patients in need of care warrants the implementation of organized
EPCRC criteria were not completely concordant in one study (Bozzetti, screening regardless of the screening tool used. Some tools (especially
2013); further highlighting the need for a universally accepted and the PG-SGA and NRS-2002 tools) have found widespread use and have
validated definition for cancer cachexia. proven useful in early recognition of malnutrition in cancer patients.
Therefore, it may be argued that, currently, the implementation of
routine screening of nutritional status in our healthcare centers out-
3. Assessment weighs the need for a fully optimized screening tool.

Cancer cachexia is a multidimensional disease, which requires a 3.2. Muscularity and body composition
broad range of assessments for the adequate characterization of the
condition in each patient. Furthermore, determining the overall con- As one of the hallmarks of cachexia, muscle depletion should be
dition of the patient is important as it indicates the best possible monitored in cancer patients. This could be achieved by either ana-
treatment plan; e.g., whether to pursue abortive (intended to stop the lyzing muscle mass or assessment of muscle strength. In assessing
progression of the condition) or palliative (intended to alleviate the muscle mass, cross-sectional imaging techniques, including computed
distress caused by the condition) therapy. Many of these evaluations are tomography (CT) or magnetic resonance imaging (MRI) are preferred
patient reported. This is because these assessments and subsequent over other methods (Fearon et al., 2011). In addition to determining
treatment commonly take place in the palliative setting. Therefore, the muscle mass, these methods are better evaluators of resting energy
goal of any therapy is to provide a subjectively tangible benefit over the expenditure (REE) (Gallagher et al., 1998) as they separate skeletal
patient’s current status (Wheelwright and Johnson, 2015). Here we muscle and adipose tissue more accurately (Jacquelin-Ravel and
summarize the 4 major aspects of assessing cancer cachexia. Pichard, 2012). Other methods include dual energy x-ray imaging
(DEXA), bio-impedance analysis (BIA), and mid upper-arm muscle area
by anthropometry (Fearon et al., 2011). Type II fast-twitch muscles are

93
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Table 3
Cancer- Methods of muscularity assessment in Caucasian cachectic patients.
specific
Assessment method Indication of muscle
Yes

No

No

No

No
depletion
Completed by patient/

Lumbar (L3) skeletal muscle index determined Men < 55 cm2/m2


by CT imaging (Prado et al., 2008) Women < 39 cm2/m2
Appendicular skeletal muscle index determined Men < 7.26 kg/m2
by DEXA (Baumgartner et al., 1998) Women < 5.45 kg/m2
Practitioner

Practitioner

Practitioner
practitioner

Whole body fat-free mass (FFM) index without Men < 14.6 kg/m2
Patient

bone determined by BIA (Janssen et al., Women < 11.4 kg/m2


Both

2002)
Mid upper-arm muscle area determined by Men < 32 cm2
anthropometry (Jetté et al., 1983) Women < 18 cm2
Assessment in addition

assessed as they are affected most commonly in cancer cachexia


to screening

(Mondello et al., 2015). Although less sensitive than cross-sectional


imaging, the DEXA and BIA have some advantages. The DEXA method
Yes

Yes

No

No

No

exposes the patient to less radiation (Mondello et al., 2015) while the
BIA method has widespread use due to its simplicity and low cost (Tuca
Yes (Neelemaat et al., 2011; Ferguson

Contradictory results (Thoresen et al.,


et al., 2011a; Neelemaat et al., 2011)
Yes (Martin et al., 2010; Kubrak and

Contradictory results (Velasco et al.,

et al., 2013). Unlike CT and MRI, however, these methods are not able
Validated in population with cancer

Contradictory results (Gioulbasanis

to assess muscle quality as determined by intra- and intermuscular fat


2011; Bauer and Capra, 2003)

(myosteatosis). This especially undermines the assessment of muscu-


2013; Velasco et al., 2011)

larity in the elderly or the obese (Barbat-Artigas et al., 2012; Erlandson


et al., 2016; Correa-de-Araujo et al., 2017).
While body composition serves as an indicator of muscularity, it
could also serve as a predictor of chemotherapy toxicity (Prado et al.,
Jensen, 2007)

et al., 1999)

2009). For the assessment of muscle strength, upper-limb hand-grip


dynamometry is the preferred method (Fearon et al., 2011). However,
this may be questioned with the observed lack of improvement in hand-
grip strength in the ROMANA trials (Temel et al., 2016). Muscle
strength is a rough manifestation of muscle mass and it might be a
Weight loss, appetite and food intake, activity

better indicator of cachexia than overall physical activity (Strasser,


Dietary and anthropometric measurement,
and function; metabolic demand, physical

2008) (Table 3).


Weight loss, appetite and food intake

Weight loss, BMI, acute disease

Weight loss, BMI, food intake

3.3. Quality of life and psychosocial assessment


Main assessment criteria

One of the much-neglected aspects of cancer cachexia is its psy-


chosocial effects on patients. These effects often present themselves as
negative emotions due to involuntary weight loss and reduced dietary
assessment

intake (Hopkinson et al., 2010). Weight loss may have different psy-
activity

chological effects on cancer patients. Most commonly, the changes in


physical appearance lead to altered self-perception and even embar-
Available nutritional assessment and screening tools for cachectic patients.

rassment (Reid et al., 2009). This leads to distress and social isolation.
Malnutrition Advisory Group of the British

However, some patients, particularly the obese, regard this weight loss
and Metabolism) (Kondrup et al., 2003)
(European Society for Clinical Nutrition
Association of Parenteral and Enteral

as beneficial (Hopkinson et al., 2006) and may even experience im-


provements in self-image. This welcoming attitude towards weight loss
might result in complacency in management of diet and physical ac-
tivity, thus accelerating the progression of the disease (Hopkinson,
(Ferguson et al., 1999)

2014).
(Guigoz et al., 1996)

Alterations in dietary intake could also lead to negative psychoso-


cial effects. Food performs many non-biological functions in our ev-
(Ottery, 1996)
Developed by

eryday lives and carries great symbolic importance (Bayer et al., 1983;
Nutrition

Chamberlain, 2004; Mathieson and Stam, 1995). Changes in eating


patterns may result in relational conflicts within both the family and
wider social contexts. Consequently, caretakers are often afflicted with
Malnutrition Universal Screening

their own psychological effects. As they try to halt the decline, they
Global Assessment (PG-SGA)

Nutritional Risk Screening-2002

attempt to increase nutritional intake through various methods


Patient-Generated Subjective

Mini-Nutritional Assessment

Malnutrition Screening Tool

(McClement et al., 2004; Hopkinson and Corner, 2006). The lack of


improvement could lead them to assume responsibility for the failure
and thus feel incompetent (McClement et al., 2004). This highlights the
Tool (MUST)

(NRS-2002)

necessity of providing psychological support for caretakers in addition


to patients in the clinic. As for the patient, the senses of failure, help-
(MNA)

(MST)

lessness, loss of independence, imminent death, and conflict with fa-


Table 2

Tool

mily members (Hopkinson, 2014) present themselves in the form of


various negative emotions (Oberholzer et al., 2013). Different factors

94
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Table 4
Quality of life (QoL) assessment tools in cachectic patients.
Assessment tool Number of items for Domains of health-related QoL Condition
evaluation covered

Cancer Rehabilitation 139-long form Physical, Functional, Social, Cancer


Evaluation System (CARES) (Schag et al., 1991; Schag and Heinrich, 1990) 59-short form Psychological, Sexual, Treatment
European Organization for the 30 Physical, Functional, Social, Cancer
Treatment and Research of Psychological, Treatment
Cancer-Quality of Life Questionnaire
(EORTC QLQ-C30) (Aaronson et al., 1993; Sprangers et al., 1998; Stukan et al.,
2017) **
EuroQOL (EuroQol, 1990; Park et al., 2006) 16 Physical, Functional, Social, General (Chronic Illness)
Psychological
Functional Assessment of Anorexia/Cachexia Therapy (FAACT) –(Functional 39 Physical, Functional, Social, Cancer Cachexia
Assessment of Cancer Therapy [FACT] combined with anorexia and cachexia Emotional, Sexual
subscale [ACS]) (Chang et al., 2005; Ribaudo et al., 2000; Salsman et al., 2015)
Functional Living Index-Cancer (FLIC) (Bektas and Akdemir, 2008; Fong et al., 2014; 22 Physical, Functional, Social, Cancer
Laenen and Alonso, 2010) Psychological, Treatment
Nottingham Health Profile (NHP) (Hunt et al., 1981; Montazeri et al., 2003) 45 Physical, Functional, Social, General Illness (Chronic
Psychological, Sexual illness-cancer)
Quality of Life Index: Cancer Version - III (Ferrans, 1990; Ferrans and Powers, 1992; 66 Physical, Functional, Social, Sexual Cancer
So et al., 2004)
World health Organization Quality of Life-100 item (WHOQOL-100) (The World 100 Physical, Functional, Social, General Illness
Health Organization Quality of Life Assessment, 1998; Lin et al., 2015; Paredes Psychological, Sexual
et al., 2010)

** The 24-item EORTC QLQ-CAX24 has been proposed as an update on this core questionnaire with promising results in the provisional phases. Studies for full
validation are currently underway (Wheelwright et al., 2017).

influence the severity of these emotions. Young people, for example, patients used in most of the mentioned studies. Also, many confounding
suffer more as they are more active in the community (Oberholzer elements could blur the relationship between these markers and ca-
et al., 2013; Lovgren et al., 2008). chexia. In one study, for example, leptin showed gender-dependent
Assessment of psychosocial and functional properties of patients association, decreasing in cachectic women while increasing in ca-
usually takes place through quality of life (QoL) assessments. Several chectic men (Wolf et al., 2006). Moreover, these serum markers might
subjective questionnaire-based methods are available and have been have intra-day variations, thus affecting research results (Fujiwara
validated, a summary of which is presented in Table 4. Even though et al., 2014). Finally, the degree of significance between the measured
these methods have been shown to be competent evaluators of quality amounts of these markers between cachectic and non-cachectic patients
of life in cancer cachexia patients, they have two fundamental flaws. may depend on the diagnostic criteria used (Bye et al., 2016). There-
Firstly, only the FAACT tool is cancer-cachexia specific and yet it has fore, more studies with more uniform and larger study groups are
been criticized for insufficient coverage of the social domain. Secondly, warranted in this field.
these tools require more general evaluation to be considered as fully
validated tools. Therefore, there remains the need for an internationally
validated tool, which adequately covers all psychosocial aspects of 4. Treatment
cancer cachexia (Wheelwright et al., 2013).
To date, an international standard care guideline for cancer ca-
3.4. Biomarkers of cachexia chexia with perfect effectivity does not exist. This hampers efforts to-
wards better treatment options as there is no universal gold standard to
Cancer cachexia has been associated with alterations in the con- which new methods could be compared with. For any treatment of
centrations of a broad array of compounds in the body fluids. These cachexia, the primary endpoints should be improvements in lean body
changes are associated with the different domains of the physiological mass, resting energy expenditure, fatigue, anorexia, quality of life,
functions affected in cancer-related anorexia and cachexia. The word performance status, and a reduction in pro-inflammatory cytokines
biomarker is intended to convey the broad meaning of any measurable (Donohoe et al., 2011). Patient-reported outcomes and other relevant
substance in the body which is either influential or predictive of the biomarkers also serve as endpoints. However, it should be noted that
incidence, outcome, or treatment response of the disease (Strimbu and the best treatment approach depends on the stage of the disease, as
Tavel, 2010). These markers have been proposed to not only identify palliative options are prioritized over curative modalities at end stages
cachectic patients, but also to distinguish different stages of cachexia (Maltoni et al., 2005). Currently, a consensus has formed that the
(Bilir et al., 2015). The most recognized serum components of cancer multifactorial nature of cancer cachexia requires a multimodal treat-
cachexia are the mediators of systemic inflammation, notably the acute ment approach (Madeddu et al., 2012). For example, while some clin-
phase response proteins (APRPs). In an effort to quantify the systemic ical studies have demonstrated that provision of high amino acid loads,
inflammation observed in cancer, the Glasgow Prognostic Score either through feeding or i.v. infusion, improves muscle protein
(mGPS/GPS) was developed in 2003 and has since been validated as a synthesis, these effects were not enough to reverse cachexia (Dillon
tool to objectively define cancer cachexia in numerous studies et al., 2007; Deutz et al., 2011; van Dijk et al., 2015;Bozzetti and
(McMillan, 2013). Other markers are associated with anorexia, lipo- Bozzetti, 2013; Lundholm et al., 2004). The same goes for trials with
lysis, and muscle degradation (Table 5). However, caution must be conventional and single-drug therapies (Gramignano et al., 2006).
taken in regarding these factors as validated markers of cachexia. Evi- Thus, further trials integrating these different therapeutic modalities
dence for the prognostic and predictive value of most of these markers seem to be the way forward. We will now consider the current treat-
is contradictory. In addition, cutoff values, which could be utilized in ment options available in each modality.
the clinic, are lacking for most of these markers. The contradictory
results could be attributed to the heterogeneity and small sample size of

95
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Table 5
Biomarkers of cancer cachexia.
Domain of Cancer Cachexia Associated biomarkers Observed change

Systemic Inflammation CRP Increase (Bilir et al., 2015; Batista et al., 2013; Kerem et al., 2008; Kemik et al., 2010; Burney
et al., 2012; Scheede-Bergdahl et al., 2012; Dev et al., 2014)
IL-1α Increase (Bilir et al., 2015; Kemik et al., 2010)
IL-1β Increase (Kemik et al., 2010; Scheede-Bergdahl et al., 2012), insignificant difference (Bilir
et al., 2015)
IL-6 Increase (Batista et al., 2013; Kemik et al., 2010; Burney et al., 2012; Scheede-Bergdahl et al.,
2012; Kim et al., 2012; Krzystek-Korpacka et al., 2007)
IFN-γ Insignificant difference (Kim et al., 2012)
IL-8 Increase (Kemik et al., 2010; Scheede-Bergdahl et al., 2012; Krzystek-Korpacka et al.,
2007;Song et al., 2009)
TNF-α Increase (Bilir et al., 2015; Kemik et al., 2010), insignificant difference (Kim et al., 2012)
IL-10 Increase (Kemik et al., 2010)
Albumin Decrease (Kerem et al., 2008; Kemik et al., 2010; Burney et al., 2012; Scheede-Bergdahl et al.,
2012; Dev et al., 2014)
Hormonal dysregulation Ghrelin Increase (Wolf et al., 2006; Kerem et al., 2008; Mondello et al., 2014; Shimizu et al., 2003)
insignificant difference (Kim et al., 2012), decrease(Kemik et al., 2010)
Obestatin Decrease (Mondello et al., 2014)
Testosterone Decrease (Bilir et al., 2015; Burney et al., 2012; Dev et al., 2014)
IGF-1 Decrease (Kerem et al., 2008; Huang et al., 2005)
Adipose-derived factors Adiponectin Increase (Wolf et al., 2006; Batista et al., 2013; Kerem et al., 2008), insignificant difference
(Kim et al., 2012), decrease (Kemik et al., 2010)
Resistin Increase (Kerem et al., 2008)
Leptin Increase (Wolf et al., 2006; Kerem et al., 2008; Kemik et al., 2010), insignificant difference,
decrease (Wolf et al., 2006; Kim et al., 2012; Mondello et al., 2014)
Tumor-derived factors Zinc-α2-glycoprotein (ZAG), also known as lipid- Increase (Felix et al., 2011), observed in urine (Todorov et al., 1998)
mobilizing factor (LMF)
Proteolysis Inducing Factor (PIF) Increase (Belizario et al., 1991), observed in urine (Wigmore et al., 2000)
VEGF-A and VEGF-C Increase (Kemik et al., 2010; Krzystek-Korpacka et al., 2007)
Midkine Increase (Kemik et al., 2010; Krzystek-Korpacka et al., 2007)

Several other factors have been proposed as markers, namely: angiotensin II, neutrophil-derived proteases, TGF-1β (Penafuerte et al., 2016); fentanyl (Suno et al.,
2015); PTHrP (Hong et al., 2016); glucagon-like peptide 1 (GLP1)(Felix et al., 2011); hemoglobin, NPY, orexin, and galanin (Bilir et al., 2015); myosin and other
muscle-related proteins in urine (Skipworth et al., 2010).

4.1. Pharmacologic treatment preclinical settings, the most important of which are listed in Tables 6.1
and 6.2 . Other notable drug categories include melanocortin-4-re-
Many different drugs have been suggested to be effective against ceptor (MC-4R) antagonists, TNF-α inhibitors, and monoclonal anti-
cancer cachexia. The rationale is usually based on the mechanism of bodies. Activation of the MC-4R in murine models decreases food-
action of the mediators of cachexia. Therefore, in a general classifica- seeking behavior, increases basal metabolic rate, and decreases lean
tion, these drugs perform 3 basic functions (Argiles et al., 2010): body mass (Marks et al., 2001). The effectiveness of MC-4R antagonists
has been demonstrated in murine models (Dallmann et al., 2011);
1 Reduction of the tumor-associated inflammation however, clinical trials are yet to be initiated. TNF-α inhibitors had
2 Capitalization on the anabolic potential of the body to counter the been proposed as potential drugs based on the large role TNF-α plays in
wasting and hypercatabolic state the mediation of cachexia. Surprisingly, several clinical trials were
3 Appetite stimulation unable to achieve any attenuation of the disease using infliximab, a
monoclonal antibody against TNF-α (Jatoi et al., 2010). Other biologic
Of course, the pathways involved in these phenomena are com- drugs, such as Clazakizumab (formerly ALD518) (Alder Biopharma-
pletely entangled, making the translation of laboratory results into ceuticals, Inc., Bothell, WA, USA) and OHR/AVR118 (Ohr Pharma-
clinical observations difficult to interpret. One solution is to identify ceutical, Inc., New York, NY, USA) have been studied in cancer ca-
convergence points where the different mediating cellular pathways are chexia. ALD518 is a humanized anti-IL-6 antibody which has been
integrated. An example is the ubiquitin-proteasome system, which ap- shown to prevent the reduction of lean body mass in phase II clinical
pears to be the dominant system in muscle protein breakdown (Shum trials in non-small cell lung cancer patients (Rigas et al., 2010). OHR/
and Polly, 2012). Another problematic factor is the lack of well-de- AVR118 targets both TNF-α and IL-6, and was observed to improve
signed randomized clinical trials on large study groups. The small size functional status, weight stabilization, appetite stimulation, and even
and heterogeneity of the patient groups, in addition to the widely weight gain (Chasen et al., 2011). Tocilizumab, an anti-IL-6 receptor
varying inclusion criteria and primary endpoints, render clear conclu- antibody sold under the trade name ACTEMRA® (Genentech, Inc.,
sions almost impossible to reach. In spite of all this, several drugs with South San Francisco, CA, USA), has been shown to bring significant
substantial promise have been identified. Currently, megestrol acetate improvements in body weight, nutritional status, inflammation, anemia
(MA) is the only drug in clinical use for the treatment of cachexia and and performance status in a number of case reports (Ando et al., 2014;
has been approved by the FDA for treating AIDS-associated cachexia. To Ando et al., 2013; Berti et al., 2013). Still other drugs have been pro-
date, no other drug has been shown to be superior to MA in efficacy and posed based on the pathogenesis of cancer cachexia but have not been
tolerability (Tuca et al., 2013). However, some researchers have ques- validated, such as growth hormone (GH) (Tuca et al., 2013), hydrazine
tioned the value of MA, stating that weight gain as a result of MA may sulfate (Chlebowski et al., 1990; Kosty et al., 1994), metoclopramide
only be the result of increased body fat and fluid retention without (Chen et al., 1997), olanzapine (Naing et al., 2015), and bortezomib
significant improvement in lean body mass (Madeddu et al., 2009; (Jatoi et al., 2005). It should be noted that such drugs with good ra-
Perez De Oteyza et al., 1998). Nevertheless, extensive studies have been tionale might still prove effective since, as was mentioned before, the
conducted on a broad range of compounds in both clinical and heterogeneity and small size of study groups limit the confidence

96
Table 6.1
Pharmacologic treatment options for cancer cachexia in clinical research phase.
Drug Proposed mechanism of action Observed improvements Drug category Shortcomings
Confirmed efficacy
M. Sadeghi et al.

Megestrol acetate Reduction and inhibition of pro-inflammatory Appetite, food intake, weight gain, Progestogens Increased risk of thromboembolic events (most
Medroxyprogesterone acetate (MPA) cytokines (Mantovani et al., 1995) and appetite quality of life (Lesniak et al., 2008; important), peripheral
stimulation through increasing the release of Pascual Lopez et al., 2004; Garcia et al., edema, breakthrough bleeding, hyperglycemia,
neuropeptide Y in the hypothalamus (McCarthy 2013) hypertension,
et al., 1994) and Cushing’s syndrome (Maccio et al., 2012a;
Marshall, 2003)
Cannabinoids (e.g., dronabinol) Interaction with endorphin receptors, interference Appetite, food Cannabinoids Severe adverse effects, especially on the central
with IL-1 synthesis, activation of cannabinoid intake, homeostatic mechanisms of nervous system, e.g., hallucinations, vertigo,
receptors involved in the neuronal circuit of leptin energy storage (Gamage and Lichtman, psychosis (Bagshaw and Hagen, 2002; Tafelski
and prostaglandin synthesis inhibition. 2012) et al., 2016)
.
Based on the current literature, evidence for the
use of cannabinoids for the treatment of cancer-
related cachexia-anorexia syndrome remains
equivocal (Reuter and Martin, 2016; Cannabis In
Cachexia Study, Group et al., 2006; Jatoi et al.,
2002).
Cyproheptadine Serotonergic blockade Mild stimulation of appetite (Kardinal Antiserotonergic drugs Sedating effects (Mantovani et al., 2001)
et al., 1990) Further clinical trials are needed following
inconsistent results (Kardinal et al., 1990; Couluris
et al., 2008).
Non-steroidal anti-inflammatory drugs (NSAIDs), Reduction of tumor-associated inflammation Lean body mass, TNF-α levels, grip Cytokine inhibitors Risk of renal and hepatic impairment and gastro-
including cyclooxygenase-2 (COX-2) inhibitors strength, quality of life, Glasgow intestinal bleeding (NSAIDs) (Reid et al., 2013)
such as celecoxib, ibuprofen, and indomethacin prognostic score (Mantovani et al., The initial results are positive (Maccio et al.,

97
2010; McMillan et al., 1999; Lai et al., 2012b); however, they are not sufficient to
2008) recommend widespread use outside of clinical
trials (Reid et al., 2013; Solheim et al., 2013).
Thalidomide Immunomodulation, anti-inflammatory and TNF-α Appetite, lean body mass, weight Cytokine inhibitors Poor tolerability in esophageal cancer (Wilkes
and IL-6 inhibition properties, inhibition of NF-κB (Gordon et al., 2005), quality of life et al., 2011)
(Jin et al., 2002; Keifer et al., 2001), and blockade of (Davis et al., 2012) Larger clinical trials are needed based on
COX-2 (Fujita et al., 2001) promising initial results (Mantovani, 2010;
Yennurajalingam et al., 2012; Wen et al., 2012).
Melatonin Cytokine and TNF-α inhibition (Lissoni et al., 1996) Contradicting results (Lissoni et al., Cytokine inhibitors More clinical trials specifically designed for the
1996; Del Fabbro et al., 2013) study of the effect of this hormone are needed.
Phase 3 clinical trials on melatonin have been
conducted without observation of any
improvement (NCT0051337).
Ghrelin (and the agonists of its receptor, anamorelin Stimulation of GH secretion (Takaya et al., 2000), Body weight, appetite, lean body mass Ghrelin might stimulate tumor growth (Murphy
and macimorelin) suppression of pro-inflammatory cytokines and (Neary et al., 2004; Garcia et al., 2007; and Lynch, 2012).
inhibition of NF-κB (Waseem et al., 2008; Li et al., Garcia et al., 2015) Confirmation requires further clinical research.
2004), orexigenic effects (Kamegai et al., 2001; Phase 3 clinical trials
Cowley et al., 2003) have been conducted (NCT01395914,
NCT01387282).
Selective Androgen Receptor Modulators (e.g., Act selectively on androgen receptors of the skeletal Physical performance and lean body Selective androgen receptor Phase 3 clinical trials have been conducted
enobosarm) muscle and bone, minimizing stimulation of other mass (Dalton et al., 2011; Dobs et al., modulators (SARMs) (NCT01355484, NCT01355497).
organs such as prostate, skin, and liver. 2013)
Espindolol Non-selective β blocker with central 5-HT1a and Lean body mass, weight, hand grip Non-selective β blocker with More efficacy and safety data needed but the
partial β2 receptor agonist effects strength (Stewart Coats et al., 2016) central 5-HT1a and partial β2 initial data have been promising (Stewart Coats
receptor agonist effects et al., 2016).
Oxymetholone, oxandrolone, nandrolone and Marked anabolic activity with minimal androgenic Lean body mass, subjective anorexia Synthetic anabolic steroids
fluoxymesterone effects (Lesser et al., 2006) score (Lesser et al., 2006)
(continued on next page)
Critical Reviews in Oncology / Hematology 127 (2018) 91–104
Table 6.1 (continued)

Drug Proposed mechanism of action Observed improvements Drug category Shortcomings


Confirmed efficacy
M. Sadeghi et al.

High hepatotoxicity (García-Cortés et al., 2016)


Confirmatory data are still lacking and have
sometimes been shown to be inferior compared to
other treatments (Loprinzi et al., 1999). Most
studies have been conducted in patients with
cachexia of a non-oncological origin (Storer et al.,
2005).
Pentoxifylline (a methylxanthine derivative) Anti-inflammatory and TNF-α inhibition properties Not significant (Goldberg et al., 1995; Cytokine inhibitors Its efficacy in cancer-associated cachexia has not
(immunomodulation through inhibition of Mehrzad et al., 2016) been demonstrated (Goldberg et al., 1995;
phosphodiesterase) Mehrzad et al., 2016).
Dexamethasone Suppression of pro-inflammatory cytokines such as Appetite, calorie intake, sensation of Corticosteroids The effect is short lived (less than 4 weeks).
Prednisone TNF-α (Han et al., 1990) and IL-1 (Uehara et al., wellbeing, and nausea (Bruera et al., Furthermore, long-term side effects (insulin
Methylprednisone 1989) 1985; Della Cuna et al., 1989; Willox resistance, fluid retention, steroidal myopathy,
et al., 1984) skin fragility, adrenal insufficiency, sleep and
cognitive disorders) have been observed (Tuca
et al., 2013).
Clinical trials have been conducted
(Yennurajalingam et al., 2012; Goldberg et al.,
1995).

98
Table 6.2
Pharmacologic treatment options for cancer cachexia in preclinical research phase.
Drug Proposed mechanism of action Observed improvements Drug category Shortcomings
Confirmed efficacy

Clenbuterol, salbutamol, salmeterol, and Increase muscle mass through inhibition of protein degradation Muscle mass and function (Busquets et al., Beta-2-agonists They might have cardiovascular side effects (Toledo et al.,
formoterol (Benson et al., 1991) and activation of muscle protein synthesis 2004; Greig et al., 2014; Toledo et al., 2014; Jeppsson et al., 1986).
(Choo et al., 1992); activation of the AKT/mTOR pathway (Kline 2016) Based on the initial promising results in animal models (Pinto
et al., 2007) et al., 2004), randomized trials are warranted.
Insulin and insulin sensitizers Counter the peripheral insulin resistance that is frequently Calorie intake, body fat (Lundholm et al., Insulin and TZDs have proven to be promising agents against cancer-
(Thiazolidinediones such as associated with cancer cachexia. Thiazolidinediones decrease 2007), and lean body mass (Yki-Jarvinen, insulin sensitizers associated cachexia in animal models (Chen and Xiao, 2014;
rosiglitazone) pro-inflammatory cytokines and increase adiponectin and also 2004; Chen and Xiao, 2014) Asp et al., 2011). Clinical trials in human cancer patients
stimulate peroxisome proliferator-activated receptor (PPAR γ) present an interesting area for future research.
(Hauner, 2002).
Metformin Increases the activity of AMP-activated protein kinase (AMPK), Body composition, protein degradation Insulin and Clinical studies are yet to be conducted.
and the PI3K pathway, in muscle cells, 2 mechanisms involved in (Oliveira and Gomes-Marcondes, 2016) insulin sensitizers
muscle wasting (Mondello et al., 2015).
Critical Reviews in Oncology / Hematology 127 (2018) 91–104
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

associated with the conclusion of each trial. Therefore, it is best to in- loss (Alves et al., 2015). In human cancer patients and survivors, ex-
tegrate these drugs into multimodal approaches in order to better de- ercise leads to a wide range of improvements, including decreased in-
termine their effectiveness (Solheim and Laird, 2012). Recently flammation and oxidative stress, reduced depressive symptoms, and
emerged drug categories such as ruxolitinib, a JAK/STAT3 inhibitor, increased anabolic hormones such as IGF-1 (Lira et al., 2015). In ad-
and BYM338, a myostatin/activin inhibitor (Novartis Pharmaceuticals, dition, in a randomized controlled trial, Oldervoll et al. found that
East Hanover, NJ, USA), should also be considered. exercise could maintain physical performance as measured by the
shuttle walk and hand grip tests (Oldervoll et al., 2011). Nevertheless,
4.2. Nutritional treatment several considerations must be made before accepting physical activity
as a feasible therapeutic approach. Cancer patients regularly suffer from
Cancer patients often have reduced calorie intakes (Bosaeus et al., chronic fatigue and thus have a diminished capacity for full time ex-
2001; Hutton et al., 2006; Bovio et al., 2008). This could be the direct ercise regiments (Argiles et al., 2012). This is further demonstrated by
result of the presence of the tumor (e.g., obstruction of the gastro- the high drop-out rates from clinical trials including exercise
intestinal tract) or the host’s response to the tumor and treatment. (Maddocks et al., 2011). In addition, patient condition should be con-
Systemic inflammation due to the tumor may lead to anorexia while sidered before prescription of exercise. For example, acute physical
nausea, mucositis, and vomiting as a result of anticancer therapy could activity must be avoided in anemic cancer patients (Argiles et al.,
severely decrease food intake (Nicolini et al., 2013). Therefore, it is of 2012). Another consideration in the prescription of exercise is the re-
utmost importance to reverse these symptoms when possible. In addi- lative efficacies of aerobic and resistance training. Although some stu-
tion, the deficiency in calorie intake should be compensated assuming dies have favored resistance training (Courneya et al., 2007; Schwartz
total energy expenditure in cancer patients to be equal to healthy and Winters-Stone, 2009), the general data is inconclusive. This must
controls (Arends et al., 2017). Other aspects of nutritional intervention not prevent, however, the assignment of training programs containing
include dietary counseling, nutritional supplementation, and artificial both types of physical activity. Overall, exercise has shown positive
nutrition. Nutritional counseling must be done, preferably by a pro- effects mostly in early stages of cancer and sufficient data on patients in
fessional dietician, through calculation of energy and nutritional re- advanced stages of disease is lacking (Stene et al., 2013; Navigante and
quirements (Arends et al., 2017). It should be noted, however, that the Morgado, 2016). Since cancer patients usually experience cachexia at
value of dietary counseling as an intervention has been assessed in such advanced stages, it is difficult to interpret these data in the context
several studies with heterogeneous results. Therefore, further evidence of cancer cachexia. Clearly, as is common with other aspects of cancer
is needed to prove this intervention as a means of improving weight cachexia, there have been too few robust randomized controlled trials
gain (Balstad et al., 2014). At this stage, recommendations include to determine the best therapeutic approach using physical activity and,
dietary changes, such as intake of an energy- and protein-rich diet, and therefore, this presents a possible area for future research.
lifestyle changes, such as increasing meal frequency and decreasing
meal size. If these changes could not satisfy the patient’s energy re- 4.4. Psychosocial treatment
quirement, oral supplementations may be offered. Several compounds,
notably, β-hydroxy-β-methylbutyrate (HMB) (Aversa et al., 2011; As was true for the assessment of the psychosocial aspect of ca-
Fitschen et al., 2013), eicosapentaenoic acid (Murphy et al., 2011; chexia, management of these symptoms has been much neglected in
Fearon et al., 2006b; Sanchez-Lara et al., 2014), and L-carnitine many healthcare settings. A unique feature of this therapeutic modality
(Gramignano et al., 2006; Kraft et al., 2012) have been shown in some is that the patient’s caregivers should also be included in the inter-
trials to positively affect cachectic patients; however, insufficient data ventions (Hudson and Payne, 2011). With the growing importance of
exist for recommending HMB and L-carnitine for medical use (Arends home care, fiscal and psychosocial support of family caregivers would
et al., 2017). The therapeutic potential of omega-3 fatty acids for cancer lead to better quality of care (Reid, 2014). The negativity of the emo-
cachexia has been widely studied. The results have been inconclusive tions experienced by the patient and caregivers is inversely propor-
thus far (Dewey et al., 2007; Mazzotta and Jeney, 2009; Ries et al., tional to acceptance of the situation and the effectiveness of their
2012; Bruera et al., 2003); however, recent studies with better study coping strategies (Oberholzer et al., 2013). As a result, it has been
design have been in favor of their use (Ryan et al., 2009; Weed et al., proposed to base psychosocial interventions on Lazarus and Folkman’s
2011; van der Meij et al., 2010). These results, in addition to mild side Coping and Adaptation Theory (Reid, 2014; Lazarus and Folkman,
effects and sound biological rationale, support the use of these fatty 1984). Currently, Reid et al. are conducting a clinical trial to test a new
acids as nutritional supplements (Arends et al., 2017). If patients be- psychoeducation intervention which focuses on practicality as well as
come unable to eat adequately for a long time, artificial nutrition will the aforementioned concepts (Reid et al., 2014).
become necessary (Arends et al., 2017). Even though numerous studies
have attempted to compare enteral and parenteral nutrition with no 5. Conclusion
conclusive result, it is now generally accepted that each mode of de-
livery has its unique indications and contraindications, thus rendering With the growing understanding of cancer cachexia, it has become a
comparisons between them superfluous. It is widely accepted that the more important focus of research. Numerous trials have been conducted
enteral route should have priority, while parenteral nutrition could be towards the development of better treatment options and various
applied when the enteral route is not sufficient or feasible (Arends treatment guidelines have been proposed (Arends et al., 2017).
et al., 2017; Cotogni, 2016). Nevertheless, several elements have frequently limited the applicability
of their results. Individually, many studies suffer from the small size
4.3. Exercise treatment and heterogeneity of their patient samples. These factors contribute to
the contradictory results observed in different trials. In addition, dif-
One of the main goals of cancer cachexia therapy is preservation of ferences in inclusion criteria used for generating patient samples have
muscle mass and physical performance. A very good rationale supports made comparing study results even more difficult. Consequently, pro-
exercise as a therapeutic approach to cachexia (Maddocks et al., 2013; posing an international standard guideline treatment has proven to be
Stene et al., 2013). Exercise has an anti-inflammatory effect (Petersen impossible so far. It is clear that constructing a standard study protocol
and Pedersen, 2005) and could counter the inflammation-induced for cancer cachexia, which would unify inclusion criteria and endpoints
muscle degradation and insulin resistance (Gould et al., 2013). Fur- in all trials, would bear immense benefits. Conduction of better de-
thermore, experiments with animal models have shown the effect of signed trials using these criteria would enable the data to be better
exercise on decreasing skeletal muscle degradation and body weight compared and integrated, thus paving the way towards a universally

99
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

accepted standard clinical approach. Thereafter, this standard guideline Blauwhoff-Buskermolen, S., et al., 2014. ’Pre-cachexia’: a non-existing phenomenon in
would form the baseline to which every other regiment should be cancer? Ann. Oncol. 25 (8), 1668–1669.
Blum, D., Strasser, F., 2011. Cachexia assessment tools. Curr. Opin. Support Palliat. Care 5
compared. Such a setting would provide a much more efficient foun- (4), 350–355.
dation for future research and much faster progress towards the best Blum, D., et al., 2014. Validation of the consensus-definition for cancer cachexia and
possible treatment for cancer cachexia. Standardization would be evaluation of a classification model–a study based on data from an international
multicentre project (EPCRC-CSA). Ann. Oncol. 25 (8), 1635–1642.
highly dependent upon the definition of cachexia (inclusion criteria) Bo, Y., et al., 2015. Preoperative nutritional risk index to predict postoperative survival
and its assessment methods (endpoints), which as noted before, remain time in primary liver cancer patients. Asia Pac. J. Clin. Nutr. 24 (4), 591–597.
controversial matters in the field. Therefore, it seems that while great Bosaeus, I., et al., 2001. Dietary intake and resting energy expenditure in relation to
weight loss in unselected cancer patients. Int. J. Cancer 93 (3), 380–383.
effort is currently being directed towards formulation of novel man- Bovio, G., et al., 2008. Evaluation of nutritional status and dietary intake in patients with
agement techniques for cancer cachexia, one could expect only a finite advanced cancer on palliative care. Minerva Gastroenterol Dietol 54 (3), 243–250.
improvement in current prevailing management regiments. A greater Bozzetti, F., 2013. Nutritional status, cachexia and survival in patients with advanced
colorectal carcinoma. Different assessment criteria for nutritional status provide
focus on the definition and assessment of cancer cachexia in order to
unequal results. Clin. Nutr. 32 (5), 876.
finally formulate widely accepted international consensuses is needed Bozzetti, F., Bozzetti, V., 2013. Is the intravenous supplementation of amino acid to
as the first step towards a standard guideline. cancer patients adequate? A critical appraisal of literature. Clin. Nutr 32 (1),
142–146.
Bozzetti, F., Mariani, L., 2009. Defining and classifying cancer cachexia: a proposal by the
Conflict of Interest SCRINIO Working Group. JPEN J. Parenter. Enter. Nutr 33 (4), 361–367.
Bruera, E., et al., 1985. Action of oral methylprednisolone in terminal cancer patients: a
The authors declare that they have no conflict of interest. prospective randomized double-blind study. Cancer Treat. Rep. 69 (7-8), 751–754.
Bruera, E., et al., 2003. Effect of fish oil on appetite and other symptoms in patients with
advanced cancer and anorexia/cachexia: a double-blind, placebo-controlled study. J.
References Clin. Oncol. 21 (1), 129–134.
Burney, B.O., et al., 2012. Low testosterone levels and increased inflammatory markers in
patients with cancer and relationship with cachexia. J. Clin. Endocrinol. Metab. 97
Aaronson, N.K., et al., 1993. The European Organization for Research and Treatment of
(5), E700–9.
Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in
Busquets, S., et al., 2004. Anticachectic effects of formoterol: a drug for potential treat-
oncology. J. Natl. Cancer Inst. 85 (5), 365–376.
ment of muscle wasting. Cancer Res. 64 (18), 6725–6731.
Alves, C.R., et al., 2015. Aerobic exercise training as therapy for cardiac and cancer ca-
Bye, A., et al., 2016. Alterations in inflammatory biomarkers and energy intake in cancer
chexia. Life Sci. 125, 9–14.
cachexia: a prospective study in patients with inoperable pancreatic cancer. Med.
Ambrus, J.L., et al., 1975. Causes of death in cancer patients. J. Med. 6 (1), 61–64.
Oncol. 33 (6), 54.
Ando, K., et al., 2014. Tocilizumab, a proposed therapy for the cachexia of Interleukin6-
Cannabis In Cachexia Study, Group, et al., 2006. Comparison of orally administered
expressing lung cancer. PLoS One 9 (7), e102436.
cannabis extract and delta-9-tetrahydrocannabinol in treating patients with cancer-
Ando, K., et al., 2013. Possible role for tocilizumab, an anti–interleukin-6 receptor anti-
related anorexia-cachexia syndrome: a multicenter, phase III, randomized, double-
body, in treating cancer cachexia. J. Clin. Oncol. 31 (6), e69–e72.
blind, placebo-controlled clinical trial from the Cannabis-In-Cachexia-Study-Group.
Arends, J., et al., 2017. ESPEN guidelines on nutrition in cancer patients. Clin. Nutr. 36
J. Clin. Oncol. 24 (21), 3394–3400.
(1), 11–48.
Cederholm, T., et al., 2015. Diagnostic criteria for malnutrition - an ESPEN consensus
Argiles, J.M., et al., 2005. Molecular mechanisms involved in muscle wasting in cancer
statement. Clin. Nutr. 34 (3), 335–340.
and ageing: cachexia versus sarcopenia. Int. J. Biochem. Cell. Biol. 37 (5),
Chamberlain, K., 2004. Food and health: expanding the agenda for health psychology. J.
1084–1104.
Health Psychol. 9 (4), 467–481.
Argiles, J.M., et al., 2009. The role of cytokines in cancer cachexia. Curr. Opin. Support
Chang, V.T., Xia, Q., Kasimis, B., 2005. The functional assessment of Anorexia/Cachexia
Palliat. Care 3 (4), 263–268.
therapy (FAACT) appetite scale in veteran cancer patients. J. Support Oncol. 3 (5),
Argiles, J.M., et al., 2014. Cancer cachexia: understanding the molecular basis. Nat. Rev.
377–382.
Cancer 14 (11), 754–762.
Chasen, M., Hirschman, S.Z., Bhargava, R., 2011. Phase II study of the novel peptide-
Argiles, J.M., et al., 2011. The cachexia score (CASCO): a new tool for staging cachectic
nucleic acid OHR118 in the management of cancer-related anorexia/cachexia. J. Am.
cancer patients. J. Cachexia Sarcopenia Muscle 2 (2), 87–93.
Med. Dir. Assoc. 12 (1), 62–67.
Argiles, J.M., et al., 2010. Optimal management of cancer anorexia-cachexia syndrome.
Chen, S.Z., Xiao, J.D., 2014. Rosiglitazone and imidapril alone or in combination alleviate
Cancer Manag Res. (2), 27–38.
muscle and adipose depletion in a murine cancer cachexia model. Tumour Biol. 35
Argiles, J.M., et al., 2012. Are there any benefits of exercise training in cancer cachexia?
(1), 323–332.
J. Cachexia Sarcopenia Muscle 3 (2), 73–76.
Chen, H.C., et al., 1997. Effect of megestrol acetate and prepulsid on nutritional im-
Asp, M.L., et al., 2011. Rosiglitazone delayed weight loss and anorexia while attenuating
provement in patients with head and neck cancers undergoing radiotherapy.
adipose depletion in mice with cancer cachexia. Cancer Biol. Ther. 12 (11), 957–965.
Radiother. Oncol. 43 (1), 75–79.
Aversa, Z., et al., 2011. Beta-hydroxy-beta-methylbutyrate (HMB) attenuates muscle and
Chlebowski, R.T., et al., 1990. Hydrazine sulfate influence on nutritional status and
body weight loss in experimental cancer cachexia. Int. J. Oncol. 38 (3), 713–720.
survival in non-small-cell lung cancer. J. Clin. Oncol. 8 (1), 9–15.
Bachmann, J., et al., 2008. Cachexia worsens prognosis in patients with resectable pan-
Choo, J.J., et al., 1992. Anabolic effects of clenbuterol on skeletal muscle are mediated by
creatic cancer. J. Gastrointest. Surg. 12 (7), 1193–1201.
beta 2-adrenoceptor activation. Am. J. Physiol. 263 (Pt (1)), E50–E56.
Bagshaw, S.M., Hagen, N.A., 2002. Medical efficacy of cannabinoids and marijuana: a
Chow, E., et al., 2005. Accuracy of survival prediction by palliative radiation oncologists.
comprehensive review of the literature. J. Palliat. Care 18 (2), 111–122.
Int. J. Radiat. Oncol.*Biol.*Phys. 61 (3), 870–873.
Balstad, T.R., et al., 2014. Dietary treatment of weight loss in patients with advanced
Churm, D., et al., 2009. A questionnaire study of the approach to the anorexia-cachexia
cancer and cachexia: a systematic literature review. Crit. Rev. Oncol. Hematol. 91 (2),
syndrome in patients with cancer by staff in a district general hospital. Support Care
210–221.
Cancer 17 (5), 503–507.
Barbat-Artigas, S., et al., 2012. How to Assess Functional Status: A New Muscle Quality
Correa-de-Araujo, R., et al., 2017. The need for standardized assessment of muscle quality
index, vol. 16. pp. 67–77.
in skeletal muscle function deficit and other aging-related muscle dysfunctions: a
Batista Jr., M.L., et al., 2013. Adipose tissue-derived factors as potential biomarkers in
symposium report. Front. Physiol. (8), 87.
cachectic cancer patients. Cytokine 61 (2), 532–539.
Cotogni, P., 2016. Enteral versus parenteral nutrition in cancer patients: evidences and
Bauer, J., Capra, S., 2003. Comparison of a malnutrition screening tool with subjective
controversies. Ann. Palliat. Med. 5 (1), 42–49.
global assessment in hospitalised patients with cancer–sensitivity and specificity. Asia
Couluris, M., et al., 2008. The effect of cyproheptadine hydrochloride (periactin) and
Pac. J. Clin. Nutr. 12 (3), 257–260.
megestrol acetate (megace) on weight in children with cancer/treatment-related
Baumgartner, R.N., et al., 1998. Epidemiology of sarcopenia among the elderly in New
cachexia. J. Pediatr. Hematol. Oncol. 30 (11), 791–797.
Mexico. Am. J. Epidemiol. 147 (8), 755–763.
Courneya, K.S., et al., 2007. Effects of aerobic and resistance exercise in breast cancer
Bayer, L.M., Bauers, C.M., Kapp, S.R., 1983. Psychosocial aspects of nutritional support.
patients receiving adjuvant chemotherapy: a multicenter randomized controlled trial.
Nurs. Clin. North Am. 18 (1), 119–128.
J. Clin. Oncol. 25 (28), 4396–4404.
Bektas, H.A., Akdemir, N., 2008. Reliability and validity of the functional living index-
Cowley, M.A., et al., 2003. The distribution and mechanism of action of ghrelin in the
cancer in Turkish cancer patients. Cancer Nurs. 31 (1), E1–7.
CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis.
Belizario, J.E., et al., 1991. Bioactivity of skeletal muscle proteolysis-inducing factors in
Neuron 37 (4), 649–661.
the plasma proteins from cancer patients with weight loss. Br. J. Cancer 63 (5),
Dallmann, R., et al., 2011. The orally active melanocortin-4 receptor antagonist BL-6020/
705–710.
979: a promising candidate for the treatment of cancer cachexia. J. Cachexia
Benson, D.W., et al., 1991. Decreased myofibrillar protein breakdown following treat-
Sarcopenia Muscle 2 (3), 163–174.
ment with clenbuterol. J. Surg. Res. 50 (1), 1–5.
Dalton, J.T., et al., 2011. The selective androgen receptor modulator GTx-024 (en-
Berti, A., et al., 2013. Assessment of tocilizumab in the treatment of cancer cachexia. J.
obosarm) improves lean body mass and physical function in healthy elderly men and
Clin. Oncol. 31 (23), 2970.
postmenopausal women: results of a double-blind, placebo-controlled phase II trial. J.
Bilir, C., et al., 2015. The prognostic role of inflammation and hormones in patients with
Cachexia Sarcopenia Muscle 2 (3), 153–161.
metastatic cancer with cachexia. Med. Oncol. 32 (3), 56.
Davis, M., et al., 2012. A phase II dose titration study of thalidomide for cancer-associated

100
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

anorexia. J. Pain Symptom Manage 43 (1), 78–86. with prognosis. Lung Cancer 74 (3), 516–520.
Del Fabbro, E., et al., 2013. Effects of melatonin on appetite and other symptoms in Gioulbasanis, I., et al., 2011b. Baseline nutritional evaluation in metastatic lung cancer
patients with advanced cancer and cachexia: a double-blind placebo-controlled trial. patients: mini nutritional assessment versus weight loss history. Ann. Oncol. 22 (4),
J. Clin. Oncol. 31 (10), 1271–1276. 835–841.
Della Cuna, G.R., Pellegrini, A., Piazzi, M., 1989. Effect of methylprednisolone sodium Goldberg, R.M., et al., 1995. Pentoxifylline for treatment of cancer anorexia and ca-
succinate on quality of life in preterminal cancer patients: a placebo-controlled, chexia? A randomized, double-blind, placebo-controlled trial. J. Clin. Oncol. 13 (11),
multicenter study. The Methylprednisolone Preterminal Cancer Study Group. Eur. J. 2856–2859.
Cancer Clin. Oncol. 25 (12), 1817–1821. Gordon, J.N., et al., 2005. Thalidomide in the treatment of cancer cachexia: a randomised
Deutsch, J., Kolhouse, J.F., 2004. Assessment of gastrointestinal function and response to placebo controlled trial. Gut 54 (4), 540–545.
megesterol acetate in subjects with gastrointestinal cancers and weight loss. Support Gorenc, M., Kozjek, N.R., Strojan, 2015. Malnutrition and cachexia in patients with head
Care Cancer 12 (7), 503–510. and neck cancer treated with (chemo)radiotherapy. Rep. Pract. Oncol. Radiother. 20
Deutz, N.E., et al., 2011. Muscle protein synthesis in cancer patients can be stimulated (4), 249–258.
with a specially formulated medical food. Clin. Nutr. (Edinb., Scotl.) 30 (6), 759–768. Gould, D.W., et al., 2013. Cancer cachexia prevention via physical exercise: molecular
Dev, R., et al., 2014. Association between hypogonadism, symptom burden, and survival mechanisms. J. Cachexia Sarcopenia Muscle 4 (2), 111–124.
in male patients with advanced cancer. Cancer 120 (10), 1586–1593. Gramignano, G., et al., 2006. Efficacy of l-carnitine administration on fatigue, nutritional
Dewey, A., et al., 2007. Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) status, oxidative stress, and related quality of life in 12 advanced cancer patients
for the treatment of cancer cachexia. Cochrane Database Syst. Rev.(1) Cd004597. undergoing anticancer therapy. Nutrition 22 (2), 136–145.
Dewys, W.D., et al., 1980. Prognostic effect of weight loss prior to chemotherapy in Greig, C.A., et al., 2014. Phase I/II trial of formoterol fumarate combined with megestrol
cancer patients. Eastern Cooperative Oncology Group. Am J. Med. 69 (4), 491–497. acetate in cachectic patients with advanced malignancy. Support Care Cancer 22 (5),
Dillon, E.L., et al., 2007. Amino acid metabolism and inflammatory burden in ovarian 1269–1275.
cancer patients undergoing intense oncological therapy. Clin. Nutr. 26 (6), 736–743. Grossberg, A.J., Scarlett, J.M., Marks, D.L., 2010. Hypothalamic mechanisms in cachexia.
Dobs, A.S., et al., 2013. Effects of enobosarm on muscle wasting and physical function in Physiol Behav. 100 (5), 478–489.
patients with cancer: a double-blind, randomised controlled phase 2 trial. Lancet Grunfeld, C., et al., 1996. Endotoxin and cytokines induce expression of leptin, the ob
Oncol. 14 (4), 335–345. gene product, in hamsters. J. Clin. Invest. 97 (9), 2152–2157.
Dodson, S., et al., 2011. Muscle wasting in cancer cachexia: clinical implications, diag- Guigoz, Y., Vellas, B., Garry, J., 1996. Assessing the nutritional status of the elderly: the
nosis, and emerging treatment strategies. Annu. Rev. Med. 62, 265–279. mini nutritional assessment as part of the geriatric evaluation. Nutr. Rev. 54 (Pt (1)),
Donohoe, C.L., Ryan, A.M., Reynolds, J.V., 2011. Cancer cachexia: mechanisms and S59–65.
clinical implications. Gastroenterol. Res. Pract. 2011, 601434. Han, J., Thompson, Beutler, B., 1990. Dexamethasone and pentoxifylline inhibit en-
Elia, M., Stratton, R.J., 2012. An analytic appraisal of nutrition screening tools supported dotoxin-induced cachectin/tumor necrosis factor synthesis at separate points in the
by original data with particular reference to age. Nutrition 28 (5), 477–494. signaling pathway. J. Exp. Med. 172 (1), 391–394.
Erlandson, M.C., et al., 2016. Muscle analysis using pQCT, DXA and MRI. Eur. J. Radiol. Hauner, H., 2002. The mode of action of thiazolidinediones. Diabetes Metab. Res. Rev. 18
85 (8), 1505–1511. (Suppl. (2)), S10–S15.
EuroQol, 1990. EuroQol–a new facility for the measurement of health-related quality of Hirai, K., et al., 1998. Biological evaluation of a lipid-mobilizing factor isolated from the
life. Health Policy 16 (3), 199–208. urine of cancer patients. Cancer Res. 58 (11), 2359–2365.
Evans, W.J., et al., 2008. Cachexia: a new definition. Clin. Nutr. 27 (6), 793–799. Hong, N., et al., 2016. Serum PTHrP predicts weight loss in cancer patients independent
Faggioni, R., et al., 1997. LPS-induced anorexia in leptin-deficient (ob/ob) and leptin of hypercalcemia, inflammation, and tumor burden. J. Clin. Endocrinol. Metab. 101
receptor-deficient (db/db) mice. Am. J. Physiol. 273 (Pt. (1)), R181–R186. (3), 1207–1214.
Fearon, K.C., 2008. Cancer cachexia: developing multimodal therapy for a multi- Hopkinson, J.B., 2014. Psychosocial impact of cancer cachexia. J. Cachexia Sarcopenia
dimensional problem. Eur. J. Cancer 44 (8), 1124–1132. Muscle 5 (2), 89–94.
Fearon, K., et al., 2011. Definition and classification of cancer cachexia: an international Hopkinson, J., Corner, J., 2006. Helping patients with advanced cancer live with concerns
consensus. Lancet Oncol. 12 (5), 489–495. about eating: a challenge for palliative care professionals. J. Pain Symptom Manage.
Fearon, K., Arends, J., Baracos, V., 2013. Understanding the mechanisms and treatment 31 (4), 293–305.
options in cancer cachexia. Nat. Rev. Clin. Oncol. 10 (2), 90–99. Hopkinson, J.B., et al., 2010. The deliverability, acceptability, and perceived effect of the
Fearon, K.C., et al., 1999. Pancreatic cancer as a model: inflammatory mediators, acute- Macmillan approach to weight loss and eating difficulties: a phase II, cluster-rando-
phase response, and cancer cachexia. World J. Surg. 23 (6), 584–588. mized, exploratory trial of a psychosocial intervention for weight- and eating-related
Fearon, K.C., Glass, D.J., Guttridge, D.C., 2012. Cancer cachexia: mediators, signaling, distress in people with advanced cancer. J. Pain Symptom Manage. 40 (5), 684–695.
and metabolic pathways. Cell Metab. 16 (2), 153–166. Hopkinson, J., Wright, D., Corner, J., 2006. Exploring the experience of weight loss in
Fearon, K.C., Voss, A.C., Hustead, D.S., 2006a. Definition of cancer cachexia: effect of people with advanced cancer. J. Adv. Nurs. 54 (3), 304–312.
weight loss, reduced food intake, and systemic inflammation on functional status and Huang, Q., et al., 2005. Change of the growth hormone-insulin-like growth factor-I axis in
prognosis. Am. J. Clin. Nutr. 83 (6), 1345–1350. patients with gastrointestinal cancer: related to tumour type and nutritional status.
Fearon, K.C., et al., 2006b. Double-blind, placebo-controlled, randomized study of eico- Br. J. Nutr. 93 (6), 853–858.
sapentaenoic acid diester in patients with cancer cachexia. J. Clin. Oncol. 24 (21), Hudson, P., Payne, S., 2011. Family caregivers and palliative care: current status and
3401–3407. agenda for the future. J. Palliat. Med. 14 (7), 864–869.
Felix, K., et al., 2011. Identification of serum proteins involved in pancreatic cancer ca- Hunt, S.M., et al., 1981. The Nottingham Health Profile: subjective health status and
chexia. Life Sci. 88 (5-6), 218–225. medical consultations. Soc. Sci. Med. A 15 (Pt (3)), 221–229.
Ferguson, M.L., et al., 1999. Validation of a malnutrition screening tool for patients re- Hutton, J.L., et al., 2006. Dietary patterns in patients with advanced cancer: implications
ceiving radiotherapy. Australas. Radiol. 43 (3), 325–327. for anorexia-cachexia therapy. Am. J. Clin. Nutr. 84 (5), 1163–1170.
Ferrans, C.E., 1990. Development of a quality of life index for patients with cancer. Oncol. Inagaki, J., Rodriguez, V., Bodey, G., 1974. Proceedings: causes of death in cancer pa-
Nurs. Forum 17 (Suppl. 3), 15–19 discussion 20-1. tients. Cancer 33 (2), 568–573.
Ferrans, C.E., Powers, M.J., 1992. Psychometric assessment of the quality of life index. Isenring, E.A., Capra, S., Bauer, J.D., 2004. Nutrition intervention is beneficial in on-
Res. Nurs. Health 15 (1), 29–38. cology outpatients receiving radiotherapy to the gastrointestinal or head and neck
Fitschen, J., et al., 2013. Efficacy of beta-hydroxy-beta-methylbutyrate supplementation area. Br. J. Cancer 91 (3), 447–452.
in elderly and clinical populations. Nutrition 29 (1), 29–36. Jacquelin-Ravel, N., Pichard, C., 2012. Clinical nutrition, body composition and on-
Fong, D.Y., et al., 2014. The functional living index-cancer is a reliable and valid in- cology: a critical literature review of the synergies. Crit. Rev. Oncol. Hematol. 84 (1),
strument in Chinese cancer patients. Qual. Life Res. 23 (1), 311–316. 37–46.
Fujita, J., et al., 2001. Thalidomide and its analogues inhibit lipopolysaccharide-mediated Janik, J.E., et al., 1997. Interleukin 1 alpha increases serum leptin concentrations in
iinduction of cyclooxygenase-2. Clin. Cancer Res. 7 (11), 3349–3355. humans. J. Clin. Endocrinol. Metab. 82 (9), 3084–3086.
Fujiwara, Y., et al., 2014. Metabolomics evaluation of serum markers for cachexia and Janssen, I., Heymsfield, S.B., Ross, R., 2002. Low relative skeletal muscle mass (sarco-
their intra-day variation in patients with advanced pancreatic cancer. PLoS One 9 penia) in older persons is associated with functional impairment and physical dis-
(11), e113259. ability. J. Am. Geriatr. Soc. 50 (5), 889–896.
Gallagher, D., et al., 1998. Organ-tissue mass measurement allows modeling of REE and Jatoi, A., et al., 2010. A placebo-controlled, double-blind trial of infliximab for cancer-
metabolically active tissue mass. Am. J. Physiol. 275 (Pt (2)), E249–58. associated weight loss in elderly and/or poor performance non-small cell lung cancer
Gamage, T.F., Lichtman, A.H., 2012. The endocannabinoid system: role in energy reg- patients (N01C9). Lung Cancer 68 (2), 234–239.
ulation. Pediatr. Blood Cancer 58 (1), 144–148. Jatoi, A., et al., 2005. Is bortezomib, a proteasome inhibitor, effective in treating cancer-
Ruiz Garcia, V., et al., 2013. Megestrol acetate for treatment of anorexia-cachexia syn- associated weight loss? Preliminary results from the North Central Treatment Group.
drome. Cochrane Database Syst. Rev.(3) Cd004310. Support Care Cancer 13 (6), 381–386.
Garcia, J., et al., 2007. A phase II randomized, placebo-controlled, double-blind study of Jatoi, A., et al., 2002. Dronabinol versus megestrol acetate versus combination therapy
the efficacy and safety of RC-1291 (RC) for the treatment of cancer cachexia. J. Clin. for cancer-associated anorexia: a North Central Cancer Treatment Group study. J.
Oncol. (Meet. Abstr.) 25 (Suppl. (18)), 9133. Clin. Oncol. 20 (2), 567–573.
Garcia, J.M., et al., 2015. Anamorelin for patients with cancer cachexia: an integrated Jeppsson, A.B., Waldeck, B., Widmark, E., 1986. Further studies on the cardiomegaly
analysis of two phase 2, randomised, placebo-controlled, double-blind trials. Lancet induced by beta-adrenoceptor agonists. Acta Pharmacol. Toxicol. (Copenh.) 58 (2),
Oncol. 16 (1), 108–116. 121–125.
García-Cortés, M., et al., 2016. Hepatotoxicity by dietary supplements: a tabular listing Jetté, M., Management, C.T., Consultant, and, 1983. Guide for Anthropometric
and clinical characteristics. Int. J. Mol. Sci. 17 (4). Measurements of Canadian Adults. C.T. Management & Consultant, Montréal.
Gioulbasanis, I., et al., 2011a. Mini nutritional assessment (MNA) and biochemical Jin, S.H., et al., 2002. Thalidomide suppresses the interleukin 1beta-induced NFkappaB
markers of cachexia in metastatic lung cancer patients: interrelations and associations signaling pathway in colon cancer cells. Ann. N. Y. Acad. Sci. 973, 414–418.

101
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Kamegai, J., et al., 2001. Chronic central infusion of ghrelin increases hypothalamic Maltoni, M., et al., 2005. Prognostic factors in advanced cancer patients: evidence-based
neuropeptide Y and Agouti-related protein mRNA levels and body weight in rats. clinical recommendations–a study by the Steering Committee of the European
Diabetes 50 (11), 2438–2443. Association for Palliative Care. J. Clin. Oncol. 23 (25), 6240–6248.
Kardinal, C.G., et al., 1990. A controlled trial of cyproheptadine in cancer patients with Mantovani, G., 2010. Randomised phase III clinical trial of 5 different arms of treatment
anorexia and/or cachexia. Cancer 65 (12), 2657–2662. on 332 patients with cancer cachexia. Eur. Rev. Med. Pharmacol. Sci. 14 (4),
Keifer, J.A., et al., 2001. Inhibition of NF-kappa B activity by thalidomide through sup- 292–301.
pression of IkappaB kinase activity. J. Biol. Chem. 276 (25), 22382–22387. Mantovani, G., et al., 1995. Megestrol acetate in neoplastic anorexia/cachexia: clinical
Kemik, O., et al., 2010. The relationship among acute-phase response proteins, cytokines evaluation and comparison with cytokine levels in patients with head and neck
and hormones in cachectic patients with colon cancer. World J. Surg. Oncol. (8), 85. carcinoma treated with neoadjuvant chemotherapy. Int. J. Clin. Lab Res. 25 (3),
Kerem, M., et al., 2008. Adipokines and ghrelin in gastric cancer cachexia. World J. 135–141.
Gastroenterol. 14 (23), 3633–3641. Mantovani, G., et al., 2001. Managing cancer-related anorexia/cachexia. Drugs 61 (4),
Kim, H.J., et al., 2012. Pathophysiological role of hormones and cytokines in cancer 499–514.
cachexia. J. Korean Med. Sci. 27 (2), 128–134. Mantovani, G., et al., 2010. Phase II nonrandomized study of the efficacy and safety of
Kir, S., et al., 2014. Tumour-derived PTH-related protein triggers adipose tissue browning COX-2 inhibitor celecoxib on patients with cancer cachexia. J. Mol. Med. (Berl.) 88
and cancer cachexia. Nature 513 (7516), 100–104. (1), 85–92.
Kline, W.O., et al., 2007. Rapamycin inhibits the growth and muscle-sparing effects of Marks, D.L., Ling, N., Cone, R.D., 2001. Role of the central melanocortin system in ca-
clenbuterol. J. Appl. Physiol. (1985) 102 (2), 740–747. chexia. Cancer Res. 61 (4), 1432–1438.
Kondrup, J., et al., 2003. ESPEN guidelines for nutrition screening 2002. Clin. Nutr. 22 Marshall, L.L., 2003. Megestrol acetate therapy in geriatric patients: case reviews and
(4), 415–421. associated deep vein thrombosis. Consult Pharm. 18 (9), 764–773.
Kosty, M., et al., 1994. Cisplatin, vinblastine, and hydrazine sulfate in advanced, non- Martin, L., et al., 2015. Diagnostic criteria for the classification of cancer-associated
small-cell lung cancer: a randomized placebo-controlled, double-blind phase III study weight loss. J. Clin. Oncol. 33 (1), 90–99.
of the Cancer and Leukemia Group B. J. Clin. Oncol. 12 (6), 1113–1120. Martin, L., et al., 2010. Prognostic factors in patients with advanced cancer: use of the
Kraft, M., et al., 2012. L-carnitine-supplementation in advanced pancreatic cancer patient-generated subjective global assessment in survival prediction. J. Clin. Oncol.
(CARPAN)–a randomized multicentre trial. Nutr. J. 11, 52. 28 (28), 4376–4383.
Krzystek-Korpacka, M., et al., 2007. Impact of weight loss on circulating IL-1, IL-6, IL-8, Mathieson, C.M., Stam, H.J., 1995. Reneotiating identity: cancer narratives. Sociol.
TNF-alpha, VEGF-A, VEGF-C and midkine in gastroesophageal cancer patients. Clin. Health Illn. 17 (3), 283–306.
Biochem. 40 (18), 1353–1360. Mazzotta, P., Jeney, C.M., 2009. Anorexia-cachexia syndrome: a systematic review of the
Kubrak, C., Jensen, L., 2007. Critical evaluation of nutrition screening tools re- role of dietary polyunsaturated fatty acids in the management of symptoms, survival,
commended for oncology patients. Cancer Nurs. 30 (5), E1–6. and quality of life. J. Pain Symptom Manage. 37 (6), 1069–1077.
Laenen, A., Alonso, A., 2010. The functional living index-cancer: estimating its reliability McCarthy, H.D., et al., 1994. Megestrol acetate stimulates food and water intake in the
based on clinical trial data. Qual. Life Res. 19 (1), 103–109. rat: effects on regional hypothalamic neuropeptide Y concentrations. Eur. J.
Lai, V., et al., 2008. Results of a pilot study of the effects of celecoxib on cancer cachexia Pharmacol. 265 (1-2), 99–102.
in patients with cancer of the head, neck, and gastrointestinal tract. Head Neck 30 McClement, S.E., Degner, L.F., Harlos, M., 2004. Family responses to declining intake and
(1), 67–74. weight loss in a terminally ill relative. Part 1: fighting back. J. Palliat. Care 20 (2),
Langer, C.J., Hoffman, J., Ottery, F.D., 2001. Clinical significance of weight loss in cancer 93–100.
patients: rationale for the use of anabolic agents in the treatment of cancer-related McGrath, P., 2002. Reflections on nutritional issues associated with cancer therapy.
cachexia. Nutrition 17 (Suppl 1), S1–20. Cancer Pract. 10 (2), 94–101.
Lazarus, R.S., Folkman, S., 1984. Stress, Appraisal, and Coping. Springer Publishing McMillan, D.C., 2013. The systemic inflammation-based Glasgow prognostic score: a
Company. decade of experience in patients with cancer. Cancer Treat. Rev. 39 (5), 534–540.
Lesniak, W., et al., 2008. Effects of megestrol acetate in patients with cancer anorexia- McMillan, D.C., et al., 1999. A prospective randomized study of megestrol acetate and
cachexia syndrome–a systematic review and meta-analysis. Pol. Arch. Med. Wewn ibuprofen in gastrointestinal cancer patients with weight loss. Br. J. Cancer 79 (3-4),
118 (11), 636–644. 495–500.
Lesser, G.J., et al., 2006. A phase III randomized study comparing the effects of Mehrzad, V., Afshar, R., Akbari, M., 2016. Pentoxifylline treatment in patients with
Oxandrolone (Ox) and Megestrol acetate (Meg) on weight (wt), lean body mass cancer cachexia: a double-blind, randomized, placebo-controlled clinical trial. Adv.
(LBM) and quality of life (QOL) in solid tumor patients (pts) receiving chemotherapy Biomed. Res. (5), 60.
(chemo). J. Clin. Oncol. 24 (90180), 18546. Mondello, et al., 2015. Cancer cachexia syndrome: pathogenesis, diagnosis, and new
Letilovic, T., Vrhovac, R., 2013. Influence of additional criteria from a definition of ca- therapeutic options. Nutr Cancer 67 (1), 12–26.
chexia on its prevalence–good or bad thing? Eur. J. Clin. Nutr. 67 (8), 797–801. Mondello, P., et al., 2014. Emerging markers of cachexia predict survival in cancer pa-
Letilovic, T., et al., 2013. Differences in routine laboratory parameters related to cachexia tients. BMC Cancer 14, 828.
between patients with hematological diseases and patients with solid tumors or heart Montazeri, A., et al., 2003. Quality of life in lung cancer patients: does socioeconomic
failure - is there only one cachexia? Nutr. J. 12, 6. status matter? Health Qual. Life Outcomes 1, 19.
Li, W.G., et al., 2004. Ghrelin inhibits proinflammatory responses and nuclear factor- Murphy, K.T., Lynch, G.S., 2012. Editorial update on emerging drugs for cancer cachexia.
kappaB activation in human endothelial cells. Circulation 109 (18), 2221–2226. Expert Opin. Emerg. Drugs 17 (1), 5–9.
Lin, C.Y., et al., 2015. Rasch models suggested the satisfactory psychometric properties of Murphy, R.A., et al., 2011. Nutritional intervention with fish oil provides a benefit over
the World Health Organization Quality of life-brief among lung cancer patients. J. standard of care for weight and skeletal muscle mass in patients with nonsmall cell
Health Psychol. lung cancer receiving chemotherapy. Cancer 117 (8), 1775–1782.
Lira, F.S., et al., 2015. The therapeutic potential of exercise to treat cachexia. Curr. Opin. Muscaritoli, M., et al., 2006. Prevention and treatment of cancer cachexia: new insights
Support Palliat. Care 9 (4), 317–324. into an old problem. Eur. J. Cancer 42 (1), 31–41.
Lissoni, et al., 1996. Is there a role for melatonin in the treatment of neoplastic cachexia? Muscaritoli, M., Rossi Fanelli, F., Molfino, A., 2016. Perspectives of health care profes-
Eur. J. Cancer 32a (8), 1340–1343. sionals on cancer cachexia: results from three global surveys. Ann. Oncol. 27 (12),
Loprinzi, C.L., et al., 1999. Randomized comparison of megestrol acetate versus dex- 2230–2236.
amethasone versus fluoxymesterone for the treatment of cancer anorexia/cachexia. J. Muscaritoli, M., et al., 2010. Consensus definition of sarcopenia, cachexia and pre-ca-
Clin. Oncol. 17 (10), 3299–3306. chexia: joint document elaborated by Special Interest Groups (SIG) "cachexia-anor-
Lovgren, M., et al., 2008. Symptoms and problems with functioning among women and exia in chronic wasting diseases" and "nutrition in geriatrics". Clin. Nutr. 29 (2),
men with inoperable lung cancer–a longitudinal study. Lung Cancer 60 (1), 113–124. 154–159.
Lundholm, K., et al., 2004. Palliative nutritional intervention in addition to cycloox- Naing, A., et al., 2015. Olanzapine for cachexia in patients with advanced cancer: an
ygenase and erythropoietin treatment for patients with malignant disease: effects on exploratory study of effects on weight and metabolic cytokines. Support Care Cancer
survival, metabolism, and function. Cancer 100 (9), 1967–1977. 23 (9), 2649–2654.
Lundholm, K., et al., 2007. Insulin treatment in cancer cachexia: effects on survival, Navigante, A., Morgado, C., 2016. Does physical exercise improve quality of life of ad-
metabolism, and physical functioning. Clin. Cancer Res. 13 (9), 2699–2706. vanced cancer patients? Curr. Opin. Supportive Palliat. Care 10 (4), 306–309.
Maccio, A., Madeddu, C., Mantovani, G., 2012a. Current pharmacotherapy options for Neary, N.M., et al., 2004. Ghrelin increases energy intake in cancer patients with im-
cancer anorexia and cachexia. Expert Opin. Pharmacother. 13 (17), 2453–2472. paired appetite: acute, randomized, placebo-controlled trial. J. Clin. Endocrinol.
Maccio, A., et al., 2012b. A randomized phase III clinical trial of a combined treatment for Metab. 89 (6), 2832–2836.
cachexia in patients with gynecological cancers: evaluating the impact on metabolic Neelemaat, F., et al., 2011. Comparison of five malnutrition screening tools in one hos-
and inflammatory profiles and quality of life. Gynecol. Oncol. 124 (3), 417–425. pital inpatient sample. J. Clin. Nurs. 20 (15–16), 2144–2152.
Mackillop, W.J., Quirt, C.F., 1997. Measuring the accuracy of prognostic judgments in Nicolini, A., et al., 2013. Malnutrition, anorexia and cachexia in cancer patients: a mini-
oncology. J. Clin. Epidemiol. 50 (1), 21–29. review on pathogenesis and treatment. Biomed. Pharmacother. 67 (8), 807–817.
Maddocks, M., Jones, L.W., Wilcock, A., 2013. Immunological and hormonal effects of Oberholzer, R., et al., 2013. Psychosocial effects of cancer cachexia: a systematic litera-
exercise: implications for cancer cachexia. Curr. Opin. Support Palliat. Care 7 (4), ture search and qualitative analysis. J. Pain Symptom Manage. 46 (1), 77–95.
376–382. Oldervoll, L.M., et al., 2011. Physical exercise for cancer patients with advanced disease:
Maddocks, M., Murton, A.J., Wilcock, A., 2011. Improving muscle mass and function in a randomized controlled trial. Oncologist 16 (11), 1649–1657.
cachexia: non-drug approaches. Curr. Opin. Support Palliat. Care 5 (4), 361–364. Oliveira, A.G., Gomes-Marcondes, M.C., 2016. Metformin treatment modulates the tu-
Madeddu, C., Maccio, A., Mantovani, G., 2012. Multitargeted treatment of cancer ca- mour-induced wasting effects in muscle protein metabolism minimising the cachexia
chexia. Crit. Rev. Oncog. 17 (3), 305–314. in tumour-bearing rats. BMC Cancer 16, 418.
Madeddu, C., et al., 2009. Medroxyprogesterone acetate in the management of cancer Ottery, F.D., 1996. Definition of standardized nutritional assessment and interventional
cachexia. Expert Opin. Pharmacother. 10 (8), 1359–1366. pathways in oncology. Nutrition 12 (Suppl. 1), S15–9.

102
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Paredes, T., Simoes, M.R., Canavarro, M.C., 2010. Psychometric properties of the World Song, B., et al., 2009. Association of interleukin-8 with cachexia from patients with low-
Health Organization Quality of Life Questionnaire (WHOQOL-100) in Portuguese third gastric cancer. Comp. Funct. Genomics 212345.
patients with sarcoma. Psychol. Health Med. 15 (4), 420–433. Spiro, A., et al., 2006. The views and practice of oncologists towards nutritional support
Park, S.M., et al., 2006. EuroQol and survival prediction in terminal cancer patients: a in patients receiving chemotherapy. Br. J. Cancer 95 (4), 431–434.
multicenter prospective study in hospice-palliative care units. Support Care Cancer Sprangers, M.A., et al., 1998. The European Organization for Research and Treatment of
14 (4), 329–333. Cancer approach to developing questionnaire modules: an update and overview.
Pascual Lopez, A., et al., 2004. Systematic review of megestrol acetate in the treatment of EORTC Qual. Life Study Group Qual. Life Res. 7 (4), 291–300.
anorexia-cachexia syndrome. J. Pain Symptom Manage. 27 (4), 360–369. Stene, G.B., et al., 2013. Effect of physical exercise on muscle mass and strength in cancer
Penafuerte, C.A., et al., 2016. Identification of neutrophil-derived proteases and angio- patients during treatment–a systematic review. Crit. Rev. Oncol. Hematol. 88 (3),
tensin II as biomarkers of cancer cachexia. Br. J. Cancer 114 (6), 680–687. 573–593.
Penet, M.F., Bhujwalla, Z.M., 2015. Cancer cachexia, recent advances, and future direc- Stewart Coats, A.J., et al., 2016. Espindolol for the treatment and prevention of cachexia
tions. Cancer J. 21 (2), 117–122. in patients with stage III/IV non-small cell lung cancer or colorectal cancer: a ran-
Perez De Oteyza, C., et al., 1998. [Megestrol in the treatment of AIDS associated cachexia. domized, double-blind, placebo-controlled, international multicentre phase II study
Evaluation by bioelectric impedance analysis of body composition]. An. Med. Interna (the ACT-ONE trial). J. Cachexia Sarcopenia Muscle 7 (3), 355–365.
15 (5), 255–258. Storer, T.W., et al., 2005. A randomized, placebo-controlled trial of nandrolone decanoate
Petersen, A.M., Pedersen, B.K., 2005. The anti-inflammatory effect of exercise. J. Appl. in human immunodeficiency virus-infected men with mild to moderate weight loss
Physiol. (1985) 98 (4), 1154–1162. with recombinant human growth hormone as active reference treatment. J. Clin.
Petruzzelli, M., et al., 2014. A switch from white to brown fat increases energy ex- Endocrinol. Metab. 90 (8), 4474–4482.
penditure in cancer-associated cachexia. Cell Metab. 20 (3), 433–447. Strasser, F., 2008. Diagnostic criteria of cachexia and their assessment: decreased muscle
Pinto Jr., J.A., et al., 2004. Fish oil supplementation in F1 generation associated with strength and fatigue. Curr. Opin. Clin. Nutr. Metab. Care 11 (4), 417–421.
naproxen, clenbuterol, and insulin administration reduce tumor growth and cachexia Strimbu, K., Tavel, J.A., 2010. What are biomarkers? Curr. Opin. HIV AIDS 5 (6),
in Walker 256 tumor-bearing rats. J. Nutr. Biochem. 15 (6), 358–365. 463–466.
Prado, C.M., et al., 2009. Sarcopenia as a determinant of chemotherapy toxicity and time Stukan, M., et al., 2017. Independent psychometric validation of European Organization
to tumor progression in metastatic breast cancer patients receiving capecitabine for Research and Treatment of Cancer Quality of Life Questionnaire-Endometrial
treatment. Clin. Cancer Res. 15 (8), 2920–2926. Cancer Module (EORTC QLQ-EN24). Eur. J. Cancer Care (Engl.).
Prado, C.M., et al., 2008. Prevalence and clinical implications of sarcopenic obesity in Sun, L., Quan, X.Q., Yu, S., 2015. An epidemiological survey of cachexia in advanced
patients with solid tumours of the respiratory and gastrointestinal tracts: a popula- cancer patients and analysis on its diagnostic and treatment Status. Nutr Cancer 67
tion-based study. Lancet Oncol. 9 (7), 629–635. (7), 1056–1062.
Reid, J., 2014. Psychosocial, educational and communicative interventions for patients Suno, M., et al., 2015. Refractory cachexia is associated with increased plasma con-
with cachexia and their family carers. Curr. Opin. Support Palliat. Care 8 (4), centrations of fentanyl in cancer patients. Ther. Clin. Risk Manage. 11, 751–757.
334–338. Tafelski, S., Hauser, W., Schafer, M., 2016. Efficacy, tolerability, and safety of cannabi-
Reid, J., et al., 2009. The experience of cancer cachexia: a qualitative study of advanced noids for chemotherapy-induced nausea and vomiting–a systematic review of sys-
cancer patients and their family members. Int. J. Nurs. Stud. 46 (5), 606–616. tematic reviews. Schmerz 30 (1), 14–24.
Reid, J., et al., 2013. Non-steroidal anti-inflammatory drugs for the treatment of cancer Takaya, K., et al., 2000. Ghrelin strongly stimulates growth hormone release in humans.
cachexia: a systematic review. Palliat. Med. 27 (4), 295–303. J. Clin. Endocrinol. Metab. 85 (12), 4908–4911.
Reid, J., et al., 2014. Evaluation of a psychoeducational intervention for patients with Tan, C.R., et al., 2014. Pancreatic cancer cachexia: a review of mechanisms and ther-
advanced cancer who have cachexia and their lay carers (EPACaCC): study protocol. apeutics. Front. Physiol. (5), 88.
J. Adv. Nurs. 70 (5), 1174–1183. Temel, J.S., et al., 2016. Anamorelin in patients with non-small-cell lung cancer and
Reuter, S.E., Martin, J.H., 2016. Pharmacokinetics of cannabis in cancer cachexia-anor- cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind,
exia syndrome. Clin. Pharmacokinet. 55 (7), 807–812. phase 3 trials. Lancet Oncol. 17 (4), 519–531.
Ribaudo, J.M., et al., 2000. Re-validation and shortening of the Functional Assessment of The World Health Organization Quality of Life Assessment, 1998. (WHOQOL): develop-
Anorexia/Cachexia Therapy (FAACT) questionnaire. Qual. Life Res. 9 (10), ment and general psychometric properties. Soc. Sci. Med. 46 (12), 1569–1585.
1137–1146. Thoresen, L., et al., 2013. Nutritional status, cachexia and survival in patients with ad-
Ries, A., et al., 2012. A systematic review on the role of fish oil for the treatment of vanced colorectal carcinoma. Different assessment criteria for nutritional status
cachexia in advanced cancer: an EPCRC cachexia guidelines project. Palliat. Med. 26 provide unequal results. Clin. Nutr. 32 (1), 65–72.
(4), 294–304. Tisdale, M.J., 2005. Molecular pathways leading to cancer cachexia. Physiology
Rigas, J.R., et al., 2010. Efect of ALD518, a humanized anti-IL-6 antibody, on lean body (Bethesda) 20, 340–348.
mass loss and symptoms in patients with advanced non-small cell lung cancer Todorov, T., et al., 1998. Purification and characterization of a tumor lipid-mobilizing
(NSCLC): results of a phase II randomized, double-blind safety and efficacy trial. J. factor. Cancer Res. 58 (11), 2353–2358.
Clin. Oncol. 28 (Suppl. 15), 7622. Toledo, M., et al., 2016. A multifactorial anti-cachectic approach for cancer cachexia in a
Ryan, A.M., et al., 2009. Enteral nutrition enriched with eicosapentaenoic acid (EPA) rat model undergoing chemotherapy. J. Cachexia Sarcopenia Muscle 7 (1), 48–59.
preserves lean body mass following esophageal cancer surgery: results of a double- Toledo, M., et al., 2014. Formoterol in the treatment of experimental cancer cachexia:
blinded randomized controlled trial. Ann. Surg. 249 (3), 355–363. effects on heart function. J. Cachexia Sarcopenia Muscle 5 (4), 315–320.
Salsman, J.M., et al., 2015. Brief versions of the FACIT-fatigue and FAACT subscales for Tuca, A., Jimenez-Fonseca, Gascon, 2013. Clinical evaluation and optimal management of
patients with non-small cell lung cancer cachexia. Support Care Cancer 23 (5), cancer cachexia. Crit. Rev. Oncol. Hematol. 88 (3), 625–636.
1355–1364. Uehara, A., et al., 1989. Anorexia induced by interleukin 1: involvement of corticotropin-
Sanchez-Lara, K., et al., 2014. Effects of an oral nutritional supplement containing eico- releasing factor. Am. J. Physiol. 257 (Pt (3)), R613–7.
sapentaenoic acid on nutritional and clinical outcomes in patients with advanced van der Meij, B.S., et al., 2013. Pre-cachexia and cachexia at diagnosis of stage III non-
non-small cell lung cancer: randomised trial. Clin. Nutr. 33 (6), 1017–1023. small-cell lung carcinoma: an exploratory study comparing two consensus-based
Schag, C.A., Heinrich, R.L., 1990. Development of a comprehensive quality of life mea- frameworks. Br. J. Nutr. 109 (12), 2231–2239.
surement tool: CARES. Oncology (Williston Park) 4 (5), 135–138 discussion 147. van der Meij, B.S., et al., 2010. Oral nutritional supplements containing (n-3) poly-
Schag, C.A., Ganz, A., Heinrich, R.L., 1991. CAncer rehabilitation evaluation system– unsaturated fatty acids affect the nutritional status of patients with stage III non-small
short form (CARES-SF). A cancer specific rehabilitation and quality of life instrument. cell lung cancer during multimodality treatment. J. Nutr. 140 (10), 1774–1780.
Cancer 68 (6), 1406–1413. van Dijk, D.J., et al., 2015. Effects of oral meal feeding on whole body protein breakdown
Scheede-Bergdahl, C., et al., 2012. Is IL-6 the best pro-inflammatory biomarker of clinical and protein synthesis in cachectic pancreatic cancer patients. J. Cachexia, Sarcopenia
outcomes of cancer cachexia? Clin. Nutr. 31 (1), 85–88. Muscle 6 (3), 212–221.
Schwartz, A.L., Winters-Stone, K., 2009. Effects of a 12-month randomized controlled trial Vanhoutte, G., et al., 2016. Cachexia in cancer: what is in the definition? BMJ
of aerobic or resistance exercise during and following cancer treatment in women. Gastroenterol. 3 (1), e000097.
Phys. Sportsmed. 37 (3), 62–67. Velasco, C., et al., 2011. Comparison of four nutritional screening tools to detect nutri-
Senesse, P., et al., 2008. Nutritional support during oncologic treatment of patients with tional risk in hospitalized patients: a multicentre study. Eur. J. Clin. Nutr. 65 (2),
gastrointestinal cancer: who could benefit? Cancer Treat. Rev. 34 (6), 568–575. 269–274.
Sesterhenn, A.M., et al., 2012. [Significance of autopsy in patients with head and neck Wallengren, O., Lundholm, K., Bosaeus, I., 2013. Diagnostic criteria of cancer cachexia:
cancer]. Laryngorhinootologie 91 (6), 375–380. relation to quality of life, exercise capacity and survival in unselected palliative care
Shimizu, Y., et al., 2003. Increased plasma ghrelin level in lung cancer cachexia. Clin. patients. Support Care Cancer 21 (6), 1569–1577.
Cancer Res. 9 (2), 774–778. Warren, S., 1932. The immediate causes of death in cancer. Am. J. Med. Sci. 184 (5),
Shum, A.M., Polly, 2012. Cancer cachexia: molecular targets and pathways for diagnosis 610–615.
and drug intervention. Endocr. Metab. Immune Disord. Drug Targets 12 (3), Waseem, T., et al., 2008. Exogenous ghrelin modulates release of pro-inflammatory and
247–259. anti-inflammatory cytokines in LPS-stimulated macrophages through distinct sig-
Skipworth, R.J., et al., 2010. Mass spectrometric detection of candidate protein bio- naling pathways. Surgery 143 (3), 334–342.
markers of cancer cachexia in human urine. Int. J. Oncol. 36 (4), 973–982. Weed, H.G., et al., 2011. Lean body mass gain in patients with head and neck squamous
So, H.S., et al., 2004. Validation of quality of life index-cancer among korean patients cell cancer treated perioperatively with a protein- and energy-dense nutritional
with cancer. Taehan Kanho Hakhoe Chi 34 (5), 693–701. supplement containing eicosapentaenoic acid. Head Neck 33 (7), 1027–1033.
Solheim, T.S., Laird, B.J., 2012. Evidence base for multimodal therapy in cachexia. Curr. Wen, H.S., et al., 2012. Clinical studies on the treatment of cancer cachexia with me-
Opin. Support Palliat Care 6 (4), 424–431. gestrol acetate plus thalidomide. Chemotherapy 58 (6), 461–467.
Solheim, T.S., et al., 2013. Non-steroidal anti-inflammatory treatment in cancer cachexia: Wesseltoft-Rao, N., et al., 2015. Comparing two classifications of cancer cachexia and
a systematic literature review. Acta Oncol. 52 (1), 6–17. their association with survival in patients with unresected pancreatic cancer. Nutr

103
M. Sadeghi et al. Critical Reviews in Oncology / Hematology 127 (2018) 91–104

Cancer 67 (3), 472–480. cancer. Eur. J. Cancer Care (Engl.) 20 (5), 593–600.
Wheelwright, S.J., Johnson, C.D., 2015. Patient-reported outcomes in cancer cachexia Willox, J.C., et al., 1984. Prednisolone as an appetite stimulant in patients with cancer.
clinical trials. Curr. Opin. Support Palliat. Care 9 (4), 325–332. Br. Med. J. (Clin. Res. Ed.) 288 (6410), 27.
Wheelwright, S., et al., 2013. A systematic review of health-related quality of life in- Wilson, J.R.M., et al., 2005. The assessment of patient life-expectancy: how accurate are
struments in patients with cancer cachexia. Support Care Cancer 21 (9), 2625–2636. urologists and oncologists? BJU Int. 95 (6), 794–798.
Wheelwright, S.J., et al., 2017. Development of the EORTC QLQ-CAX24, a questionnaire Wolf, I., et al., 2006. Adiponectin, ghrelin, and leptin in cancer cachexia in breast and
for cancer patients with cachexia. J. Pain Symptom Manage. 53 (2), 232–242. colon cancer patients. Cancer 106 (4), 966–973.
White, N., et al., 2016. A systematic review of predictions of survival in palliative care: Wyke, S.M., Tisdale, M.J., 2005. NF-kappaB mediates proteolysis-inducing factor induced
how accurate are clinicians and who are the experts? PLoS ONE 11 (8), e0161407. protein degradation and expression of the ubiquitin-proteasome system in skeletal
Wigmore, S.J., et al., 1997. Contribution of anorexia and hypermetabolism to weight loss muscle. Br. J. Cancer 92 (4), 711–721.
in anicteric patients with pancreatic cancer. Br. J. Surg. 84 (2), 196–197. Yennurajalingam, S., et al., 2012. The role of thalidomide and placebo for the treatment
Wigmore, S.J., et al., 2000. Characteristics of patients with pancreatic cancer expressing a of cancer-related anorexia-cachexia symptoms: results of a double-blind placebo-
novel cancer cachectic factor. Br. J. Surg. 87 (1), 53–58. controlled randomized study. J. Palliat. Med. 15 (10), 1059–1064.
Wilkes, E.A., et al., 2011. Poor tolerability of thalidomide in end-stage oesophageal Yki-Jarvinen, H., 2004. Thiazolidinediones. New Engl. J. Med. 351 (11), 1106–1118.

104

You might also like