SlideShare a Scribd company logo
1 of 35
1
Efficacy of Omega-3 Polyunsaturated Fatty
Acids for Preventing Cancer-Induced
Cachexia
Josh Nooner, BS, CSCS
NSCI – 5843
2/29/16
2
Introduction
Cachexia is an extremely debilitating condition that is seen in many chronic and terminal
diseases such as cancer. This syndrome is characterized by unintentional weight loss, anorexia,
and severe muscle wasting. Stewart reports, “20% of all cancer deaths are caused directly by
cachexia (Stewart, 2006).” As the cause of death for 1 in 5 cancer patients, an individual who
develops this wasting syndrome has a very poor prognosis. In fact, “Up to 50% of cancer patients
suffer from a progressive atrophy of adipose tissue and skeletal muscle, called cachexia,
resulting in weight loss, a reduced quality of life, and a shortened survival time (Tisdale, 2009).”
As we can see, cachexia is a very prevalent condition, affecting half of all individuals diagnosed
with cancer and is the direct cause of death for 20% of cancer deaths. Additionally, this
syndrome results in a lower quality of life and a reduced survival time, making it an extremely
important target for therapy. Cachexia is currently being heavily studied, as we aim to find an
effective intervention strategy that will reduce the occurrence of this devastating syndrome,
increase the quality of patient’s life’s, and lengthen their survival time. Omega-3 polyunsaturated
fatty acids (PUFAs) are one area of research that is being investigated for their use in alleviating
this wasting syndrome, specifically through their anti-inflammatory properties and their ability to
mediate specific metabolic processes that are seen in cachexia. After studying the altered
metabolic pathways that present in cachexia, we can better understand how the omega-3 fatty
acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), work to alleviate cancer-
induced cachexia.
In order to better understand this syndrome, we must first define exactly what it is. In 2011
Fearon brought together an international panel of experts that formed a consensus on the
definition, diagnosis, and the classification of cachexia. They reported that, “Cancer cachexia is
3
defined as a multifactorial syndrome characterized by an ongoing loss of skeletal muscle mass
(with or without loss of fat mass) that cannot be fully reversed by conventional nutritional
support and leads to progressive functional impairment (Fearon, 2011).” This definition brings
up two very important characteristics of cachexia that are crucial to understand. First, cachexia
cannot be fully reversed by conventional nutritional support. Unlike with starvation induced
weight loss, simply adding calories into the diet does not reverse cachexia. Secondly, cachexia
leads to progressive functional impairment. The longer that someone is in a cachectic state, the
more functionally impaired they become, eventually losing all strength which is often the cause
of death. Knowing this helps us to understand why practitioners need to actively manage this
condition and why it is such an important area of research.
We know that cachexia occurs along a spectrum with three stages of severity, and that
there are specific clinical aspects of each stage. The three stages are precachexia, cachexia, and
refractory cachexia. The main clinical characteristics of cachexia are weight loss, anorexia or
loss of appetite, reduced food intake, complex metabolic changes, systemic inflammation, and
reduced functional capacity. There is a stream of negative side effects, each one causing another,
with each further step downward compounding the entire cycle. This is an extremely viscous
catabolic cycle that is repeated over and over eventually leading to death in the majority of
cancer patients.
The metabolic processes seen in cachexia are quite complex, as the cellular mechanisms are
not typically seen in normal healthy cells, or in cells of those experiencing starvation induced
weight loss. Barber reports, “The fundamental difference between the weight loss observed in
cachexia and that seen in, e.g., starvation, is the lack of reversibility with feeding (Barber,
2001).” Simply adding calories to the diet does not reverse weight loss seen in cachexia. These
4
metabolic alterations are thought to be induced by a combination of both tumor and host
changes. Barber continues and states, “Candidate mediators include cytokines, neuroendocrine
hormones, and tumor specific products. Several pro-inflammatory cytokines, including tumor
necrosis factor (TNF), interleukin-1 (IL-1), interleukin-6 (IL-6), and interferon-y, have been
implicated in cachexia (Barber, 2001).” Chronic inflammation is one of the major clinical
manifestations of cancer, resulting in an elevation of pro-inflammatory cytokines, which mediate
cachexia.
Hypermetabolism is another complication of cachexia that leads to muscle and fat loss.
“Approximately 50% of patients are hypermetabolic (REE > 110% of predicted) …
hypermetabolism has been related to an elevated adrenergic state or systemic inflammation
(Fearon, 2012).” Cachectic patients are in a hypermetabolic state, burning more calories than
they would under normal conditions, yet have very limited appetites, thus they are also
consuming fewer calories than they would under normal conditions. The hypermetabolic state is
thought to be caused, in part, by futile cycles. Futile cycles are when opposing metabolic cycles
(glycolysis and gluconeogenesis) occur simultaneously, resulting in the use of ATP and
production of heat.
Tumor cells are known to have an increased uptake of glucose, even in the presence of
adequate oxygen levels. This phenomenon, known as the Warburg effect, causes excess
production of lactate and upregulates the Cori cycle. Fearon continues, “Increased Cori cycle
activity has been documented in weight-losing cancer patients… Overall glucose flux has been
shown to be increased in weight-losing cancer patients, and such flux has been estimated to
contribute up to 40% of the increase in energy expenditure in metastatic cancer (Fearon, 2012).”
Here Fearon describes one of the main causes of hypermetabolism seen in cachexia. The second
5
known cause of increased metabolism is a heightened expression of mitochondrial uncoupling
proteins (UCPs). This causes, as Fearon explains, “Dissipation of the proton gradient so that
oxygen consumption is no longer coupled to ATP synthesis and heat is generated (Fearon,
2012).” This effect is commonly seen in the brown adipose tissue (BAT) of cachectic cancer
patients, causing a thermogenic effect. The combination of an increased glucose flux and an
increased expression of UCPs in adipose tissue creates a rise in heat production. These are the
two main causes of hypermetabolism seen in cancer-induced cachexia.
Lastly, cachexia is characterized by alterations in gene expression and protein levels. As
previously stated, there is an overexpression of mitochondrial uncoupling proteins. This effect
increases heat production, ATP loss, and creates a hypermetabolic effect. Another gene that is
altered is the GLUT-4 receptor protein gene. GLUT-4 expression is downregulated in cachexia
and causes insulin insensitivity. Reductions in insulin sensitivity create a pro-catabolic state,
leading to increased use of the Cori cycle and loss of muscle mass.
Additionally, both muscle and adipose tissue loss are due to expression of specific proteins
seen in cachexia. Tisdale describes, “Loss of adipose tissue is due to increased lipolysis by tumor
or host products. Loss of skeletal muscle in cachexia results from a depression in protein
synthesis combined with an increase in protein degradation. The increase in protein degradation
may include increased activity of the ubiquitin-proteasome pathway (Tisdale, 2009).” Tisdale
explains that loss of adipose tissue is due to lipolysis brought about by certain factors. One such
proposed factor is zinc alpha2-glycoprotein (ZAG) which was discovered to be a lipid
mobilizing factor. This protein has been shown to cause adipose tissue loss in many animal and
human cell studies. Tisdale concludes, “An increased ZAG expression may be responsible for
the increased lipolytic response of adipose tissue in cancer cachexia (Tisdale, 2009).” Mimicking
6
this fat wasting protein is a muscle wasting glycoprotein called Proteolysis-Inducing Factor
(PIF). PIF has been found to induce protein degradation and prevent protein synthesis in
numerous animal, cell, and human models of cancer cachexia. Furthermore, the ubiquitin-
proteasome pathway is highly active in cachexia, causing the majority of muscle loss. Jatoi
explains that, “The ubiquitin-proteasome pathway is responsible for >80% of lean tissue wasting
from cancer (Jatoi, 2005).” A direct correlation exists between the degree of expression of the
ubiquitin-proteasome pathway and how advanced a tumor is. In summary, although not an
extensive list, there are many proteins that are known to be over expressed in cancer-induced
cachexia.
Now that we have a better understanding of the mediators and mechanisms involved in
cachexia we can look at how omega-3 PUFAs act on these pathways to reduce cachexia. There
have been a number of quality papers published detailing the beneficial effects of omega-3
PUFAs on cachexia. In 2005 Jatoi reported that, “The purported mechanisms of n-3 PUFA in
preventing lean tissue wasting are through the suppression of the ubiquitin-proteasome pathway,
inflammatory cytokines, and the cancer cachectic factor (Jatoi, 2005).” As previously discussed,
the ubiquitin-proteasome pathway accounts for a vast majority of muscle loss and the
inflammatory cytokines lead to hypermetabolism.
In 2008 Rondanelli reported, “Production of pro-inflammatory cytokines such as IL-6, IL-1,
and TNF can be downregulated by EPA. Furthermore, the effects of PIF, are also inhibited by
EPA (Rondanelli, 2008).” Rondanelli discusses EPA’s effect on cytokines, which are chronically
elevated in cachexia. Adding to this, Fetterman elaborates, “Supplementation with n-3 PUFA
also appears able to reduce production of proinflammatory cytokines, such as IL-1, IL-6, IL-8,
and TNF-a…Excess activity of these substances contributes to pathological inflammation
7
(Fetterman, 2009).” Reduction in pro-inflammatory cytokines is one of the main reported
mechanisms and a major benefit of n-3 supplementation for cachexia patients.
Furthermore, a recent report summarized the antineoplastic activity of n-3 PUFAs. “The
three main antineoplastic activities of n-3 PUFAs that have been proposed are (i) modulation of
COX activity; (ii) alteration of membrane dynamics and cell surface receptor function and (iii)
increased cellular oxidative stress (Cockbain, 2011).” This evidence suggests n-3 PUFAs can
beneficially effect eicosanoid synthesis through COX enzyme modification, can be incorporated
into the lipid bilayer and alter the structure and function of the cell surface and its downstream
signaling pathways, and can also beneficially alter the intracellular redox status of target tissues.
Adding to these beneficial effects, Hardman states, “n-3 fatty acids may be detrimental to
the growth of metastatic or residual cancer cells by altering eicosanoid metabolism, slowing
cancer cell mitosis, increasing cancer cell death, inducing differentiation, suppressing
angiogenesis, and altering estrogen metabolism (Hardman, 2002).” Hardman shows us two more
benefits of n-3 PUFAs that have yet to be discussed, angiogenesis and estrogen. By preventing
vascular supply to the tumor, fish oil deprives the tumor of nutrients needed for growth. By
decreasing estrogen production, n-3 PUFAs prevent the activation of estrogen receptors on
breast, colon, and prostate cancer cells, which all have estrogen receptors needed for growth.
More recently, Murphy reported many more benefits of n-3 PUFAs, stating, “EPA may
support the anabolic potential of muscle through sensitizing skeletal muscle to insulin. EPA has
been shown to improve glucose uptake and increase GLUT-4 expression in skeletal muscle
(Murphy, 2011).” This is a highly desirable effect, noting that muscle protein synthesis must
occur in an anabolic state created by the hormone insulin. Murphy continues, “EPA and DHA
reduced the side effects from chemotherapy, and limited weight loss and anorexia. EPA and
8
DHA have also been reported to enhance tumor response to chemotherapy (Murphy, 2011).”
These reports that EPA and DHA can increase the positive effects of chemotherapy while also
reducing the side effects just add to the list of reasons someone would consider taking fish oil to
treat cancer-induced cachexia.
Now that we have reviewed the main pathways through which omega-3 fatty acids inhibit
cancer-induced cachexia, we can take a deeper look into the literature supporting both sides of
the argument. In order to fully understand this complex issue a full literature review is necessary.
The following section will be a review of the major studies that support the use of fish oil for
cancer patients with cachexia. I will then review the major studies that discourage using fish oil
to treat cachexia. Lastly, I will summarize the major findings of these studies, discuss the
strengths and weaknesses of each study, and provide a recommendation for their use.
Review of Supporting Articles
The first point of view on this issue that will be discussed is that Omega-3’s are effective
for the treatment of cancer induced cachexia. Beginning with the earliest studies on this issue and
progressing chronologically, the main studies on this subject will be discussed in detail. Included
in the review will be the purpose of each study, the methodology, the results of the study, and the
researcher’s conclusions. The opposing point of view on this issue will be discussed
subsequently, with each study supporting this view point presented chronologically and
discussed in a similar fashion.
Omega-3 PUFAs began to be investigated for their potential ameliorating effects on
cachexia as early as 1975. The study of fish oil began when epidemiological studies, often
studying Greenland Inuits or Native Alaskans, first showed that cultures eating higher amounts
9
of fish had lower incidence of chronic diseases such as heart disease, diabetes, and cancer. One
study reported, “Alaskan and Greenland Eskimos have lower rates of colon cancer and a higher
consumption of n-3 fatty acids than other North Americans (Chang, 1998).” This study, among
others, showed that there was a beneficial effect of consuming higher amounts of n-3 PUFAs,
mainly from fish, and kick started the investigation into fish oil. Since then, thousands of studies
have been done, investigating fish oil’s effects on hundreds of clinical outcomes, including
cancer-induced cachexia.
Many studies have been conducted to determine the appropriate dosage of fish oil and to
test for adverse side effects and toxicity. In 1999 a phase 1 clinical trial was conducted to
determine the maximal tolerated dose of fish oil. Patients were given .05g/kg/day fish oil
supplements that were to be increased by .05g/kg/day every 2 weeks unless a complication arose.
The authors found that, “At doses of 0.35 g/kg/day and above, at least one-third of patients
suffered a dose-limiting toxicity. Therfore, the maximum tolerable dose was considered to be 0.3
g/kg/day (Burns, 1999).” Important to note is that the “toxicity” discussed in this study were all
very minimal side effects. Burns explains, “These included excessive belching, fish taste in
mouth, fish taste of food, fish smell on their own body, flatulence, and diarrhea… In summary
we found that patients with advanced cancer can tolerate a large dose of fish oil with only minor
side effects (Burns, 1999).” This study used much larger doses of fish oil than most other studies
and found only minor side effects.
One of the earliest studies that will be discussed was published in 1990 in the Journal of
Cancer Research. The purpose of this study by Tisdale and Dhesi was to determine if omega-3
fatty acids could inhibit weight loss in an experimental cachexia model (Tisdale, 1990). In order
to test this, they transplanted MAC16 tumor fragments into the flanks of NMRI mice to induce
10
experimental colon adenocarcinoma. After tumor implantation was confirmed, the mice were
randomized into one of two dietary treatments. “The standard diet was comprised of mouse
breeding nuts which contained 50% carbohydrate, 20% protein, 4.1% crude oil and supplied
11.5% of the energy as fat (Tisdale, 1990).” All other treatment arms were isocaloric but
decreased the carbohydrate amount and provided an increasing amount of calories from fish oil,
from 5% up to 50% of total energy being derived from fish oil. The mice were weighed daily and
remained on the diet for 24 days.
The authors found that, “Weight loss was prevented in proportion to the concentration of
fish oil in the diet with almost complete protection occurring when the fish oil comprised 50% of
the diet (Tisdale, 1990).” Here we can see that the fish oil supplemented groups did show a
reversal of weight loss. The authors also reported that the weight loss inhibition was seen in a
dose dependent manner, with the groups receiving the largest amounts of calories from fish oil
losing the least amount of weight. Secondly, the authors reported, “Both the tumor weight gain
per day and the tumor volume increase were reduced in animals fed the fish oil diets (Tisdale,
1990).” Interestingly, the fish oil not only prevented weight loss, but it also prevented the tumor
from growing as quickly as it did in the control group. This is important to note, because many
studies on fish oil report there being a beneficial effect on both the host and the tumor cells. The
authors conclude by stating, “Diets enriched with fish oil have been shown to reduce both the
growth rate and the extent of weight loss produced by the MAC16 adenocarcinoma…Thus an
EPA-containing oil has been shown to exert both antitumor and anticachectic activity with no
toxicity.” This study was an important early study that shed a hopeful light on the cachexia
dilemma and opened the gateway for more research into the effects of fish oil.
11
Five years later, Tisdale was involved in another study with six other researchers that
investigated how polyunsaturated fatty acids affected cachexia progression in a group of patients
with pancreatic cancer (Wigmore, 1996). This study looked at pancreatic cancer patients because
cachexia is highly prevalent in this type of cancer. The authors were specifically looking at the
effect of fish oil supplementation on weight loss and the acute-phase response in these patients. It
has been noted that acute-phase protein elevation, “Is associated with both increased resting
energy expenditure and shortened duration of survival. This elevation is thought to be the result
of proinflammatory cytokine release. (Wigmore, 1996).” The authors measured C-reactive
protein (CRP) levels at baseline and 3 months after intervention to examine the acute-phase
response.
A total of eighteen patients with pancreatic adenocarcinoma were included in this study.
The patients were given fish oil capsules of 1g that contained 18% EPA and 12% DHA. Each
week the dose was increased by 2g/day, starting with 2g and rising to 16g/day. Weight, CRP
levels, and a nutritional assessment were taken at baseline, 1 month after treatment, and 3 months
after treatment initiation. Results of this study showed that, “All patients were losing weight
before supplementation, whereas on receiving fish oil there was a median weight gain of
0.3kg/month (Wigmore, 1996).” There was a significant decrease in weight loss in the majority
of the patients after 3 months of supplementation with fish oil. In addition, “After 1 month of
oral fish oil, patients showed an attenuation of the acute phase response by a reduced C-reactive
protein concentration (Wigmore, 1996).” The change in weight was mirrored by changes in CRP
levels. The authors conclude by stating, “Before supplementation, all of the study group
experienced progressive weigh loss; however, following administration of fish oil three quarters
of the group either become weight stable or actually gained a small amount of weight (Wigmore,
12
1996).” This study showed that polyunsaturated fatty acids prevented weight loss in pancreatic
cancer patients with cachexia.
The next study in the review was a prospective randomized control trial that was
published in 1998 by Gogos et al. The aim of this study was to investigate the omega-3 PUFAs
and vitamin E effects on survival rates and the immune system in both malnourished and well-
nourished patients with malignancy (Gogos, 1998). The 60 patients were randomized into two
groups. The first group was supplemented with fish oil, and the second group received a placebo.
There were 15 malnourished and 15 well-nourished patients in each group The groups were
followed until the time of death. Before treatment and on day 40 measurements of T cells, T
helper cells, T-suppressor cells, natural killer cells, and synthesis of interleukin-1 (IL-1),
interleukin-6(IL-6), and tumor necrosis factor (TNF) were taken in blood mononuclear cells
(Gogos, 1998).
The results of this study showed that omega-3 polyunsaturated fatty acids produced an
immunomodulating effect and increased the ratio of T-helper cells to T-suppressor cells in the
malnourished group. However, there were no significant changes in cytokine production between
the groups. In terms of survival, the well-nourished group supplemented with fish oil had the
greatest survival whereas the malnourished group receiving the placebo had the least survival.
Additionally, both the well-nourished and the malnourished groups receiving fish oil showed a
significant increase in survival compared to the placebo groups. The authors conclude by saying
that omega-3 PUFAs could offer support, especially to malnourished patients, due to their unique
anticachectic, antitumor, and immunomodulating effects (Gogos, 1998).
The next study supporting the use of omega-3 PUFAs to treat cancer cachexia was
published in 1999 in The Journal of Nutrition. This study by Barber investigated the use of a
13
supplement enriched with fish oil on acute-phase protein concentrations in patients presenting
with pancreatic cancer (Barber, 1999). For three weeks, 18 patients were given a nutritional
supplement (2g EPA/day and 1g DA/day), and an additional 18 patients who did not receive the
supplement were used as a control. The APP concentrations were measured at baseline and again
after three weeks of intervention.
The APP measurements taken at the three-week mark revealed that only transferrin was
increased in the fish oil supplemented group. However, in the control group, there were
significant decreases in albumin, pre-albumin, and transferrin, as well as a significant increase in
CRP levels, suggesting an attenuation of the APP response by fish oil. In summary, the authors
conclude, “Fish oil, in combination with a nutritional supplement, is able to prevent progression
of the APP response and cachexia in weight losing patients with advanced cancer (Barber,
1999).”
The same year as the previous study, Barber published another study looking at the
effects of a fish oil enriched nutritional supplement in those with advanced pancreatic cancer.
The purpose of this study was to determine if weight gain can be produced in pancreatic cancer
patients through the use of a conventional nutrition supplement in combination with EPA
(Barber, 1999). Barber administered nutritional supplements enriched with fish oil to 20 patients
with pancreatic adenocarcinoma. There were asked to consume two cans a day, each can
containing 1.09g EPA, 16.1g of protein, and 310 calories, in addition to their normal daily food
intake. Assessments were taken at 3 and 7 weeks for weight, body composition, dietary intake,
resting energy expenditure, and performance status (Barber, 1999).
The results showed improvements in every area that was being studied. The authors
reported, “Patients had significant weight gain at both 3 and 7 weeks (median 2 kg), dietary
14
intake increased significantly by almost 400 kcal/day, resting energy expenditure fell
significantly, and performance status and appetite were significantly improved (Barber, 1999).”
This study was able to show that fish oil not only stopped weight loss, but also lead to
improvements in appetite, increased functional performance, as well as a reduction in resting
energy expenditure, going above and beyond the results of previous studies. The very promising
results from this study lead the authors to conclude that cachexia can be countered through the
use of a nutritional supplement enriched with EPA.
Following this encouraging study, Barber decided to look into some of the metabolic
components of cachexia to see how fish oil affected these pathways. In 2001, he published
another study using the same fish oil enriched nutritional supplement, this time looking at how
the supplement effected metabolic mediators that are known to have a role in cachexia (Barber,
2001). Barber provided a supplement that contained 2g of EPA and 600 kcal to 20 patients with
pancreatic cancer who were to consume it daily. Barber reported that, “At baseline and at 3
weeks, patients were weighed and serum concentration samples were taken to measure IL-6,
cortisol, insulin, leptin, blood mononuclear cell production of IL-1B, IL-6, and TNF-a, and
urinary excretion of proteolysis inducing factor (PIF) (Barber, 2001).”
Interestingly, Barber found that after 3 weeks of fish oil supplementation, levels of IL-6
were significantly decrease, the serum insulin concentration was elevated, the cortisol-to-insulin
ratio decreased, and there was also a reduction in the number of patients excreting PIF in the
urine. Notably, there was also weight gain in addition to the beneficial changes in these
metabolic mediators. In conclusion, Barber stated that the fish oil-enriched supplement
modulates different mediators involved in cachexia catabolism. Normalization of cancer
associated metabolic changes, that normally prevent weight gain, are suggested to be brought
15
about by the fish oil in the supplement (Barber, 2001). Here we begin to see some of the
mechanisms by which fish oil exerts its positive anti-cachexia effects, specifically through the
modulation of key metabolic pathways. The last three studies by Barber build upon each other
and present a strong argument for the use of omega-3 fatty acids in preventing cachexia in cancer
patients.
Further strengthening the argument for the use of fish oil to treat cachexia is a study that
was published in 2001 by Hardman. The purpose of this study was to determine if concentrated
fish oil will be a useful adjuvant treatment for chemo, as well as to determine if the concentrated
fish oil would cause detrimental increases in the drug toxicity to either the host or tumor cell
(Hardman, 2001). The methods of this study used MDA-MB 231 breast cancer cell xenografts
and implanted them into 120 athymic mice. The mice were fed a normal diet for three weeks to
allow the tumor to develop. At the three-week mark, the mice were randomly divided into two
dietary groups. The first group receiving the standard diet of 5% corn oil, and the second group
receiving a diet containing 3% fish oil concentrate and 2% corn oil. The mice were kept on their
respective diets for two weeks at which point mice from each group were randomly selected to
receive doxorubicin (DOX) therapy for a total of five weeks. Tumor size was measured weekly.
The data from the results of this experiment showed that the 3% FOC group increased the
efficacy of doxorubicin against tumor growth without causing an increase in toxicity to the host
cells. Additionally, “Consumption of the 3% FOC alone was as effective against the growth of
the MDA-MB 231 breast carcinoma as was DOX chemotherapy with the 5% corn oil diet
(Hardman, 2001).” These results are intriguing, showing that a fish oil concentrate increased the
effectiveness of doxorubicin chemotherapy without causing toxicity to the host. Even more
16
interesting, this study showed that fish oil concentrate alone was just as effective as
chemotherapy in preventing tumor growth in breast cancer xenografts.
The next study to be reviewed looked at the use of fish oil to treat colorectal cancer. In
2003, Llor published a study in the Journal of Clinical Nutrition that looked at how fish oil
affects colorectal metabolic processes in comparison to olive oil, linoleic acid, and oleic acid
(Llor, 2003). The purpose of this study was to determine the function of these fats in the process
of colorectal carcinogenesis. The authors used both Caco-2 and HT-29 colon adenocarcinoma
cell lines. These cells were then supplemented with fish oil (50% EPA and DHA), olive oil, oleic
acid, or linoleic acid. At 48 and 72 hours after the addition of the fats, the cells were tested for
apoptosis, proliferation, differentiation, and Cox-2 and Bcl-2 expression was assessed.
The results showed that both fish oil and olive oil increased the induction of apoptosis in
both colon cancer cell lines. Fish oil supplementation drastically decreased cell proliferation in
the Caco-2 and HT-29 cell lines. All four of the fats increased cellular differentiation of the
Caco-2 cells. Lastly, both COX-2 and Bcl-2 expression was downregulated in both the fish oil
and olive oil supplemented cells. The authors conclude by stating, “Fish oil and olive oil alter
different cellular processes leading to prevention of colorectal cancer development. COX-2 and
Bcl-2 could be partially responsible for some of the observed effects (Llor, 2003).” Notably, fish
oil not only prevented further cell proliferation of these cancer cells, but it also increased cellular
differentiation and apoptosis. These results help to explain some of the metabolic pathways that
are mediated by fish oil and shows us that fish oil effects gene expression of specific pro-
carcinogenic genes such as COX-2 and Bcl-2.
The final and most recent study supporting the use of omega-3 PUFAs to treat cancer
cachexia is a study by Murphy et al (Murphy, 2011). The purpose of Murphy’s study was to
17
assess the effect of a fish oil intervention on body composition and weight compared to standard
of care during chemotherapy. There were forty patients in this study that were divided into two
groups. Sixteen patients were placed in the fish oil (2.2g of EPA/day) group and the remaining
24 were placed into the standard of care group. Body composition was measured using computed
tomography imaging. Blood and weight measurements were taken at baseline and throughout
chemotherapy.
The authors found that those in the standard of care group had an average weight loss of
2.3 kg, however those in the fish oil group maintained their weight. Secondly, the authors
reported that those who had the highest plasma EPA levels showed the greatest muscle gains.
There was a maintenance or gain in muscle mass in 69% of patients in the fish oil group. In
comparison, there was a maintenance of muscle mass in only 29% of patients in the standard of
care group (Murphy, 2011). Here we see that fish oil was superior to standard of care treatment
for maintaining body composition. Finally, the authors conclude that, “Nutritional intervention
with 2.2g of fish oil per day appears to provide a benefit over standard of care, resulting in
maintenance of weight and muscle mass during chemotherapy (Murphy, 2011).” This study
shows us that fish oil helps to maintain muscle mass in cancer patients, even during
chemotherapy treatment, very promising results for individuals with cancer cachexia.
Adding support to the argument for omega 3 PUFAs to treat cachexia is a double-blind,
placebo-controlled study looking that looked at advanced lung cancer patients (Finocchiaro,
2012). This study looked into how EPA and DHA would effect inflammation and oxidative
stress in lung cancer patients. They randomly divided 33 patients with non-small-cell lung cancer
into two groups. The first group received 850 mg of EPA/DHA and the second group received a
18
placebo. The subjects were followed for 66 days and biochemical and anthropometric
measurements were taken at day 8, 22, and 66.
The results showed a significant reduction in inflammatory cytokines CRP, TNF-a, and
IL-6 in the n-3 group. Additionally, there was a significant reduction in plasma ROS compared
to the placebo group. Lastly, the study showed a slight increase in protein and energy intake in
the n-3 group and a significant increase in body weight in the treatment group. These positive
results support the use of n-3 PUFAs in cachexia treatment, however, the authors concluded that
there is still not enough clinical evidence to justify using n-3 PUFAs in a clinical setting.
Following this study was a study out of Taiwan that looked at omega-3 fatty acids, in
combination with micronutrients and probiotics, for their effects on weight gain in head and neck
cancer patients (Yeh, 2013). The authors examined the effectiveness of a nutritional supplement
for modifying body weight, albumin and prealbumin levels, and survival. After being randomly
assigned to receive either the nutritional supplement or an isocaloric control supplement, 68
subjects were followed for 3 months. Changes in body weight and albumin levels were
monitored.
The results indicated that the group taking the nutritional supplement enriched with n-3
fatty acids maintained their body weight throughout the treatment period whereas the control
group did not. Additionally, the treatment group had significantly improved albumin and
prealbumin levels compared to the control group. The ability of patients to maintain body weight
seen in this study supports an anti-cachectic effect of n-3 fatty acids. Moreover, a rise in protein
levels suggest another benefit of n-3 fatty acids and endorses their use in cachexia patients.
Lastly, a randomized control trial was published in 2014 that compared the use of an EPA
enriched nutritional supplement versus an isocaloric control to treat clinical and biochemical
19
outcomes in patients with advanced non-small-cell lung cancer (Sanchez-Lara, 2014). This study
looked at the effects on body weight, inflammation, energy intake, quality of life, chemotherapy
response, and survival. The 96 patients were randomized and followed for 8 weeks, with
nutritional, clinical, and biochemical measurements taken after each round of chemo.
The omega-3 fatty acid supplemented group was able to maintain their weight and even
significantly increased their lean body mass, whereas the control group showed the opposite.
Inflammation was also improved in the treatment group, which displayed a significant reduction
in CRP and TNF-a levels, while the control group showed no changes. Energy and protein intake
was reduced in the control group but significantly increased in the treatment group. Additionally,
the treatment group showed reduced appetite loss and fatigue and an increased quality of life
compared to the control group. There were no significant differences in responses to
chemotherapy between groups.
In summary, the previous nine studies have created a very strong argument for the
effectiveness of omega-3 fatty acids found in fish oil to prevent cancer cachexia. We have seen
that fish oil has been effective in reversing cachexia in many different types of cancer including
pancreatic, breast, colon, and lung cancers. These studies have shown fish oil to inhibit weight
loss in a cachexia model, to delay the progression of cachexia in pancreatic cancer, and to restore
immunodeficiency and prolong survival of malnourished patients. Additionally, fish oil enriched
nutritional supplements caused pancreatic cancer patients to gain weight. Fish oil has also shown
to positively affect metabolic mediators of cachexia such as cytokine production, in addition to
being able to modulate gene expression of key mediators of cachexia such as COX-2 and Bcl-2.
Furthermore, these studies have shown that omega-3 PUFAs from fish oil increase the
effectiveness of chemotherapy such as doxorubicin without causing toxicity to the host cell, in
20
addition to fish oil providing a benefit over standard of care for preserving muscle mass and
improving body composition in patients with advanced cachexia. Most recently, randomized
control trials have showed us that omega-3 fatty acids are effective at reducing inflammation and
ROS levels, increasing total energy intake, and maintaining body weight, with the most recent
study showing an increase in lean body mass and an improvement in quality of life. When all of
these studies are taken into account we cannot deny that there is very strong evidence supporting
the use of omega-3 PUFAs from fish oil for the treatment of cancer cachexia. However, before
any conclusion can be made, the opposing side of this argument must be presented and all
relevant literature will be reviewed and discussed.
Review of Refuting Articles
The earliest study that will be discussed was published in 1992 by Calder and
Newsholme (Calder, 1992). The aim of this study was to study the effects of fatty acids on
proliferation of blood lymphocytes and on IL-2 concentration. The authors wanted to look at the
effects on the immune system because numerous studies have shown fatty acids to be able to
modulate the immune system. Blood was drawn from five female subjects and transferred to
Leucoprep tubes for centrifugation. PUFAs were added to the tubes and after 48 hours the blood
was assayed for lymphocyte proliferation and IL-2 levels.
The results from the assays showed that EPA caused a significant inhibition of blood
lymphocyte proliferation. In addition, EPA also significantly decreased the concentration of IL-2
in blood lymphocytes. These findings are important to discuss because they cause a reduction in
the immune system after the addition of PUFAs. Lymphocyte proliferation has been shown to be
dependent on its ability to secrete IL-2. In this study PUFAs were shown to prevent the secretion
21
of IL-2, therefore halting lymphocyte proliferation and weakening the immune system. This
raises serious concerns for cancer patients who are already severely immunocompromised.
The following year another study was published that also looked at omega-3s effect on
the immune system, specifically IL-2 and mononuclear cells (Endres, 1993). This study by
Endres reinforced the previous study, finding similar results. Eighteen male volunteers were
given 18g (Each gram containing 153mg EPA and 103 mg DHA) of lipid concentrate per day
and continued this for six weeks. Blood was drawn at baseline and at the six-week mark and was
assessed for mononuclear cell proliferation and IL-2 production.
IL-2 production was remarkably reduced by 65% compared to baseline levels.
Furthermore, mononuclear cell proliferation mirrored the IL-2 response and showed a 70%
decrease compared to baseline. While these results may be useful in diseases that show an
overexpression of the immune system such as rheumatoid arthritis, these effects would be
detrimental to cancer patients who are already severely immunocompromised. The reduction in
immune function seen in these two early studies highlights one of the negative side effects of fish
oil supplementation.
The next study that will be reviewed showed that EPA had no effect on preventing
cachexia (Costelli, 1995). This study assessed if omega 3 polyunsaturated fatty acids, especially
EPA, effectively prevents hypercatabolism in tissues or changes tumor growth rates. Male wistar
rats were injected with Yoshida AH-130, a rapid growing hepatoma that causes weight loss and
tissue wasting. They were randomly divided into two groups, the first receiving daily EPA and
the second receiving a control diet. At 0, 4, and 7 days after injection of the hepatoma, the mice
were killed and tumor volume was assessed.
22
The hepatoma created a rapid loss of body weight and muscle in the rates. The rats in the
group receiving the EPA showed no alteration to the tumor growth rate compared to the control
group. Furthermore, the EPA was also ineffective at reducing the rate of wasting as compared to
the control group. The authors conclusion states, “The use of n-3 PUFA to prevent cancer
associated cachexia may be limited by the type of tumor. Very undifferentiated tumors are
probably insensitive to growth modulation by such agents (Costelli, 1995).” This study shows no
effect of EPA in reducing cachexia, at least in this tumor type, and weakens the argument for
using n-3 PUFAs to treat cachexia.
Moving ahead three years, we go to a study looking into the effects of PUFAs on colon
cancer in rats (Griffini, 1998). This study is one of the strongest advocates against the use of
omega 3 PUFAs in cancer patients. The purpose of this study was to investigate the effects of n-3
and PUFAs on the development of colon carcinoma metastasis in rat liver (Griffini, 1998). Wag-
Rij rats were divided into three dietary groups, one receiving a fish oil diet, one receiving a
safflower oil diet, and the last group receiving a low fat control diet. The rats were maintained on
their diets for three weeks. At the three-week mark, CC531 colon carcinoma cells were
transplanted into the rats. At 1 and 3 weeks, rats were euthanized and liver metastasis was
measured.
The data showed that at 1 week after transplantation there were 7x more metastasis in the
fish oil group compared to the low fat diet group. Additionally, at 3 weeks after transplantation
there were 10x more metastasis in the fish oil group compared to the low fat diet group. Adding
to these alarming results, the size of the metastasis in the fish oil group were 1000 fold larger
than those found in the low fat diet group. Griffini concludes, “N-3 PUFAs promote colon cancer
metastases in the rat liver. This finding has serious implications for the treatment of cancer
23
patients with fish oil to fight cachexia (Griffini, 1998).” This study is very alarming, showing
that n-3 PUFAs not only promoted extreme increases in number of metastases but also in the size
of the metastases.
In 2000 the Journal of Clinical Nutrition published a study out of the Netherlands that
investigated the effect of EPA on rates of lipolysis and lipid oxidation in cachectic patients and
healthy individuals (Zuijdgeest-Van Leeuwen, 2000). This was being investigated because it was
proposed that weight loss in cancer patients can be prevented through lipolysis inhibition. There
were 17 cancer patients and 16 healthy subjects, all randomized to receive either a EPA (6g/day)
or a placebo. The subjects were followed for seven days, with whole body lipolysis, palmitate
oxidation, and palmitic acid release being measured at baseline, day 2, and day 7 of the
intervention.
The authors reported no significant differences in lipolysis between groups. Secondly,
palmitate oxidation and palmitic acid release did not differ between the two groups. In
conclusion, the authors state, “Supplementation of EPA does not significantly inhibit lipolysis or
lipid oxidation in weight-losing cancer patients or in healthy subjects (Zuijdgeest-Van Leeuwen,
2000).” By showing fish oil is ineffective at reducing wasting of adipose tissue and preventing
lipolysis in cachectic patients, this study refutes the use of omega-3 PUFAs in treating cachexia.
The ensuing study in this review is just as bleak as the previous by Van Leeuwen. In
2003, Gut published a randomized double blind trial by Fearon et al. that assessed the effect of a
n-3 enriched supplement on weight loss (Fearon, 2003). This study compared a protein and
energy dense supplement enriched with n-3 fatty acids with an isocaloric control supplement.
They tested for effects on weight, lean body mass, dietary intake, and quality of life in cachectic
24
patients (Fearon, 2003). There were 200 patients in this trial, randomized to either receive the n-3
fatty acid or the control supplement and followed for eight weeks.
At the end of the 8-week intervention period there was no significant difference in the
two groups in terms of weight loss and lean body mass. Reduction in the rate of weight loss was
the same in both groups. Changes in lean body mass were also similar in both groups. The
authors concluding statement said, “Enrichment with n-3 fatty acids did not provide a therapeutic
advantage and both supplements were equally effective in arresting weight loss (Fearon, 2003).”
There was not a clear advantage and no difference in outcomes between the experimental and
control arms. We can see from this study that the use of omega-3 fatty acids to prevent cachexia
would be ill advised as it provides no additional benefit when compared to traditional treatments.
Jatoi lead a similar study in 2004 that investigated whether an EPA supplement would be
more effective than megestrol acetate at augmenting appetite and weight gain in patients with
cancer associated cachexia (Jatoi, 2004). This study randomly assigned 421 cachectic patients to
one of three groups. The first group received 1.09g EPA supplement plus a placebo, the second
group received 600 mg/d megestrol acetate plus an isocaloric supplement, and the third group
received both the EPA supplement as well as megestrol acetate. Weight measurements and
physical examinations were taken at baseline and weekly.
In terms of appetite improvement, there was no significant difference between the three
treatment arms. However, both of the treatment arms receiving megestrol acetate showed an
increase in functional assessment compared to the fish oil treatment arm. In addition, there was a
smaller percentage of patients who gained greater than 10% of baseline weight in the EPA group
compared to those in both megestrol acetate treatment groups. Lastly, there was no significant
differences in quality of life or survival between the two groups. In conclusion, “This EPA
25
supplement, either alone or in combination with megestrol acetate, does not improve weight or
appetite better than megestrol acetate alone (Jatoi, 2004).” EPA supplementation showed zero
improvements in any of the parameters measured compared to the other treatment arms. This
suggests no benefit in using omega-3 PUFAs to treat cancer cachexia.
Next up in the review of studies opposing the use of fish oil to treat cancer cachexia is a
randomized pilot study looking at advanced gastrointestinal cancer patients (Persson, 2005). The
objective of this pilot study was to investigate the effect of fish oil supplementation in
combination with melatonin and dietary advice on the biochemical variables known to develop
with cachexia, specifically cytokines. In addition, the authors examined clinical outcomes such
as body weight, quality of life, and food intake. The twenty-four patients were randomized to
receive either fish oil (4.9g DHA and 3.2g DHA), melatonin, or a combination. At baseline and
at 4 weeks plasma was analyzed for TNF-a, IL-6, IL-1B, IL-8, DHA, EPA, linoleic acid, and
arachidonic acid (Persson, 2005).
The results showed that there was no significant difference in any of the cytokines and no
difference in biochemical variables between the fish oil and melatonin groups. Moreover, there
was not a significant difference in energy intake, functional performance status, or survival
between the two groups. The authors conclude, “Fish oil, melatonin, and their combination, did
not produce substantial anti-inflammatory effects in cachectic patients with advanced
gastrointestinal cancer (Persson, 2005).” No benefits in inflammatory cytokine concentrations
were seen when fish oil was supplemented, contrary to what has been shown in previous fish oil
studies. These results oppose the use of fish oil for treating cancer cachexia, due to an absence of
an anti-inflammatory effect that they are commonly associated with.
26
The last study that will be covered in this review was published in 2006 in the Journal of
Clinical Oncology (Fearon, 2006). This article was double-blind, randomized, and placebo-
controlled. The purpose of this study was to compare EPA diester against a placebo for their
effects on lean body mass and weight in an eight-week intervention. This was a large study, with
518 patients being randomly assigned to one of three treatment arms to be followed for eight
weeks. The first group received 2g of EPA daily (n=175), the second group received 4g EPA
daily (n=172), and the final group received a placebo (n=171). Assessments were taken at 4 and
8 weeks.
The results indicated no significant changes in weight, survival, or nutritional variables.
Fearon concludes, “Overall, no statistically significant effects of treatment on the primary
endpoint of weight were observed…There were no other obvious differences in nutrition or
quality of life measures as a result of EPA diester administration...The results indicate no
significant benefit from single agent EPA in the treatment of cancer cachexia (Fearon, 2006).”
The current study, reporting no changes in body weight or quality of life by the addition of EPA,
illustrates that EPA is not useful for preventing cancer cachexia and provides a strong argument
against its use in this population.
Taking all of these studies into account we can see that there is also a strong argument
against the use of omega-3 PUFAs to treat cancer cachexia. Opposing the views of the previous
set of studies, these articles demonstrate that fish oil can potentially be quite harmful and
advocate that it should not be used to treat cachexia. The data from these original articles have
shown that fish oil can reduce the immune system by suppressing IL-2 production and
lymphocyte proliferation, has increased the number and size of colon metastases in an animal
model, is ineffective at preventing lipolysis, and is simply ineffective at modifying many clinical
27
outcomes such as quality of life, performance status, and survival. It is very important to
consider the results of these studies when assessing the use omega-3 fatty acids to treat patients
with cancer-induced cachexia.
Discussion
This area of study has become a major controversy, especially in clinical practice. While
there are numerous studies showing that fish oil has positive effects on clinical outcomes of
cachexia, there are also just as many studies showing the opposite. For every study that came out
supporting the use of fish oil to treat cachexia, another article followed refuting its use. Because
of this dichotomy in the literature, oncologists have become torn on this issue, some saying that
yes, we should supplement fish oil in cachectic patients, whilst other clinicians are very opposed,
saying that it has no benefits and will only add to the financial burden of the patient. In addition
to the financial burden, some clinicians argue that it is already difficult to get cancer patients to
eat because of their loss of appetite and adding fish oil into their treatment plan will only
complicate this issue further. So what conclusions can be drawn after reviewing both sides of the
literature?
From reviewing both the supporting and opposing articles we can see that there is strong
evidence supporting both sides of this debate. In order to decide which side is more convincing
we must take a deeper look at each study and carefully examine the methodology and results and
compare strengths and weaknesses of each study. We can see that omega-3 fatty acids show very
positive results in the early cell and animal studies. However, when we look at the human studies
we can see that the results were a lot dimmer. Because the larger human trials show negative
results, I am persuaded to support the side against the use of omega-3 fatty acids. Cell and
28
animal studies are important for understanding the mechanisms, but phase 3 human studies are
much more applicable to a clinical setting. Cell and animal studies are easy to control for
confounding variables, but studying nutrition in humans can be very difficult because of our free
will. I will discuss both sides and why I believe that there currently is not enough evidence to
support the use of omega-3 fatty acids to treat cancer cachexia.
As discussed previously, almost all cell and animal models show that omega 3 PUFAs
produce a beneficial effect in a cachexia model. In cellular studies they were shown to reduce
inflammatory cytokine levels such as CRP, TNF-a, IL-1, and IL-6 and to reduce PIF activation
(Rondanelli, 2008). We have also seen that they can suppress the ubiquitin-proteasome pathway
and suppress the cancer cachetic factor (Jatoi, 2005). However, we also know that the immune
system can be compromised with the use of n-3 fatty acids. Lymphocyte proliferation was halted
in one study (Calder, 2002), and mononuclear cell production was greatly reduced in another
(Endres, 1993). Cancer patients typically have very weakened immune systems already so we
must carefully consider these studies when deciding whether or not to supplement n-3 fatty
acids. In terms of cell studies, the majority of the evidence supports the use of omega-3 fatty
acids for cachexia patients, but the possible reduction in immunity must be taken into account.
When we look at animal studies we see a similar pattern. The majority of evidence is in favor of
the use of omega-3 fatty acids to treat cachexia. However, one study did show 10x greater
number of colon metastasis in rats supplemented with fish oil compared to the control group
(Griffini, 1998). Cell and animal models, while important and useful, can only tell us so much.
The results of human studies have much more weight on the clinical applicability.
When we look at the earlier human studies we can see that there are many flaws. Many of
the studies had small number of subjects meaning the results can’t be generalized to the total
29
population group. The study by Wigmore only had 18 patients, the study by Gogos only had 15
subjects (Gogos, 1998), the study by Barber only had 18 subjects (Barber, 1999), and the study
by Barber in 2001 only had 20 patients (Barber, 2001). Although these sample sizes are
somewhat small we also have to keep in mind that there are many factors affecting participation
when dealing with advanced cancer patients. It is very difficult to get very sick patients to
volunteer for studies when they are undergoing chemotherapy and radiation, and this is also why
there is low compliance and a high dropout rate in some of these studies. While this is something
that can be very difficult to improve upon, it still remains a flaw of many of these studies
supporting the use of fish oil.
Furthermore, when we look at some of the larger clinical trials in humans we see that
they show no effect. A study using 91 patients compared fish oil to olive oil on its ability to
affect clinical outcomes in cachexia patients. In this well designed study there was absolutely no
change in appetite, energy intake, or functional status between the groups (Bruera, 2003).
Another large clinical trial (N=200) showed no difference in rate of weight loss between an
EPA-enriched supplement group and an isocaloric supplement group (Fearon, 2003). Lastly, a
large double blind study (N=421) showed that EPA had no added benefit for treating cachexia
compared to megestrol acetate alone (Jatoi, 2004). Each of these studies had much larger sample
sizes and better study designs than the earlier human studies. Some would argue that the dose of
fish oil was too small to see an effect and a weakness in these studies, but I do not believe that
this is the case. For this reason, these later trials hold more weight than the earlier trials showing
positive results. Based on this information, I am convinced that the use of omega-3 fatty acids is
ill advised for patients with cachexia.
30
The most recent randomized clinical trials began to show a trend in support of the use of
omega-3 fatty acids to treat cachexia. However, in one study, the intervention involved n-3
PUFAs, probiotics, and micronutrients (Yeh, 2013). It cannot be said for certain that the positive
results of this study were due to the omega-3 fatty acids. Another recent study showed that the
group receiving n-3 fatty acids showed a significant decrease in inflammatory cytokine and ROS
levels and an increase in energy intake (Finocchiaro, 2012). However, in their discussion the
authors state that they think the differences seen between the two groups are random and not
caused by the treatment. It is important to look closely at each study before drawing conclusions.
Conclusion
In summary, I believe that there is not enough evidence to support the use of omega-3
fatty acids for treating cancer-induced cachexia. While there are ample amounts of research
showing beneficial effects of omega-3 fatty acids, specifically in cell and animal models, the
larger, phase-3 clinical trials in humans show negative results and are more convincing.
Furthermore, after considering flaws in study design of the smaller human trials, I am more
swayed by the larger clinical trials that have better study designs and superior methodology.
Based on the reviewed evidence, omega-3 fatty acids may or may not have an effect depending
on numerous variables. Supplementing with fish oil, while being relatively safe and having very
minimal side effects, is simply not supported by the literature at this time. The answer is not a
definitive no at this point, but adding fish oil into a treatment regimen for an individual with
advanced cancer can possibly create more problems than it would solve and is ill advised. While
it can be highly appealing to some looking for anything to relieve the deleterious effects of
cachexia, we do not know enough about it yet and further research is necessary.
31
Future Research
While there are currently hundreds of studies on this subject and our understanding of
certain aspects of it are plentiful, we must continue to develop future research in order to fully
understand this phenomenon before we can come to a definitive conclusion. The scientific
community has made leaps and strides in understanding how omega 3 fatty acids work over the
last 50 years, however, there are still some specific questions that we have yet to answer. In
addition to these questions, I believe that future studies should change the way certain parts of
the study methodology is carried out.
Future studies should address some of the following questions. How does the stage of
cachexia progression affect the effectiveness of omega 3 supplementation? For example, what
will the difference in outcome be if treatment is started in the pre-cachexia stage, vs. the cachexia
stage, vs. the refractory cachexia stage? Future studies also need to examine how other clinical
and nutritional variables play a role in how effective omega-3 fatty acids will be. How do
comorbidities, genotype, epigenetics, environment, and other dietary factors affect the outcome
of treatment? Furthermore, it would be beneficial to have a deeper understanding of the
underlying mechanisms omega 3s use to exert their effects in order to create more effective
treatment options. Lastly, there is simply a need for additional large scale, randomized clinical
trials in humans.
In terms of the methodology, all efforts in the future should be placed on making the
studies as equal as possible so that comparisons can be made. In the past, study designs are rarely
comparable and most studies are highly heterogeneous. Tolerability of the EPA/DHA source
should be made more tolerable so that there will be less dropout. Moreover, compliance with the
treatment needs to be better addressed in future studies to ensure the desired treatment effect is
32
taking place. Making these changes will ensure that more subjects are able to complete the trial
and the results are more legitimate.
References
Barber, M. D. (2001). Cancer Cachexia and Its Treatment With Fish-Oil-Enriched Nutritional
Supplementation Nutrition, 17, 751-755.
Barber, M. D., Fearon, K.C., Tisdale, M.J., McMillan, D.C., & Ross, J.A. (2001). Effect of a
Fish-Oil Enriched Nutritional Supplement on Metabolic Mediators in Patients With
Pancreatic Cancer Cachexia Nutrition and Cancer, 40(2), 118-124.
Barber, M. D., Ross, J.A., Preston, T., Shenkin, A., & Fearon, K.C. (1999). Fish Oil-Enriched
Nutritional Supplement Attenuates Progression of the Acute-Phase Response in Weight-
Losing Patients with Advanced Pancreatic Cancer The Journal of Nutrition, 129, 1120-
1125.
Barber, M. D., Ross, J.A., Voss, A.C., Tisdale, M.J., & Fearon, K.C. (1999). The effect of an
oral nutritional supplement enriched with fish oil on weight-loss in patients with
pancreatic cancer British Journal of Cancer, 81(1), 80-86.
Bruera, E., Strasser, F., Palmer, J.L., Willey, J., Calder, K., Amyotte, G., & Baracos, V. (2003).
Effect of Fish Oil on Appetite and Other Symptoms in Patients With Advanced Cancer
and Anorexia/Cachexia: A Double-Blind, Placebo-Controlled Study Journal of Clinical
Oncology, 21(1), 129-134.
Burns, C. P., Halabi, S., Clamon, G.H., Hars, V., Wagner, B.A. Hohl, R.J., ... Paskett, E. .
(1999). Phase I Clinical Study of Fish Oil Fatty Acid Capsules for Patients with Cancer
Cachexia: Cancer and Leukemia Group B Study 9473. Clinical Cancer Research, 5,
3942-3947.
Calder, P. C., & Newsholme, E.A. (1992). Polyunsaturated fatty acids suppress human peripheral
blood lymphocyte proliferation and interleukin-2 production Clinical Science, 82, 695-
700.
Chang, W. L., Chapkin, R.S., & Lupton, J.R. (1998). Fish Oil Blocks Azoxymethane-Induced
Rat Colon Tumorigenesis by Increasing Cell Differentiation and Apoptosis Rather Than
Decreasing Cell Proliferation The Journal of Nutrition, 128, 491-497.
Cockbain, A. J., Toogood, G.J., & Hull, M.A. (2011). Omega-3 polyunsaturated fatty acids for
the treatment and prevention of colorectal cancer. Gut, 1-15.
33
Colomer, R., Moreno-Nogueira, J.M., Garcia-Luna, P.P., Garcia-Peris, P., Garcia-de-Lorenzo,
A., Zarazaga, A., ... Casimiro, C. . (2007). n-3 Fatty acids, cancer and cachexia: a
systematic review of the literature. British Journal of Nutrition, 97, 823-831.
Costelli, P., Llovera, M., Lopez-Soriana, J., Carbo, N., Tessitore, L., Lopez-Soriano, F.J., ...
Argiles, J.M. (1995). Lack of effect of eicosapentaenoic acid in preventing cancer
cachexai and inhibiting tumor growth Cancer Letters, 97, 25-32.
Dewey, A., Baughan, C., Dean, T.P., Higgins, B, & Johnson, I. (2012). Eicosapentaenoic acid
(EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia
Cochrane Database of Systematic Reviews(1).
Endres, S., Meydani, S.N., Ghorbani, R., Schnigler, R., & Dinarello, C.A. (1993). Dietary
Supplementation with n-3 fatty acids suppresses interleukin-2 production and
mononuclear cell proliferation Journal of Leukocyte Biology, 54(6), 599-603.
Fearon, K., Strasser, F., Anker, S.D., Bosaeus, I., Bruera, E., Fainsinger, R.L., ... Baracos, V.E.
(2011). Definition and classification of cancer cachexia: an international consensus
Lancet, 12, 489-495.
Fearon, K. C., Barber, M.D., Moses, A.G., Ahmedzai, S.H., Taylor, G.S., Tisdale, M.J., &
Murray, G.D. (2006). Double-Blind, Placebo-Controlled, Randomized Study of
Eicosapentaenoic Acid Diester in Patients With Cancer Cachexia Journal of Clinical
Oncology, 24(21), 3401-3407.
Fearon, K. C., Glass, D.J., Guttridge, D.C. (2012). Cancer Cachexia: Mediators, Signaling, and
Metabolic Pathways Cell Metabolism, 16, 153-166.
Fearon, K. C., von Meyenfeldt, M.F., Moses, A.G., van Geenen, R., Roy, A., Gouma, D.J., ...
Tisdale, M.J. (2003). Effect of a protein and energy dense n-3 fatty acid enriched oral
supplement on loss of weight and lean tissue in cancer cachexia: a randomised double
blind trial. Gut, 52, 1479-1486.
Fetterman Jr., J. W., & Zdanowicz, M.M. (2009). Therapeutic potential of n-3 polyunsaturated
fatty acids in disease. American Journal of Health-System Pharmacy, 66, 1169-1179.
Finocchiaro, C., Segre, O., Fadda, M., Monge, T., Scigliano, M., Schena, M., & Tinivella, M.
(2012). "Effect of n-3 fatty acids on patients with advanced lung cancer: a double blind,
placebo-controlled study " British Journal of Nutrition 108: 327-333.
Furst, P., & Kuhn, K.S. (2000). Fish oil emulsions: what benefits can they bring? . Clinical
Nutrition, 19(1), 7-14.
Giacosa, A., & Rondanelli, M. . (2008). Fish oil and treatment of cancer cachexia Genes and
Nutrition, 3, 25-28.
Gogos, C. A., Ginopoulos, P., Salsa, B., Apostolidou, E., Zoumbos, N.C., & Kalfarentzos, F.
(1998). Dietary Omega-3 Polyunsaturated Fatty Acids plus Vitamin E Restore
34
Immunodeficiency and Prolong Survival for Severely Ill Patients with Generalized
Malignancy Cancer, 82, 395-402.
Griffini, P., Fehres, O., Klieverik, L., Vogels, I.M., Tigchelaar, W., Smorenburg, S.M., & Van
Noorden, C.J. (1998). Dietary n-3 Polyunsaturated Fatty Acids Promote Colon
Carcinoma Metastasis in Rat Liver. Cancer Research, 58, 3312-3319.
Hardman, W. E. (2002). Omega-3 Fatty Acids to Augment Cancer Therapy The Journal of
Nutrition, 132, 3508-3512.
Hardman, W. E., & Avula, C.P. (2001). Three Percent DIetary Fish Oil Concentrate Increased
Efficacy of Doxorubicin Against MDA-MB 231 Breast Cancer Xenografts Clinical
Cancer Research, 7, 2041-2049.
Jatoi, A. (2005). Fish oil, lean tissue, and cancer: is there a role for eicosapentaenoic acid in
treating the cancer anorexia/weight loss syndrome? . Critical Reviews in
Oncology/Hematology, 55, 37-43.
Jatoi, A. (2005). n-3 Fatty Acid Supplements for Cancer-Associated Weight Loss. Nutrition in
Clinical Practice, 20, 394-399.
Jatoi, A. R., K., Loprinzi, C.L., Sloan, J.A., Dakhil, S.R., Macdonald, N., ... Christensen, B.
(2004). An Eicosapentaenoic Acid Supplement Versus Megestrol Acetate Versus Both
for Patients With Cancer-Associated Wasting: A North Central Cancer Treatment Group
and National Cancer Institute of Canada Collaborative Effort Journal of Clinical
Oncology, 22(12), 2469-2476.
Llor, X., Pons, E., Roca, A., Alvarez, M., Mane, J., Fernandez-Banares, F., & Gassull, M.A.
(2003). The effects of fish oil, olive oil, oleic acid, and linoleic acid on colorectal
neoplastic processes. Clinical Nutrition, 22(1), 71-79.
Murphy, R. A., Mourtzakis, M., Chu, Q.S., Baracos, V.E., Reiman, T., & Mazurak, V.C. (2011).
Nutritional Intervention With Fish Oil Provides a Benefit Over Standard of Care for
Weight and Skeletal Muscle Mass in Patients With Nonsmall Cell Lung Cancer
Receiving Chemotherapy Cancer, 117, 1775-1782.
Murphy, R. A., Yeung, E., Mazurak, V.C., & Mourtzakis, M. . (2011). Influence of
eicosapentaenoic acid supplementation on lean body mass in cancer cachexia British
Journal of Cancer, 105, 1469-1473.
Persson, C., Glimelius, B., Ronnelid, J., & Nygren, P. . (2005). Impact of fish oil and melatonin
on cachexia in patients with advanced gastrointestinal cancer: A randomized pilot study.
Nutrition, 21, 170-178.
Roodhart, J. M., Daenen, L.G., Stigter, E.C., Prins, H., Gerrits, J., Houthuijzen, J.M., ... Voest,
E.E. (2011). Mesenchymal Stem Cells Induce Resistance to Chemotherapy through the
Release of Platinum-Induced Fatty Acids. Cancer Cell, 20, 370-383.
35
Roynette, C. E., Calder, P.C., Dupertuis, Y.M., & Pichard, C. (2004). n-3 Polyunsaturated fatty
acids and colon cancer prevention Clinical Nutrition, 23, 139-151.
Sanchez-Lara, K., Turcott, J.G., Juarez-Hernandez, E., Nunez-Valencia, C., Villanueva, G.,
Guevara, P., & Torre-Vallejo, M. (2014). "Effects of an oral nutritional supplement
containing eicosapentaenoic acid on nutritional and clinical outcomes in patients with
advanced non-small cell lung cancer: Randomised trial " Clinical Nutrition 33: 1017-
1023.
Stewart, G. D., Skipworth, R.J., & Fearon, K.C. (2006). Cancer cachexia and fatigue Clinical
Medicine, 6(2), 140-143.
Tisdale, M. J. (2009). Mechanisms of Cancer Cachexia. Physiological Reviews, 89, 381-410.
Tisdale, M. J., & Dhesi, J.K. (1990). Inhibition of Weight Loss by n-3 Fatty Acids in an
Experimental Cachexia Model. Cancer Research, 50, 5022-5026.
Wigmore, S. J., Ross, J.A., Falconer, S., Plester, C.E., Tisdale, M.J., Carter, D.C., & Fearon,
K.C. (1996). The Effect of Polyunsaturated Fatty Acids on the Progress of Cachexia in
Patients with Pancreatic Cancer Nutrition, 12(1), 27-30.
Wu, D., & Meydani, S.N. . (1998). n-3 Polyunsaturated fatty acids and immune function
Proceedings of the Nutrition Society, 57, 503-509.
Yeh, K., Wang, H., Chang, J. W., Huang, J., Lai, C., Lan, Y., & Wu, T. (2013). "Omega-3 fatty
acid-, micronutrient-, and probiotic-enriched nutrition helps body weight stabilization in
head and neck cancer cachexia." Oral Surgery Oral Medicine Oral Pathology Oral
Radiology 116(1): 41-48.
Zuijdgeest-Van Leeuwen, S. D., Dagnelie, P.C., Wattimena, J.L., Van Den Berg, J.W., Van Der
Gaast, A., Swart, G.R., & Paul Wilson, J.H. (2000). Eicosapentaenoic acid ethyl ester
supplementation in cachectic cancer patients and healthy subjects: effects on lipolysis and
lipid oxidation. Clinical Nutrition, 19(6), 417-423.

More Related Content

What's hot

The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
guestdb1ebe
 
Lapband Seminar Port Lap Surgery
Lapband Seminar Port Lap SurgeryLapband Seminar Port Lap Surgery
Lapband Seminar Port Lap Surgery
guestc242dc
 
Presentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic SyndromePresentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic Syndrome
Josh Nooner
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
MD Specialclass
 

What's hot (17)

Nutritional managment of cachexia
Nutritional managment of cachexiaNutritional managment of cachexia
Nutritional managment of cachexia
 
Present management of obesity in adult
Present management of obesity in adultPresent management of obesity in adult
Present management of obesity in adult
 
Obesity and Low Carbohydrate Diets
Obesity and Low Carbohydrate DietsObesity and Low Carbohydrate Diets
Obesity and Low Carbohydrate Diets
 
Metabolic syndrome: an Asian perspective
Metabolic syndrome: an Asian perspectiveMetabolic syndrome: an Asian perspective
Metabolic syndrome: an Asian perspective
 
Obesity -pathophysiology,Evaluation & pharmacotherapy
Obesity -pathophysiology,Evaluation & pharmacotherapy Obesity -pathophysiology,Evaluation & pharmacotherapy
Obesity -pathophysiology,Evaluation & pharmacotherapy
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
 
Pharmacotherpy & Recent Advances in Obesity Management
Pharmacotherpy & Recent Advances in Obesity ManagementPharmacotherpy & Recent Advances in Obesity Management
Pharmacotherpy & Recent Advances in Obesity Management
 
Intermittent Fasting; Beneficial or Harmful?
Intermittent Fasting; Beneficial or Harmful?Intermittent Fasting; Beneficial or Harmful?
Intermittent Fasting; Beneficial or Harmful?
 
082409 understanding cachexia
082409 understanding cachexia082409 understanding cachexia
082409 understanding cachexia
 
Morbid obesity
Morbid obesityMorbid obesity
Morbid obesity
 
Lapband Seminar Port Lap Surgery
Lapband Seminar Port Lap SurgeryLapband Seminar Port Lap Surgery
Lapband Seminar Port Lap Surgery
 
Update on Obesity Management & Surgical Options
Update on Obesity Management & Surgical OptionsUpdate on Obesity Management & Surgical Options
Update on Obesity Management & Surgical Options
 
Presentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic SyndromePresentation - Omega-3 PUFAs and Metabolic Syndrome
Presentation - Omega-3 PUFAs and Metabolic Syndrome
 
Ueda 2016 bariatric surgery -fawzy el mosalamy
Ueda 2016 bariatric surgery -fawzy el mosalamyUeda 2016 bariatric surgery -fawzy el mosalamy
Ueda 2016 bariatric surgery -fawzy el mosalamy
 
Type 2 Diabetes and Cancer
Type 2 Diabetes and CancerType 2 Diabetes and Cancer
Type 2 Diabetes and Cancer
 
Malnutrition in the hospital
Malnutrition in the hospitalMalnutrition in the hospital
Malnutrition in the hospital
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
 

Similar to Efficacy of Omeg-3 Fatty Acids in Preventing Cancer Induced Cachexia

Phytosterols for Cancer Treatment
Phytosterols for Cancer TreatmentPhytosterols for Cancer Treatment
Phytosterols for Cancer Treatment
Josh Nooner
 
Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02
MD Specialclass
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
MD Specialclass
 
Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02
Saurabh
 
Lapband Seminar Port Lap Surgery
Lapband Seminar Port Lap SurgeryLapband Seminar Port Lap Surgery
Lapband Seminar Port Lap Surgery
guestc242dc
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
guestdb1ebe
 
Effect of the use of intragastric balloon to reduce weight in the management...
Effect of the use of intragastric balloon to reduce  weight in the management...Effect of the use of intragastric balloon to reduce  weight in the management...
Effect of the use of intragastric balloon to reduce weight in the management...
Shendy Sherif
 
obesity presentation american hospital
obesity presentation american hospitalobesity presentation american hospital
obesity presentation american hospital
heba abou diab
 
Esteatose Framminghan
Esteatose FramminghanEsteatose Framminghan
Esteatose Framminghan
docanto
 

Similar to Efficacy of Omeg-3 Fatty Acids in Preventing Cancer Induced Cachexia (20)

Obesity and cancer
Obesity and cancerObesity and cancer
Obesity and cancer
 
Phytosterols for Cancer Treatment
Phytosterols for Cancer TreatmentPhytosterols for Cancer Treatment
Phytosterols for Cancer Treatment
 
Phytosterols for cancer treatment
Phytosterols for cancer treatmentPhytosterols for cancer treatment
Phytosterols for cancer treatment
 
Nutrition Care process for Oncology Patients
Nutrition Care process for Oncology PatientsNutrition Care process for Oncology Patients
Nutrition Care process for Oncology Patients
 
Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
 
Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02Themanagementofobesity 100312193454 Phpapp02
Themanagementofobesity 100312193454 Phpapp02
 
Resurge - The Godzilla Of Offers - Resurge weight loss.
Resurge - The Godzilla Of Offers - Resurge weight loss.Resurge - The Godzilla Of Offers - Resurge weight loss.
Resurge - The Godzilla Of Offers - Resurge weight loss.
 
Lapband Seminar Port Lap Surgery
Lapband Seminar Port Lap SurgeryLapband Seminar Port Lap Surgery
Lapband Seminar Port Lap Surgery
 
Fishing Clues for the Efficacy of Chemotherapy Role of Fasting
Fishing Clues for the Efficacy of Chemotherapy Role of FastingFishing Clues for the Efficacy of Chemotherapy Role of Fasting
Fishing Clues for the Efficacy of Chemotherapy Role of Fasting
 
The Management Of Obesity
The Management Of ObesityThe Management Of Obesity
The Management Of Obesity
 
FInal Draft
FInal DraftFInal Draft
FInal Draft
 
Nutritional diseases (Obesity and starvation)
Nutritional diseases (Obesity and starvation)Nutritional diseases (Obesity and starvation)
Nutritional diseases (Obesity and starvation)
 
MULTIFACETED ROLES OF AUTOPHAGY IN CANCER & NEURODEGENERATIVE DISORDERS
MULTIFACETED ROLES OF AUTOPHAGY IN CANCER & NEURODEGENERATIVE DISORDERSMULTIFACETED ROLES OF AUTOPHAGY IN CANCER & NEURODEGENERATIVE DISORDERS
MULTIFACETED ROLES OF AUTOPHAGY IN CANCER & NEURODEGENERATIVE DISORDERS
 
Effect of the use of intragastric balloon to reduce weight in the management...
Effect of the use of intragastric balloon to reduce  weight in the management...Effect of the use of intragastric balloon to reduce  weight in the management...
Effect of the use of intragastric balloon to reduce weight in the management...
 
Cancer anorexia cachexia 23rd nov 2017 (1)
Cancer anorexia cachexia 23rd nov 2017 (1)Cancer anorexia cachexia 23rd nov 2017 (1)
Cancer anorexia cachexia 23rd nov 2017 (1)
 
1. Deepak Jain final for publication.pdf
1. Deepak Jain final for publication.pdf1. Deepak Jain final for publication.pdf
1. Deepak Jain final for publication.pdf
 
obesity presentation american hospital
obesity presentation american hospitalobesity presentation american hospital
obesity presentation american hospital
 
Esteatose Framminghan
Esteatose FramminghanEsteatose Framminghan
Esteatose Framminghan
 
Musculoskletal manifestations of Obesity
Musculoskletal manifestations of ObesityMusculoskletal manifestations of Obesity
Musculoskletal manifestations of Obesity
 

Efficacy of Omeg-3 Fatty Acids in Preventing Cancer Induced Cachexia

  • 1. 1 Efficacy of Omega-3 Polyunsaturated Fatty Acids for Preventing Cancer-Induced Cachexia Josh Nooner, BS, CSCS NSCI – 5843 2/29/16
  • 2. 2 Introduction Cachexia is an extremely debilitating condition that is seen in many chronic and terminal diseases such as cancer. This syndrome is characterized by unintentional weight loss, anorexia, and severe muscle wasting. Stewart reports, “20% of all cancer deaths are caused directly by cachexia (Stewart, 2006).” As the cause of death for 1 in 5 cancer patients, an individual who develops this wasting syndrome has a very poor prognosis. In fact, “Up to 50% of cancer patients suffer from a progressive atrophy of adipose tissue and skeletal muscle, called cachexia, resulting in weight loss, a reduced quality of life, and a shortened survival time (Tisdale, 2009).” As we can see, cachexia is a very prevalent condition, affecting half of all individuals diagnosed with cancer and is the direct cause of death for 20% of cancer deaths. Additionally, this syndrome results in a lower quality of life and a reduced survival time, making it an extremely important target for therapy. Cachexia is currently being heavily studied, as we aim to find an effective intervention strategy that will reduce the occurrence of this devastating syndrome, increase the quality of patient’s life’s, and lengthen their survival time. Omega-3 polyunsaturated fatty acids (PUFAs) are one area of research that is being investigated for their use in alleviating this wasting syndrome, specifically through their anti-inflammatory properties and their ability to mediate specific metabolic processes that are seen in cachexia. After studying the altered metabolic pathways that present in cachexia, we can better understand how the omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), work to alleviate cancer- induced cachexia. In order to better understand this syndrome, we must first define exactly what it is. In 2011 Fearon brought together an international panel of experts that formed a consensus on the definition, diagnosis, and the classification of cachexia. They reported that, “Cancer cachexia is
  • 3. 3 defined as a multifactorial syndrome characterized by an ongoing loss of skeletal muscle mass (with or without loss of fat mass) that cannot be fully reversed by conventional nutritional support and leads to progressive functional impairment (Fearon, 2011).” This definition brings up two very important characteristics of cachexia that are crucial to understand. First, cachexia cannot be fully reversed by conventional nutritional support. Unlike with starvation induced weight loss, simply adding calories into the diet does not reverse cachexia. Secondly, cachexia leads to progressive functional impairment. The longer that someone is in a cachectic state, the more functionally impaired they become, eventually losing all strength which is often the cause of death. Knowing this helps us to understand why practitioners need to actively manage this condition and why it is such an important area of research. We know that cachexia occurs along a spectrum with three stages of severity, and that there are specific clinical aspects of each stage. The three stages are precachexia, cachexia, and refractory cachexia. The main clinical characteristics of cachexia are weight loss, anorexia or loss of appetite, reduced food intake, complex metabolic changes, systemic inflammation, and reduced functional capacity. There is a stream of negative side effects, each one causing another, with each further step downward compounding the entire cycle. This is an extremely viscous catabolic cycle that is repeated over and over eventually leading to death in the majority of cancer patients. The metabolic processes seen in cachexia are quite complex, as the cellular mechanisms are not typically seen in normal healthy cells, or in cells of those experiencing starvation induced weight loss. Barber reports, “The fundamental difference between the weight loss observed in cachexia and that seen in, e.g., starvation, is the lack of reversibility with feeding (Barber, 2001).” Simply adding calories to the diet does not reverse weight loss seen in cachexia. These
  • 4. 4 metabolic alterations are thought to be induced by a combination of both tumor and host changes. Barber continues and states, “Candidate mediators include cytokines, neuroendocrine hormones, and tumor specific products. Several pro-inflammatory cytokines, including tumor necrosis factor (TNF), interleukin-1 (IL-1), interleukin-6 (IL-6), and interferon-y, have been implicated in cachexia (Barber, 2001).” Chronic inflammation is one of the major clinical manifestations of cancer, resulting in an elevation of pro-inflammatory cytokines, which mediate cachexia. Hypermetabolism is another complication of cachexia that leads to muscle and fat loss. “Approximately 50% of patients are hypermetabolic (REE > 110% of predicted) … hypermetabolism has been related to an elevated adrenergic state or systemic inflammation (Fearon, 2012).” Cachectic patients are in a hypermetabolic state, burning more calories than they would under normal conditions, yet have very limited appetites, thus they are also consuming fewer calories than they would under normal conditions. The hypermetabolic state is thought to be caused, in part, by futile cycles. Futile cycles are when opposing metabolic cycles (glycolysis and gluconeogenesis) occur simultaneously, resulting in the use of ATP and production of heat. Tumor cells are known to have an increased uptake of glucose, even in the presence of adequate oxygen levels. This phenomenon, known as the Warburg effect, causes excess production of lactate and upregulates the Cori cycle. Fearon continues, “Increased Cori cycle activity has been documented in weight-losing cancer patients… Overall glucose flux has been shown to be increased in weight-losing cancer patients, and such flux has been estimated to contribute up to 40% of the increase in energy expenditure in metastatic cancer (Fearon, 2012).” Here Fearon describes one of the main causes of hypermetabolism seen in cachexia. The second
  • 5. 5 known cause of increased metabolism is a heightened expression of mitochondrial uncoupling proteins (UCPs). This causes, as Fearon explains, “Dissipation of the proton gradient so that oxygen consumption is no longer coupled to ATP synthesis and heat is generated (Fearon, 2012).” This effect is commonly seen in the brown adipose tissue (BAT) of cachectic cancer patients, causing a thermogenic effect. The combination of an increased glucose flux and an increased expression of UCPs in adipose tissue creates a rise in heat production. These are the two main causes of hypermetabolism seen in cancer-induced cachexia. Lastly, cachexia is characterized by alterations in gene expression and protein levels. As previously stated, there is an overexpression of mitochondrial uncoupling proteins. This effect increases heat production, ATP loss, and creates a hypermetabolic effect. Another gene that is altered is the GLUT-4 receptor protein gene. GLUT-4 expression is downregulated in cachexia and causes insulin insensitivity. Reductions in insulin sensitivity create a pro-catabolic state, leading to increased use of the Cori cycle and loss of muscle mass. Additionally, both muscle and adipose tissue loss are due to expression of specific proteins seen in cachexia. Tisdale describes, “Loss of adipose tissue is due to increased lipolysis by tumor or host products. Loss of skeletal muscle in cachexia results from a depression in protein synthesis combined with an increase in protein degradation. The increase in protein degradation may include increased activity of the ubiquitin-proteasome pathway (Tisdale, 2009).” Tisdale explains that loss of adipose tissue is due to lipolysis brought about by certain factors. One such proposed factor is zinc alpha2-glycoprotein (ZAG) which was discovered to be a lipid mobilizing factor. This protein has been shown to cause adipose tissue loss in many animal and human cell studies. Tisdale concludes, “An increased ZAG expression may be responsible for the increased lipolytic response of adipose tissue in cancer cachexia (Tisdale, 2009).” Mimicking
  • 6. 6 this fat wasting protein is a muscle wasting glycoprotein called Proteolysis-Inducing Factor (PIF). PIF has been found to induce protein degradation and prevent protein synthesis in numerous animal, cell, and human models of cancer cachexia. Furthermore, the ubiquitin- proteasome pathway is highly active in cachexia, causing the majority of muscle loss. Jatoi explains that, “The ubiquitin-proteasome pathway is responsible for >80% of lean tissue wasting from cancer (Jatoi, 2005).” A direct correlation exists between the degree of expression of the ubiquitin-proteasome pathway and how advanced a tumor is. In summary, although not an extensive list, there are many proteins that are known to be over expressed in cancer-induced cachexia. Now that we have a better understanding of the mediators and mechanisms involved in cachexia we can look at how omega-3 PUFAs act on these pathways to reduce cachexia. There have been a number of quality papers published detailing the beneficial effects of omega-3 PUFAs on cachexia. In 2005 Jatoi reported that, “The purported mechanisms of n-3 PUFA in preventing lean tissue wasting are through the suppression of the ubiquitin-proteasome pathway, inflammatory cytokines, and the cancer cachectic factor (Jatoi, 2005).” As previously discussed, the ubiquitin-proteasome pathway accounts for a vast majority of muscle loss and the inflammatory cytokines lead to hypermetabolism. In 2008 Rondanelli reported, “Production of pro-inflammatory cytokines such as IL-6, IL-1, and TNF can be downregulated by EPA. Furthermore, the effects of PIF, are also inhibited by EPA (Rondanelli, 2008).” Rondanelli discusses EPA’s effect on cytokines, which are chronically elevated in cachexia. Adding to this, Fetterman elaborates, “Supplementation with n-3 PUFA also appears able to reduce production of proinflammatory cytokines, such as IL-1, IL-6, IL-8, and TNF-a…Excess activity of these substances contributes to pathological inflammation
  • 7. 7 (Fetterman, 2009).” Reduction in pro-inflammatory cytokines is one of the main reported mechanisms and a major benefit of n-3 supplementation for cachexia patients. Furthermore, a recent report summarized the antineoplastic activity of n-3 PUFAs. “The three main antineoplastic activities of n-3 PUFAs that have been proposed are (i) modulation of COX activity; (ii) alteration of membrane dynamics and cell surface receptor function and (iii) increased cellular oxidative stress (Cockbain, 2011).” This evidence suggests n-3 PUFAs can beneficially effect eicosanoid synthesis through COX enzyme modification, can be incorporated into the lipid bilayer and alter the structure and function of the cell surface and its downstream signaling pathways, and can also beneficially alter the intracellular redox status of target tissues. Adding to these beneficial effects, Hardman states, “n-3 fatty acids may be detrimental to the growth of metastatic or residual cancer cells by altering eicosanoid metabolism, slowing cancer cell mitosis, increasing cancer cell death, inducing differentiation, suppressing angiogenesis, and altering estrogen metabolism (Hardman, 2002).” Hardman shows us two more benefits of n-3 PUFAs that have yet to be discussed, angiogenesis and estrogen. By preventing vascular supply to the tumor, fish oil deprives the tumor of nutrients needed for growth. By decreasing estrogen production, n-3 PUFAs prevent the activation of estrogen receptors on breast, colon, and prostate cancer cells, which all have estrogen receptors needed for growth. More recently, Murphy reported many more benefits of n-3 PUFAs, stating, “EPA may support the anabolic potential of muscle through sensitizing skeletal muscle to insulin. EPA has been shown to improve glucose uptake and increase GLUT-4 expression in skeletal muscle (Murphy, 2011).” This is a highly desirable effect, noting that muscle protein synthesis must occur in an anabolic state created by the hormone insulin. Murphy continues, “EPA and DHA reduced the side effects from chemotherapy, and limited weight loss and anorexia. EPA and
  • 8. 8 DHA have also been reported to enhance tumor response to chemotherapy (Murphy, 2011).” These reports that EPA and DHA can increase the positive effects of chemotherapy while also reducing the side effects just add to the list of reasons someone would consider taking fish oil to treat cancer-induced cachexia. Now that we have reviewed the main pathways through which omega-3 fatty acids inhibit cancer-induced cachexia, we can take a deeper look into the literature supporting both sides of the argument. In order to fully understand this complex issue a full literature review is necessary. The following section will be a review of the major studies that support the use of fish oil for cancer patients with cachexia. I will then review the major studies that discourage using fish oil to treat cachexia. Lastly, I will summarize the major findings of these studies, discuss the strengths and weaknesses of each study, and provide a recommendation for their use. Review of Supporting Articles The first point of view on this issue that will be discussed is that Omega-3’s are effective for the treatment of cancer induced cachexia. Beginning with the earliest studies on this issue and progressing chronologically, the main studies on this subject will be discussed in detail. Included in the review will be the purpose of each study, the methodology, the results of the study, and the researcher’s conclusions. The opposing point of view on this issue will be discussed subsequently, with each study supporting this view point presented chronologically and discussed in a similar fashion. Omega-3 PUFAs began to be investigated for their potential ameliorating effects on cachexia as early as 1975. The study of fish oil began when epidemiological studies, often studying Greenland Inuits or Native Alaskans, first showed that cultures eating higher amounts
  • 9. 9 of fish had lower incidence of chronic diseases such as heart disease, diabetes, and cancer. One study reported, “Alaskan and Greenland Eskimos have lower rates of colon cancer and a higher consumption of n-3 fatty acids than other North Americans (Chang, 1998).” This study, among others, showed that there was a beneficial effect of consuming higher amounts of n-3 PUFAs, mainly from fish, and kick started the investigation into fish oil. Since then, thousands of studies have been done, investigating fish oil’s effects on hundreds of clinical outcomes, including cancer-induced cachexia. Many studies have been conducted to determine the appropriate dosage of fish oil and to test for adverse side effects and toxicity. In 1999 a phase 1 clinical trial was conducted to determine the maximal tolerated dose of fish oil. Patients were given .05g/kg/day fish oil supplements that were to be increased by .05g/kg/day every 2 weeks unless a complication arose. The authors found that, “At doses of 0.35 g/kg/day and above, at least one-third of patients suffered a dose-limiting toxicity. Therfore, the maximum tolerable dose was considered to be 0.3 g/kg/day (Burns, 1999).” Important to note is that the “toxicity” discussed in this study were all very minimal side effects. Burns explains, “These included excessive belching, fish taste in mouth, fish taste of food, fish smell on their own body, flatulence, and diarrhea… In summary we found that patients with advanced cancer can tolerate a large dose of fish oil with only minor side effects (Burns, 1999).” This study used much larger doses of fish oil than most other studies and found only minor side effects. One of the earliest studies that will be discussed was published in 1990 in the Journal of Cancer Research. The purpose of this study by Tisdale and Dhesi was to determine if omega-3 fatty acids could inhibit weight loss in an experimental cachexia model (Tisdale, 1990). In order to test this, they transplanted MAC16 tumor fragments into the flanks of NMRI mice to induce
  • 10. 10 experimental colon adenocarcinoma. After tumor implantation was confirmed, the mice were randomized into one of two dietary treatments. “The standard diet was comprised of mouse breeding nuts which contained 50% carbohydrate, 20% protein, 4.1% crude oil and supplied 11.5% of the energy as fat (Tisdale, 1990).” All other treatment arms were isocaloric but decreased the carbohydrate amount and provided an increasing amount of calories from fish oil, from 5% up to 50% of total energy being derived from fish oil. The mice were weighed daily and remained on the diet for 24 days. The authors found that, “Weight loss was prevented in proportion to the concentration of fish oil in the diet with almost complete protection occurring when the fish oil comprised 50% of the diet (Tisdale, 1990).” Here we can see that the fish oil supplemented groups did show a reversal of weight loss. The authors also reported that the weight loss inhibition was seen in a dose dependent manner, with the groups receiving the largest amounts of calories from fish oil losing the least amount of weight. Secondly, the authors reported, “Both the tumor weight gain per day and the tumor volume increase were reduced in animals fed the fish oil diets (Tisdale, 1990).” Interestingly, the fish oil not only prevented weight loss, but it also prevented the tumor from growing as quickly as it did in the control group. This is important to note, because many studies on fish oil report there being a beneficial effect on both the host and the tumor cells. The authors conclude by stating, “Diets enriched with fish oil have been shown to reduce both the growth rate and the extent of weight loss produced by the MAC16 adenocarcinoma…Thus an EPA-containing oil has been shown to exert both antitumor and anticachectic activity with no toxicity.” This study was an important early study that shed a hopeful light on the cachexia dilemma and opened the gateway for more research into the effects of fish oil.
  • 11. 11 Five years later, Tisdale was involved in another study with six other researchers that investigated how polyunsaturated fatty acids affected cachexia progression in a group of patients with pancreatic cancer (Wigmore, 1996). This study looked at pancreatic cancer patients because cachexia is highly prevalent in this type of cancer. The authors were specifically looking at the effect of fish oil supplementation on weight loss and the acute-phase response in these patients. It has been noted that acute-phase protein elevation, “Is associated with both increased resting energy expenditure and shortened duration of survival. This elevation is thought to be the result of proinflammatory cytokine release. (Wigmore, 1996).” The authors measured C-reactive protein (CRP) levels at baseline and 3 months after intervention to examine the acute-phase response. A total of eighteen patients with pancreatic adenocarcinoma were included in this study. The patients were given fish oil capsules of 1g that contained 18% EPA and 12% DHA. Each week the dose was increased by 2g/day, starting with 2g and rising to 16g/day. Weight, CRP levels, and a nutritional assessment were taken at baseline, 1 month after treatment, and 3 months after treatment initiation. Results of this study showed that, “All patients were losing weight before supplementation, whereas on receiving fish oil there was a median weight gain of 0.3kg/month (Wigmore, 1996).” There was a significant decrease in weight loss in the majority of the patients after 3 months of supplementation with fish oil. In addition, “After 1 month of oral fish oil, patients showed an attenuation of the acute phase response by a reduced C-reactive protein concentration (Wigmore, 1996).” The change in weight was mirrored by changes in CRP levels. The authors conclude by stating, “Before supplementation, all of the study group experienced progressive weigh loss; however, following administration of fish oil three quarters of the group either become weight stable or actually gained a small amount of weight (Wigmore,
  • 12. 12 1996).” This study showed that polyunsaturated fatty acids prevented weight loss in pancreatic cancer patients with cachexia. The next study in the review was a prospective randomized control trial that was published in 1998 by Gogos et al. The aim of this study was to investigate the omega-3 PUFAs and vitamin E effects on survival rates and the immune system in both malnourished and well- nourished patients with malignancy (Gogos, 1998). The 60 patients were randomized into two groups. The first group was supplemented with fish oil, and the second group received a placebo. There were 15 malnourished and 15 well-nourished patients in each group The groups were followed until the time of death. Before treatment and on day 40 measurements of T cells, T helper cells, T-suppressor cells, natural killer cells, and synthesis of interleukin-1 (IL-1), interleukin-6(IL-6), and tumor necrosis factor (TNF) were taken in blood mononuclear cells (Gogos, 1998). The results of this study showed that omega-3 polyunsaturated fatty acids produced an immunomodulating effect and increased the ratio of T-helper cells to T-suppressor cells in the malnourished group. However, there were no significant changes in cytokine production between the groups. In terms of survival, the well-nourished group supplemented with fish oil had the greatest survival whereas the malnourished group receiving the placebo had the least survival. Additionally, both the well-nourished and the malnourished groups receiving fish oil showed a significant increase in survival compared to the placebo groups. The authors conclude by saying that omega-3 PUFAs could offer support, especially to malnourished patients, due to their unique anticachectic, antitumor, and immunomodulating effects (Gogos, 1998). The next study supporting the use of omega-3 PUFAs to treat cancer cachexia was published in 1999 in The Journal of Nutrition. This study by Barber investigated the use of a
  • 13. 13 supplement enriched with fish oil on acute-phase protein concentrations in patients presenting with pancreatic cancer (Barber, 1999). For three weeks, 18 patients were given a nutritional supplement (2g EPA/day and 1g DA/day), and an additional 18 patients who did not receive the supplement were used as a control. The APP concentrations were measured at baseline and again after three weeks of intervention. The APP measurements taken at the three-week mark revealed that only transferrin was increased in the fish oil supplemented group. However, in the control group, there were significant decreases in albumin, pre-albumin, and transferrin, as well as a significant increase in CRP levels, suggesting an attenuation of the APP response by fish oil. In summary, the authors conclude, “Fish oil, in combination with a nutritional supplement, is able to prevent progression of the APP response and cachexia in weight losing patients with advanced cancer (Barber, 1999).” The same year as the previous study, Barber published another study looking at the effects of a fish oil enriched nutritional supplement in those with advanced pancreatic cancer. The purpose of this study was to determine if weight gain can be produced in pancreatic cancer patients through the use of a conventional nutrition supplement in combination with EPA (Barber, 1999). Barber administered nutritional supplements enriched with fish oil to 20 patients with pancreatic adenocarcinoma. There were asked to consume two cans a day, each can containing 1.09g EPA, 16.1g of protein, and 310 calories, in addition to their normal daily food intake. Assessments were taken at 3 and 7 weeks for weight, body composition, dietary intake, resting energy expenditure, and performance status (Barber, 1999). The results showed improvements in every area that was being studied. The authors reported, “Patients had significant weight gain at both 3 and 7 weeks (median 2 kg), dietary
  • 14. 14 intake increased significantly by almost 400 kcal/day, resting energy expenditure fell significantly, and performance status and appetite were significantly improved (Barber, 1999).” This study was able to show that fish oil not only stopped weight loss, but also lead to improvements in appetite, increased functional performance, as well as a reduction in resting energy expenditure, going above and beyond the results of previous studies. The very promising results from this study lead the authors to conclude that cachexia can be countered through the use of a nutritional supplement enriched with EPA. Following this encouraging study, Barber decided to look into some of the metabolic components of cachexia to see how fish oil affected these pathways. In 2001, he published another study using the same fish oil enriched nutritional supplement, this time looking at how the supplement effected metabolic mediators that are known to have a role in cachexia (Barber, 2001). Barber provided a supplement that contained 2g of EPA and 600 kcal to 20 patients with pancreatic cancer who were to consume it daily. Barber reported that, “At baseline and at 3 weeks, patients were weighed and serum concentration samples were taken to measure IL-6, cortisol, insulin, leptin, blood mononuclear cell production of IL-1B, IL-6, and TNF-a, and urinary excretion of proteolysis inducing factor (PIF) (Barber, 2001).” Interestingly, Barber found that after 3 weeks of fish oil supplementation, levels of IL-6 were significantly decrease, the serum insulin concentration was elevated, the cortisol-to-insulin ratio decreased, and there was also a reduction in the number of patients excreting PIF in the urine. Notably, there was also weight gain in addition to the beneficial changes in these metabolic mediators. In conclusion, Barber stated that the fish oil-enriched supplement modulates different mediators involved in cachexia catabolism. Normalization of cancer associated metabolic changes, that normally prevent weight gain, are suggested to be brought
  • 15. 15 about by the fish oil in the supplement (Barber, 2001). Here we begin to see some of the mechanisms by which fish oil exerts its positive anti-cachexia effects, specifically through the modulation of key metabolic pathways. The last three studies by Barber build upon each other and present a strong argument for the use of omega-3 fatty acids in preventing cachexia in cancer patients. Further strengthening the argument for the use of fish oil to treat cachexia is a study that was published in 2001 by Hardman. The purpose of this study was to determine if concentrated fish oil will be a useful adjuvant treatment for chemo, as well as to determine if the concentrated fish oil would cause detrimental increases in the drug toxicity to either the host or tumor cell (Hardman, 2001). The methods of this study used MDA-MB 231 breast cancer cell xenografts and implanted them into 120 athymic mice. The mice were fed a normal diet for three weeks to allow the tumor to develop. At the three-week mark, the mice were randomly divided into two dietary groups. The first group receiving the standard diet of 5% corn oil, and the second group receiving a diet containing 3% fish oil concentrate and 2% corn oil. The mice were kept on their respective diets for two weeks at which point mice from each group were randomly selected to receive doxorubicin (DOX) therapy for a total of five weeks. Tumor size was measured weekly. The data from the results of this experiment showed that the 3% FOC group increased the efficacy of doxorubicin against tumor growth without causing an increase in toxicity to the host cells. Additionally, “Consumption of the 3% FOC alone was as effective against the growth of the MDA-MB 231 breast carcinoma as was DOX chemotherapy with the 5% corn oil diet (Hardman, 2001).” These results are intriguing, showing that a fish oil concentrate increased the effectiveness of doxorubicin chemotherapy without causing toxicity to the host. Even more
  • 16. 16 interesting, this study showed that fish oil concentrate alone was just as effective as chemotherapy in preventing tumor growth in breast cancer xenografts. The next study to be reviewed looked at the use of fish oil to treat colorectal cancer. In 2003, Llor published a study in the Journal of Clinical Nutrition that looked at how fish oil affects colorectal metabolic processes in comparison to olive oil, linoleic acid, and oleic acid (Llor, 2003). The purpose of this study was to determine the function of these fats in the process of colorectal carcinogenesis. The authors used both Caco-2 and HT-29 colon adenocarcinoma cell lines. These cells were then supplemented with fish oil (50% EPA and DHA), olive oil, oleic acid, or linoleic acid. At 48 and 72 hours after the addition of the fats, the cells were tested for apoptosis, proliferation, differentiation, and Cox-2 and Bcl-2 expression was assessed. The results showed that both fish oil and olive oil increased the induction of apoptosis in both colon cancer cell lines. Fish oil supplementation drastically decreased cell proliferation in the Caco-2 and HT-29 cell lines. All four of the fats increased cellular differentiation of the Caco-2 cells. Lastly, both COX-2 and Bcl-2 expression was downregulated in both the fish oil and olive oil supplemented cells. The authors conclude by stating, “Fish oil and olive oil alter different cellular processes leading to prevention of colorectal cancer development. COX-2 and Bcl-2 could be partially responsible for some of the observed effects (Llor, 2003).” Notably, fish oil not only prevented further cell proliferation of these cancer cells, but it also increased cellular differentiation and apoptosis. These results help to explain some of the metabolic pathways that are mediated by fish oil and shows us that fish oil effects gene expression of specific pro- carcinogenic genes such as COX-2 and Bcl-2. The final and most recent study supporting the use of omega-3 PUFAs to treat cancer cachexia is a study by Murphy et al (Murphy, 2011). The purpose of Murphy’s study was to
  • 17. 17 assess the effect of a fish oil intervention on body composition and weight compared to standard of care during chemotherapy. There were forty patients in this study that were divided into two groups. Sixteen patients were placed in the fish oil (2.2g of EPA/day) group and the remaining 24 were placed into the standard of care group. Body composition was measured using computed tomography imaging. Blood and weight measurements were taken at baseline and throughout chemotherapy. The authors found that those in the standard of care group had an average weight loss of 2.3 kg, however those in the fish oil group maintained their weight. Secondly, the authors reported that those who had the highest plasma EPA levels showed the greatest muscle gains. There was a maintenance or gain in muscle mass in 69% of patients in the fish oil group. In comparison, there was a maintenance of muscle mass in only 29% of patients in the standard of care group (Murphy, 2011). Here we see that fish oil was superior to standard of care treatment for maintaining body composition. Finally, the authors conclude that, “Nutritional intervention with 2.2g of fish oil per day appears to provide a benefit over standard of care, resulting in maintenance of weight and muscle mass during chemotherapy (Murphy, 2011).” This study shows us that fish oil helps to maintain muscle mass in cancer patients, even during chemotherapy treatment, very promising results for individuals with cancer cachexia. Adding support to the argument for omega 3 PUFAs to treat cachexia is a double-blind, placebo-controlled study looking that looked at advanced lung cancer patients (Finocchiaro, 2012). This study looked into how EPA and DHA would effect inflammation and oxidative stress in lung cancer patients. They randomly divided 33 patients with non-small-cell lung cancer into two groups. The first group received 850 mg of EPA/DHA and the second group received a
  • 18. 18 placebo. The subjects were followed for 66 days and biochemical and anthropometric measurements were taken at day 8, 22, and 66. The results showed a significant reduction in inflammatory cytokines CRP, TNF-a, and IL-6 in the n-3 group. Additionally, there was a significant reduction in plasma ROS compared to the placebo group. Lastly, the study showed a slight increase in protein and energy intake in the n-3 group and a significant increase in body weight in the treatment group. These positive results support the use of n-3 PUFAs in cachexia treatment, however, the authors concluded that there is still not enough clinical evidence to justify using n-3 PUFAs in a clinical setting. Following this study was a study out of Taiwan that looked at omega-3 fatty acids, in combination with micronutrients and probiotics, for their effects on weight gain in head and neck cancer patients (Yeh, 2013). The authors examined the effectiveness of a nutritional supplement for modifying body weight, albumin and prealbumin levels, and survival. After being randomly assigned to receive either the nutritional supplement or an isocaloric control supplement, 68 subjects were followed for 3 months. Changes in body weight and albumin levels were monitored. The results indicated that the group taking the nutritional supplement enriched with n-3 fatty acids maintained their body weight throughout the treatment period whereas the control group did not. Additionally, the treatment group had significantly improved albumin and prealbumin levels compared to the control group. The ability of patients to maintain body weight seen in this study supports an anti-cachectic effect of n-3 fatty acids. Moreover, a rise in protein levels suggest another benefit of n-3 fatty acids and endorses their use in cachexia patients. Lastly, a randomized control trial was published in 2014 that compared the use of an EPA enriched nutritional supplement versus an isocaloric control to treat clinical and biochemical
  • 19. 19 outcomes in patients with advanced non-small-cell lung cancer (Sanchez-Lara, 2014). This study looked at the effects on body weight, inflammation, energy intake, quality of life, chemotherapy response, and survival. The 96 patients were randomized and followed for 8 weeks, with nutritional, clinical, and biochemical measurements taken after each round of chemo. The omega-3 fatty acid supplemented group was able to maintain their weight and even significantly increased their lean body mass, whereas the control group showed the opposite. Inflammation was also improved in the treatment group, which displayed a significant reduction in CRP and TNF-a levels, while the control group showed no changes. Energy and protein intake was reduced in the control group but significantly increased in the treatment group. Additionally, the treatment group showed reduced appetite loss and fatigue and an increased quality of life compared to the control group. There were no significant differences in responses to chemotherapy between groups. In summary, the previous nine studies have created a very strong argument for the effectiveness of omega-3 fatty acids found in fish oil to prevent cancer cachexia. We have seen that fish oil has been effective in reversing cachexia in many different types of cancer including pancreatic, breast, colon, and lung cancers. These studies have shown fish oil to inhibit weight loss in a cachexia model, to delay the progression of cachexia in pancreatic cancer, and to restore immunodeficiency and prolong survival of malnourished patients. Additionally, fish oil enriched nutritional supplements caused pancreatic cancer patients to gain weight. Fish oil has also shown to positively affect metabolic mediators of cachexia such as cytokine production, in addition to being able to modulate gene expression of key mediators of cachexia such as COX-2 and Bcl-2. Furthermore, these studies have shown that omega-3 PUFAs from fish oil increase the effectiveness of chemotherapy such as doxorubicin without causing toxicity to the host cell, in
  • 20. 20 addition to fish oil providing a benefit over standard of care for preserving muscle mass and improving body composition in patients with advanced cachexia. Most recently, randomized control trials have showed us that omega-3 fatty acids are effective at reducing inflammation and ROS levels, increasing total energy intake, and maintaining body weight, with the most recent study showing an increase in lean body mass and an improvement in quality of life. When all of these studies are taken into account we cannot deny that there is very strong evidence supporting the use of omega-3 PUFAs from fish oil for the treatment of cancer cachexia. However, before any conclusion can be made, the opposing side of this argument must be presented and all relevant literature will be reviewed and discussed. Review of Refuting Articles The earliest study that will be discussed was published in 1992 by Calder and Newsholme (Calder, 1992). The aim of this study was to study the effects of fatty acids on proliferation of blood lymphocytes and on IL-2 concentration. The authors wanted to look at the effects on the immune system because numerous studies have shown fatty acids to be able to modulate the immune system. Blood was drawn from five female subjects and transferred to Leucoprep tubes for centrifugation. PUFAs were added to the tubes and after 48 hours the blood was assayed for lymphocyte proliferation and IL-2 levels. The results from the assays showed that EPA caused a significant inhibition of blood lymphocyte proliferation. In addition, EPA also significantly decreased the concentration of IL-2 in blood lymphocytes. These findings are important to discuss because they cause a reduction in the immune system after the addition of PUFAs. Lymphocyte proliferation has been shown to be dependent on its ability to secrete IL-2. In this study PUFAs were shown to prevent the secretion
  • 21. 21 of IL-2, therefore halting lymphocyte proliferation and weakening the immune system. This raises serious concerns for cancer patients who are already severely immunocompromised. The following year another study was published that also looked at omega-3s effect on the immune system, specifically IL-2 and mononuclear cells (Endres, 1993). This study by Endres reinforced the previous study, finding similar results. Eighteen male volunteers were given 18g (Each gram containing 153mg EPA and 103 mg DHA) of lipid concentrate per day and continued this for six weeks. Blood was drawn at baseline and at the six-week mark and was assessed for mononuclear cell proliferation and IL-2 production. IL-2 production was remarkably reduced by 65% compared to baseline levels. Furthermore, mononuclear cell proliferation mirrored the IL-2 response and showed a 70% decrease compared to baseline. While these results may be useful in diseases that show an overexpression of the immune system such as rheumatoid arthritis, these effects would be detrimental to cancer patients who are already severely immunocompromised. The reduction in immune function seen in these two early studies highlights one of the negative side effects of fish oil supplementation. The next study that will be reviewed showed that EPA had no effect on preventing cachexia (Costelli, 1995). This study assessed if omega 3 polyunsaturated fatty acids, especially EPA, effectively prevents hypercatabolism in tissues or changes tumor growth rates. Male wistar rats were injected with Yoshida AH-130, a rapid growing hepatoma that causes weight loss and tissue wasting. They were randomly divided into two groups, the first receiving daily EPA and the second receiving a control diet. At 0, 4, and 7 days after injection of the hepatoma, the mice were killed and tumor volume was assessed.
  • 22. 22 The hepatoma created a rapid loss of body weight and muscle in the rates. The rats in the group receiving the EPA showed no alteration to the tumor growth rate compared to the control group. Furthermore, the EPA was also ineffective at reducing the rate of wasting as compared to the control group. The authors conclusion states, “The use of n-3 PUFA to prevent cancer associated cachexia may be limited by the type of tumor. Very undifferentiated tumors are probably insensitive to growth modulation by such agents (Costelli, 1995).” This study shows no effect of EPA in reducing cachexia, at least in this tumor type, and weakens the argument for using n-3 PUFAs to treat cachexia. Moving ahead three years, we go to a study looking into the effects of PUFAs on colon cancer in rats (Griffini, 1998). This study is one of the strongest advocates against the use of omega 3 PUFAs in cancer patients. The purpose of this study was to investigate the effects of n-3 and PUFAs on the development of colon carcinoma metastasis in rat liver (Griffini, 1998). Wag- Rij rats were divided into three dietary groups, one receiving a fish oil diet, one receiving a safflower oil diet, and the last group receiving a low fat control diet. The rats were maintained on their diets for three weeks. At the three-week mark, CC531 colon carcinoma cells were transplanted into the rats. At 1 and 3 weeks, rats were euthanized and liver metastasis was measured. The data showed that at 1 week after transplantation there were 7x more metastasis in the fish oil group compared to the low fat diet group. Additionally, at 3 weeks after transplantation there were 10x more metastasis in the fish oil group compared to the low fat diet group. Adding to these alarming results, the size of the metastasis in the fish oil group were 1000 fold larger than those found in the low fat diet group. Griffini concludes, “N-3 PUFAs promote colon cancer metastases in the rat liver. This finding has serious implications for the treatment of cancer
  • 23. 23 patients with fish oil to fight cachexia (Griffini, 1998).” This study is very alarming, showing that n-3 PUFAs not only promoted extreme increases in number of metastases but also in the size of the metastases. In 2000 the Journal of Clinical Nutrition published a study out of the Netherlands that investigated the effect of EPA on rates of lipolysis and lipid oxidation in cachectic patients and healthy individuals (Zuijdgeest-Van Leeuwen, 2000). This was being investigated because it was proposed that weight loss in cancer patients can be prevented through lipolysis inhibition. There were 17 cancer patients and 16 healthy subjects, all randomized to receive either a EPA (6g/day) or a placebo. The subjects were followed for seven days, with whole body lipolysis, palmitate oxidation, and palmitic acid release being measured at baseline, day 2, and day 7 of the intervention. The authors reported no significant differences in lipolysis between groups. Secondly, palmitate oxidation and palmitic acid release did not differ between the two groups. In conclusion, the authors state, “Supplementation of EPA does not significantly inhibit lipolysis or lipid oxidation in weight-losing cancer patients or in healthy subjects (Zuijdgeest-Van Leeuwen, 2000).” By showing fish oil is ineffective at reducing wasting of adipose tissue and preventing lipolysis in cachectic patients, this study refutes the use of omega-3 PUFAs in treating cachexia. The ensuing study in this review is just as bleak as the previous by Van Leeuwen. In 2003, Gut published a randomized double blind trial by Fearon et al. that assessed the effect of a n-3 enriched supplement on weight loss (Fearon, 2003). This study compared a protein and energy dense supplement enriched with n-3 fatty acids with an isocaloric control supplement. They tested for effects on weight, lean body mass, dietary intake, and quality of life in cachectic
  • 24. 24 patients (Fearon, 2003). There were 200 patients in this trial, randomized to either receive the n-3 fatty acid or the control supplement and followed for eight weeks. At the end of the 8-week intervention period there was no significant difference in the two groups in terms of weight loss and lean body mass. Reduction in the rate of weight loss was the same in both groups. Changes in lean body mass were also similar in both groups. The authors concluding statement said, “Enrichment with n-3 fatty acids did not provide a therapeutic advantage and both supplements were equally effective in arresting weight loss (Fearon, 2003).” There was not a clear advantage and no difference in outcomes between the experimental and control arms. We can see from this study that the use of omega-3 fatty acids to prevent cachexia would be ill advised as it provides no additional benefit when compared to traditional treatments. Jatoi lead a similar study in 2004 that investigated whether an EPA supplement would be more effective than megestrol acetate at augmenting appetite and weight gain in patients with cancer associated cachexia (Jatoi, 2004). This study randomly assigned 421 cachectic patients to one of three groups. The first group received 1.09g EPA supplement plus a placebo, the second group received 600 mg/d megestrol acetate plus an isocaloric supplement, and the third group received both the EPA supplement as well as megestrol acetate. Weight measurements and physical examinations were taken at baseline and weekly. In terms of appetite improvement, there was no significant difference between the three treatment arms. However, both of the treatment arms receiving megestrol acetate showed an increase in functional assessment compared to the fish oil treatment arm. In addition, there was a smaller percentage of patients who gained greater than 10% of baseline weight in the EPA group compared to those in both megestrol acetate treatment groups. Lastly, there was no significant differences in quality of life or survival between the two groups. In conclusion, “This EPA
  • 25. 25 supplement, either alone or in combination with megestrol acetate, does not improve weight or appetite better than megestrol acetate alone (Jatoi, 2004).” EPA supplementation showed zero improvements in any of the parameters measured compared to the other treatment arms. This suggests no benefit in using omega-3 PUFAs to treat cancer cachexia. Next up in the review of studies opposing the use of fish oil to treat cancer cachexia is a randomized pilot study looking at advanced gastrointestinal cancer patients (Persson, 2005). The objective of this pilot study was to investigate the effect of fish oil supplementation in combination with melatonin and dietary advice on the biochemical variables known to develop with cachexia, specifically cytokines. In addition, the authors examined clinical outcomes such as body weight, quality of life, and food intake. The twenty-four patients were randomized to receive either fish oil (4.9g DHA and 3.2g DHA), melatonin, or a combination. At baseline and at 4 weeks plasma was analyzed for TNF-a, IL-6, IL-1B, IL-8, DHA, EPA, linoleic acid, and arachidonic acid (Persson, 2005). The results showed that there was no significant difference in any of the cytokines and no difference in biochemical variables between the fish oil and melatonin groups. Moreover, there was not a significant difference in energy intake, functional performance status, or survival between the two groups. The authors conclude, “Fish oil, melatonin, and their combination, did not produce substantial anti-inflammatory effects in cachectic patients with advanced gastrointestinal cancer (Persson, 2005).” No benefits in inflammatory cytokine concentrations were seen when fish oil was supplemented, contrary to what has been shown in previous fish oil studies. These results oppose the use of fish oil for treating cancer cachexia, due to an absence of an anti-inflammatory effect that they are commonly associated with.
  • 26. 26 The last study that will be covered in this review was published in 2006 in the Journal of Clinical Oncology (Fearon, 2006). This article was double-blind, randomized, and placebo- controlled. The purpose of this study was to compare EPA diester against a placebo for their effects on lean body mass and weight in an eight-week intervention. This was a large study, with 518 patients being randomly assigned to one of three treatment arms to be followed for eight weeks. The first group received 2g of EPA daily (n=175), the second group received 4g EPA daily (n=172), and the final group received a placebo (n=171). Assessments were taken at 4 and 8 weeks. The results indicated no significant changes in weight, survival, or nutritional variables. Fearon concludes, “Overall, no statistically significant effects of treatment on the primary endpoint of weight were observed…There were no other obvious differences in nutrition or quality of life measures as a result of EPA diester administration...The results indicate no significant benefit from single agent EPA in the treatment of cancer cachexia (Fearon, 2006).” The current study, reporting no changes in body weight or quality of life by the addition of EPA, illustrates that EPA is not useful for preventing cancer cachexia and provides a strong argument against its use in this population. Taking all of these studies into account we can see that there is also a strong argument against the use of omega-3 PUFAs to treat cancer cachexia. Opposing the views of the previous set of studies, these articles demonstrate that fish oil can potentially be quite harmful and advocate that it should not be used to treat cachexia. The data from these original articles have shown that fish oil can reduce the immune system by suppressing IL-2 production and lymphocyte proliferation, has increased the number and size of colon metastases in an animal model, is ineffective at preventing lipolysis, and is simply ineffective at modifying many clinical
  • 27. 27 outcomes such as quality of life, performance status, and survival. It is very important to consider the results of these studies when assessing the use omega-3 fatty acids to treat patients with cancer-induced cachexia. Discussion This area of study has become a major controversy, especially in clinical practice. While there are numerous studies showing that fish oil has positive effects on clinical outcomes of cachexia, there are also just as many studies showing the opposite. For every study that came out supporting the use of fish oil to treat cachexia, another article followed refuting its use. Because of this dichotomy in the literature, oncologists have become torn on this issue, some saying that yes, we should supplement fish oil in cachectic patients, whilst other clinicians are very opposed, saying that it has no benefits and will only add to the financial burden of the patient. In addition to the financial burden, some clinicians argue that it is already difficult to get cancer patients to eat because of their loss of appetite and adding fish oil into their treatment plan will only complicate this issue further. So what conclusions can be drawn after reviewing both sides of the literature? From reviewing both the supporting and opposing articles we can see that there is strong evidence supporting both sides of this debate. In order to decide which side is more convincing we must take a deeper look at each study and carefully examine the methodology and results and compare strengths and weaknesses of each study. We can see that omega-3 fatty acids show very positive results in the early cell and animal studies. However, when we look at the human studies we can see that the results were a lot dimmer. Because the larger human trials show negative results, I am persuaded to support the side against the use of omega-3 fatty acids. Cell and
  • 28. 28 animal studies are important for understanding the mechanisms, but phase 3 human studies are much more applicable to a clinical setting. Cell and animal studies are easy to control for confounding variables, but studying nutrition in humans can be very difficult because of our free will. I will discuss both sides and why I believe that there currently is not enough evidence to support the use of omega-3 fatty acids to treat cancer cachexia. As discussed previously, almost all cell and animal models show that omega 3 PUFAs produce a beneficial effect in a cachexia model. In cellular studies they were shown to reduce inflammatory cytokine levels such as CRP, TNF-a, IL-1, and IL-6 and to reduce PIF activation (Rondanelli, 2008). We have also seen that they can suppress the ubiquitin-proteasome pathway and suppress the cancer cachetic factor (Jatoi, 2005). However, we also know that the immune system can be compromised with the use of n-3 fatty acids. Lymphocyte proliferation was halted in one study (Calder, 2002), and mononuclear cell production was greatly reduced in another (Endres, 1993). Cancer patients typically have very weakened immune systems already so we must carefully consider these studies when deciding whether or not to supplement n-3 fatty acids. In terms of cell studies, the majority of the evidence supports the use of omega-3 fatty acids for cachexia patients, but the possible reduction in immunity must be taken into account. When we look at animal studies we see a similar pattern. The majority of evidence is in favor of the use of omega-3 fatty acids to treat cachexia. However, one study did show 10x greater number of colon metastasis in rats supplemented with fish oil compared to the control group (Griffini, 1998). Cell and animal models, while important and useful, can only tell us so much. The results of human studies have much more weight on the clinical applicability. When we look at the earlier human studies we can see that there are many flaws. Many of the studies had small number of subjects meaning the results can’t be generalized to the total
  • 29. 29 population group. The study by Wigmore only had 18 patients, the study by Gogos only had 15 subjects (Gogos, 1998), the study by Barber only had 18 subjects (Barber, 1999), and the study by Barber in 2001 only had 20 patients (Barber, 2001). Although these sample sizes are somewhat small we also have to keep in mind that there are many factors affecting participation when dealing with advanced cancer patients. It is very difficult to get very sick patients to volunteer for studies when they are undergoing chemotherapy and radiation, and this is also why there is low compliance and a high dropout rate in some of these studies. While this is something that can be very difficult to improve upon, it still remains a flaw of many of these studies supporting the use of fish oil. Furthermore, when we look at some of the larger clinical trials in humans we see that they show no effect. A study using 91 patients compared fish oil to olive oil on its ability to affect clinical outcomes in cachexia patients. In this well designed study there was absolutely no change in appetite, energy intake, or functional status between the groups (Bruera, 2003). Another large clinical trial (N=200) showed no difference in rate of weight loss between an EPA-enriched supplement group and an isocaloric supplement group (Fearon, 2003). Lastly, a large double blind study (N=421) showed that EPA had no added benefit for treating cachexia compared to megestrol acetate alone (Jatoi, 2004). Each of these studies had much larger sample sizes and better study designs than the earlier human studies. Some would argue that the dose of fish oil was too small to see an effect and a weakness in these studies, but I do not believe that this is the case. For this reason, these later trials hold more weight than the earlier trials showing positive results. Based on this information, I am convinced that the use of omega-3 fatty acids is ill advised for patients with cachexia.
  • 30. 30 The most recent randomized clinical trials began to show a trend in support of the use of omega-3 fatty acids to treat cachexia. However, in one study, the intervention involved n-3 PUFAs, probiotics, and micronutrients (Yeh, 2013). It cannot be said for certain that the positive results of this study were due to the omega-3 fatty acids. Another recent study showed that the group receiving n-3 fatty acids showed a significant decrease in inflammatory cytokine and ROS levels and an increase in energy intake (Finocchiaro, 2012). However, in their discussion the authors state that they think the differences seen between the two groups are random and not caused by the treatment. It is important to look closely at each study before drawing conclusions. Conclusion In summary, I believe that there is not enough evidence to support the use of omega-3 fatty acids for treating cancer-induced cachexia. While there are ample amounts of research showing beneficial effects of omega-3 fatty acids, specifically in cell and animal models, the larger, phase-3 clinical trials in humans show negative results and are more convincing. Furthermore, after considering flaws in study design of the smaller human trials, I am more swayed by the larger clinical trials that have better study designs and superior methodology. Based on the reviewed evidence, omega-3 fatty acids may or may not have an effect depending on numerous variables. Supplementing with fish oil, while being relatively safe and having very minimal side effects, is simply not supported by the literature at this time. The answer is not a definitive no at this point, but adding fish oil into a treatment regimen for an individual with advanced cancer can possibly create more problems than it would solve and is ill advised. While it can be highly appealing to some looking for anything to relieve the deleterious effects of cachexia, we do not know enough about it yet and further research is necessary.
  • 31. 31 Future Research While there are currently hundreds of studies on this subject and our understanding of certain aspects of it are plentiful, we must continue to develop future research in order to fully understand this phenomenon before we can come to a definitive conclusion. The scientific community has made leaps and strides in understanding how omega 3 fatty acids work over the last 50 years, however, there are still some specific questions that we have yet to answer. In addition to these questions, I believe that future studies should change the way certain parts of the study methodology is carried out. Future studies should address some of the following questions. How does the stage of cachexia progression affect the effectiveness of omega 3 supplementation? For example, what will the difference in outcome be if treatment is started in the pre-cachexia stage, vs. the cachexia stage, vs. the refractory cachexia stage? Future studies also need to examine how other clinical and nutritional variables play a role in how effective omega-3 fatty acids will be. How do comorbidities, genotype, epigenetics, environment, and other dietary factors affect the outcome of treatment? Furthermore, it would be beneficial to have a deeper understanding of the underlying mechanisms omega 3s use to exert their effects in order to create more effective treatment options. Lastly, there is simply a need for additional large scale, randomized clinical trials in humans. In terms of the methodology, all efforts in the future should be placed on making the studies as equal as possible so that comparisons can be made. In the past, study designs are rarely comparable and most studies are highly heterogeneous. Tolerability of the EPA/DHA source should be made more tolerable so that there will be less dropout. Moreover, compliance with the treatment needs to be better addressed in future studies to ensure the desired treatment effect is
  • 32. 32 taking place. Making these changes will ensure that more subjects are able to complete the trial and the results are more legitimate. References Barber, M. D. (2001). Cancer Cachexia and Its Treatment With Fish-Oil-Enriched Nutritional Supplementation Nutrition, 17, 751-755. Barber, M. D., Fearon, K.C., Tisdale, M.J., McMillan, D.C., & Ross, J.A. (2001). Effect of a Fish-Oil Enriched Nutritional Supplement on Metabolic Mediators in Patients With Pancreatic Cancer Cachexia Nutrition and Cancer, 40(2), 118-124. Barber, M. D., Ross, J.A., Preston, T., Shenkin, A., & Fearon, K.C. (1999). Fish Oil-Enriched Nutritional Supplement Attenuates Progression of the Acute-Phase Response in Weight- Losing Patients with Advanced Pancreatic Cancer The Journal of Nutrition, 129, 1120- 1125. Barber, M. D., Ross, J.A., Voss, A.C., Tisdale, M.J., & Fearon, K.C. (1999). The effect of an oral nutritional supplement enriched with fish oil on weight-loss in patients with pancreatic cancer British Journal of Cancer, 81(1), 80-86. Bruera, E., Strasser, F., Palmer, J.L., Willey, J., Calder, K., Amyotte, G., & Baracos, V. (2003). Effect of Fish Oil on Appetite and Other Symptoms in Patients With Advanced Cancer and Anorexia/Cachexia: A Double-Blind, Placebo-Controlled Study Journal of Clinical Oncology, 21(1), 129-134. Burns, C. P., Halabi, S., Clamon, G.H., Hars, V., Wagner, B.A. Hohl, R.J., ... Paskett, E. . (1999). Phase I Clinical Study of Fish Oil Fatty Acid Capsules for Patients with Cancer Cachexia: Cancer and Leukemia Group B Study 9473. Clinical Cancer Research, 5, 3942-3947. Calder, P. C., & Newsholme, E.A. (1992). Polyunsaturated fatty acids suppress human peripheral blood lymphocyte proliferation and interleukin-2 production Clinical Science, 82, 695- 700. Chang, W. L., Chapkin, R.S., & Lupton, J.R. (1998). Fish Oil Blocks Azoxymethane-Induced Rat Colon Tumorigenesis by Increasing Cell Differentiation and Apoptosis Rather Than Decreasing Cell Proliferation The Journal of Nutrition, 128, 491-497. Cockbain, A. J., Toogood, G.J., & Hull, M.A. (2011). Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer. Gut, 1-15.
  • 33. 33 Colomer, R., Moreno-Nogueira, J.M., Garcia-Luna, P.P., Garcia-Peris, P., Garcia-de-Lorenzo, A., Zarazaga, A., ... Casimiro, C. . (2007). n-3 Fatty acids, cancer and cachexia: a systematic review of the literature. British Journal of Nutrition, 97, 823-831. Costelli, P., Llovera, M., Lopez-Soriana, J., Carbo, N., Tessitore, L., Lopez-Soriano, F.J., ... Argiles, J.M. (1995). Lack of effect of eicosapentaenoic acid in preventing cancer cachexai and inhibiting tumor growth Cancer Letters, 97, 25-32. Dewey, A., Baughan, C., Dean, T.P., Higgins, B, & Johnson, I. (2012). Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia Cochrane Database of Systematic Reviews(1). Endres, S., Meydani, S.N., Ghorbani, R., Schnigler, R., & Dinarello, C.A. (1993). Dietary Supplementation with n-3 fatty acids suppresses interleukin-2 production and mononuclear cell proliferation Journal of Leukocyte Biology, 54(6), 599-603. Fearon, K., Strasser, F., Anker, S.D., Bosaeus, I., Bruera, E., Fainsinger, R.L., ... Baracos, V.E. (2011). Definition and classification of cancer cachexia: an international consensus Lancet, 12, 489-495. Fearon, K. C., Barber, M.D., Moses, A.G., Ahmedzai, S.H., Taylor, G.S., Tisdale, M.J., & Murray, G.D. (2006). Double-Blind, Placebo-Controlled, Randomized Study of Eicosapentaenoic Acid Diester in Patients With Cancer Cachexia Journal of Clinical Oncology, 24(21), 3401-3407. Fearon, K. C., Glass, D.J., Guttridge, D.C. (2012). Cancer Cachexia: Mediators, Signaling, and Metabolic Pathways Cell Metabolism, 16, 153-166. Fearon, K. C., von Meyenfeldt, M.F., Moses, A.G., van Geenen, R., Roy, A., Gouma, D.J., ... Tisdale, M.J. (2003). Effect of a protein and energy dense n-3 fatty acid enriched oral supplement on loss of weight and lean tissue in cancer cachexia: a randomised double blind trial. Gut, 52, 1479-1486. Fetterman Jr., J. W., & Zdanowicz, M.M. (2009). Therapeutic potential of n-3 polyunsaturated fatty acids in disease. American Journal of Health-System Pharmacy, 66, 1169-1179. Finocchiaro, C., Segre, O., Fadda, M., Monge, T., Scigliano, M., Schena, M., & Tinivella, M. (2012). "Effect of n-3 fatty acids on patients with advanced lung cancer: a double blind, placebo-controlled study " British Journal of Nutrition 108: 327-333. Furst, P., & Kuhn, K.S. (2000). Fish oil emulsions: what benefits can they bring? . Clinical Nutrition, 19(1), 7-14. Giacosa, A., & Rondanelli, M. . (2008). Fish oil and treatment of cancer cachexia Genes and Nutrition, 3, 25-28. Gogos, C. A., Ginopoulos, P., Salsa, B., Apostolidou, E., Zoumbos, N.C., & Kalfarentzos, F. (1998). Dietary Omega-3 Polyunsaturated Fatty Acids plus Vitamin E Restore
  • 34. 34 Immunodeficiency and Prolong Survival for Severely Ill Patients with Generalized Malignancy Cancer, 82, 395-402. Griffini, P., Fehres, O., Klieverik, L., Vogels, I.M., Tigchelaar, W., Smorenburg, S.M., & Van Noorden, C.J. (1998). Dietary n-3 Polyunsaturated Fatty Acids Promote Colon Carcinoma Metastasis in Rat Liver. Cancer Research, 58, 3312-3319. Hardman, W. E. (2002). Omega-3 Fatty Acids to Augment Cancer Therapy The Journal of Nutrition, 132, 3508-3512. Hardman, W. E., & Avula, C.P. (2001). Three Percent DIetary Fish Oil Concentrate Increased Efficacy of Doxorubicin Against MDA-MB 231 Breast Cancer Xenografts Clinical Cancer Research, 7, 2041-2049. Jatoi, A. (2005). Fish oil, lean tissue, and cancer: is there a role for eicosapentaenoic acid in treating the cancer anorexia/weight loss syndrome? . Critical Reviews in Oncology/Hematology, 55, 37-43. Jatoi, A. (2005). n-3 Fatty Acid Supplements for Cancer-Associated Weight Loss. Nutrition in Clinical Practice, 20, 394-399. Jatoi, A. R., K., Loprinzi, C.L., Sloan, J.A., Dakhil, S.R., Macdonald, N., ... Christensen, B. (2004). An Eicosapentaenoic Acid Supplement Versus Megestrol Acetate Versus Both for Patients With Cancer-Associated Wasting: A North Central Cancer Treatment Group and National Cancer Institute of Canada Collaborative Effort Journal of Clinical Oncology, 22(12), 2469-2476. Llor, X., Pons, E., Roca, A., Alvarez, M., Mane, J., Fernandez-Banares, F., & Gassull, M.A. (2003). The effects of fish oil, olive oil, oleic acid, and linoleic acid on colorectal neoplastic processes. Clinical Nutrition, 22(1), 71-79. Murphy, R. A., Mourtzakis, M., Chu, Q.S., Baracos, V.E., Reiman, T., & Mazurak, V.C. (2011). Nutritional Intervention With Fish Oil Provides a Benefit Over Standard of Care for Weight and Skeletal Muscle Mass in Patients With Nonsmall Cell Lung Cancer Receiving Chemotherapy Cancer, 117, 1775-1782. Murphy, R. A., Yeung, E., Mazurak, V.C., & Mourtzakis, M. . (2011). Influence of eicosapentaenoic acid supplementation on lean body mass in cancer cachexia British Journal of Cancer, 105, 1469-1473. Persson, C., Glimelius, B., Ronnelid, J., & Nygren, P. . (2005). Impact of fish oil and melatonin on cachexia in patients with advanced gastrointestinal cancer: A randomized pilot study. Nutrition, 21, 170-178. Roodhart, J. M., Daenen, L.G., Stigter, E.C., Prins, H., Gerrits, J., Houthuijzen, J.M., ... Voest, E.E. (2011). Mesenchymal Stem Cells Induce Resistance to Chemotherapy through the Release of Platinum-Induced Fatty Acids. Cancer Cell, 20, 370-383.
  • 35. 35 Roynette, C. E., Calder, P.C., Dupertuis, Y.M., & Pichard, C. (2004). n-3 Polyunsaturated fatty acids and colon cancer prevention Clinical Nutrition, 23, 139-151. Sanchez-Lara, K., Turcott, J.G., Juarez-Hernandez, E., Nunez-Valencia, C., Villanueva, G., Guevara, P., & Torre-Vallejo, M. (2014). "Effects of an oral nutritional supplement containing eicosapentaenoic acid on nutritional and clinical outcomes in patients with advanced non-small cell lung cancer: Randomised trial " Clinical Nutrition 33: 1017- 1023. Stewart, G. D., Skipworth, R.J., & Fearon, K.C. (2006). Cancer cachexia and fatigue Clinical Medicine, 6(2), 140-143. Tisdale, M. J. (2009). Mechanisms of Cancer Cachexia. Physiological Reviews, 89, 381-410. Tisdale, M. J., & Dhesi, J.K. (1990). Inhibition of Weight Loss by n-3 Fatty Acids in an Experimental Cachexia Model. Cancer Research, 50, 5022-5026. Wigmore, S. J., Ross, J.A., Falconer, S., Plester, C.E., Tisdale, M.J., Carter, D.C., & Fearon, K.C. (1996). The Effect of Polyunsaturated Fatty Acids on the Progress of Cachexia in Patients with Pancreatic Cancer Nutrition, 12(1), 27-30. Wu, D., & Meydani, S.N. . (1998). n-3 Polyunsaturated fatty acids and immune function Proceedings of the Nutrition Society, 57, 503-509. Yeh, K., Wang, H., Chang, J. W., Huang, J., Lai, C., Lan, Y., & Wu, T. (2013). "Omega-3 fatty acid-, micronutrient-, and probiotic-enriched nutrition helps body weight stabilization in head and neck cancer cachexia." Oral Surgery Oral Medicine Oral Pathology Oral Radiology 116(1): 41-48. Zuijdgeest-Van Leeuwen, S. D., Dagnelie, P.C., Wattimena, J.L., Van Den Berg, J.W., Van Der Gaast, A., Swart, G.R., & Paul Wilson, J.H. (2000). Eicosapentaenoic acid ethyl ester supplementation in cachectic cancer patients and healthy subjects: effects on lipolysis and lipid oxidation. Clinical Nutrition, 19(6), 417-423.