Tumor cachexia

from Wikipedia, the free encyclopedia
Classification according to ICD-10
C80 Malignant neoplasm cachexia.
Annotation:
ICD-10 online (WHO version 2019)

Cancer cachexia , rare cancerous cachexia called, is the name given to an as a result of cancer occurring in patients metabolic disorder leading to wasting ( cachexia ) and emaciation leads to those affected. Tumor cachexia is a common complication of cancer, especially malignant tumors of the digestive tract , which has an unfavorable effect on the course of the disease , quality of life and prognosis and can itself be directly life-threatening. Tumor cachexia is a major factor in the high death rate in cancer.

The molecular and biochemical causes that lead to tumor cachexia are highly complex and not yet fully understood. Metabolic products of the tumor and messenger substances that - as a result of cancer - are formed by the body of the person affected play an important role in the development of this syndrome . There are also psychological factors and side effects of cancer therapy measures that can lead to a loss of appetite in the patient. In contrast to the weight loss in the case of malnutrition , tumor cachexia not only reduces fat reserves, but also the skeletal muscles.

There is currently no effective curative treatment . So far , all therapeutic approaches have been palliative , that is, merely alleviate the disease.

Classification and definition

The development of cancer to the fatal stage

Most authors prefer the term syndrome for tumor cachexia . This designation is more appropriate to the complex situation than the classification as an independent disease. The Anglo-Saxon specialist literature usually speaks of cancer cachexia syndrome (CCS) or cancer anorexia-cachexia syndrome (CACS). Cachexia is a suitcase word from the Greek κακὸς = kakos = "bad" and ἕξiς = hexis = "condition".

Cachexia is generally described as a multifactorial syndrome that affects any area of ​​the body and is most easily identified in its advanced form from the massive loss of subcutaneous fat and skeletal muscle . There is currently no binding or standardized definition for either cachexia or tumor cachexia, which, given the high prevalence and severity of this syndrome, has repeatedly been admonished as inadequate. The same applies to a classification . One of the reasons for this is the lack of reliable biomarkers with which the tumor cachexia could be diagnosed and classified using measured values. Due to the lack of classification, there are currently no treatment regimens adapted to the degree of the syndrome.

To date, very different definitions of tumor cachexia have been used for clinical studies , based on the percentage loss of body mass or falling below a certain value in the body mass index , for example . The lack of a binding definition makes it much more difficult to compare clinical studies.

Depending on the definition chosen, there are different percentages of cancer patients who also suffer from tumor cachexia. For example, a group of 8541 cancer patients with solid tumors was analyzed according to four different criteria. According to the ICD-9 diagnosis of cachexia as a criterion, 2.4 percent of these patients would have tumor cachexia. With an expanded ICD-9 classification with cachexia, anorexia, abnormal weight loss, or problems with nutrition, it would have been 5.5 percent. If the prescription of drugs such as megestrol , oxandrolone , somatotropin or dronabinol , which indicate a medication for tumor cachexia, is used as a criterion, 6.4 percent of the patients fell under this criterion. If a loss of body mass of more than 5 percent were the definition of tumor cachexia, 14.7 percent of these cancer patients would have met this criterion.

The lack of a definition of cachexia has a negative impact on the diagnosis and treatment of affected patients. This also makes the development and approval of potential active ingredients more difficult. On December 13th and 14th, 2006, a new definition of cachexia was proposed at the Cachexia Consensus Conference in Washington, DC . According to them, cachexia is “a complex metabolic syndrome which is caused by a chronic illness and is characterized by the loss of muscle mass, with or without loss of body fat. Anorexia (loss of appetite), inflammation , insulin resistance and an increased breakdown of muscle proteins are common side effects of cachexia. Cachexia, which is associated with increased morbidity , must be distinguished from: malnutrition , age-related loss of muscle mass, primary depression , malassimilation (reduced use of nutrients) and hyperthyroidism (overactive thyroid) ”.

A current recommendation defines cachexia when the

  • Loss of body mass over 5 percent within twelve months or less, and if so
  • at least three of the five following criteria are met at the same time:

However, this proposed definition has not yet been used in either epidemiological or clinical studies.

Epidemiology and relevance to health policy

Type of cancer Weight Loss Incidence
Pancreatic tumor 83%
Stomach cancer 83%
Esophageal cancer 79%
Head and neck cancer 72%
Colorectal cancer 55 to 60%
Bronchial carcinoma 50 to 66%
Prostate cancer 56%
Breast cancer 10 to 35%
All cancers together average 63%

About 50 percent of all cancer patients are affected by tumor cachexia in the course of their illness. The likelihood of tumor cachexia is very much dependent on the type of cancer. After sepsis (blood poisoning), tumor cachexia is the most common cause of cancer death. Depending on the author, the proportion of tumor cachexia among the causes of death from cancer is 20 to 50 percent. The range of fluctuation in the prevalence data (disease frequency) is causally due to the previously mentioned lack of definition of tumor cachexia.

The type of cancer plays a major role in the likelihood of developing tumor cachexia. For example, around 80 percent of patients with pancreatic tumors and 30 to 50 percent of patients with cancer in the gastrointestinal tract die of tumor cachexia. In pancreatic tumors, the incidence of tumor cachexia is highest at 80 percent at the time the carcinoma is diagnosed. Tumor cachexia occurs in the course of the disease in up to 85 percent of all patients with gastrointestinal (gastrointestinal) cancer. With solid tumors, the probability of tumor cachexia is significantly higher than with cancers of the hematopoietic system ( leukemia , myelodysplastic syndromes and other hematological neoplasias). Breast cancer is an exception among solid tumors . The likelihood of tumor cachexia is significantly lower here. The development of tumor cachexia is very individual, even with the same type of tumor, depending on the patient. Tumor cachexia can be observed in affected patients in all stages of cancer, but is particularly common in the terminal stage of the disease.

Tumor cachexia affects children and the elderly significantly more frequently.

In absolute terms, tumor cachexia is one of the most common causes of death. In Germany, roughly 50,000 people die each year from tumor cachexia. Exact data are not available due to the lack of definition of tumor cachexia and the usual death certificate practice .

Clinical picture

Distribution of body mass in cachectic patients with bronchial carcinoma (right), compared to a healthy control group (left)

Tumor cachexia is essentially characterized by a weight loss in the patient, often associated with anorexia (loss of appetite), symptoms of inflammation , insulin resistance and the breakdown of skeletal muscles . In many cases, anorexia is a symptom of tumor cachexia, but tumor cachexia can develop without anorexia. The occurrence of anorexia accelerates the progression of tumor cachexia.

While weight loss in healthy individuals is compensated for by an increased appetite and the associated higher food intake, this is not the case with cachectic patients. Tumor cachexia leads to general weakness, decreased strength and immobility in the affected patients, as well as fatigue, listlessness and depression , which in turn leads to an impairment of the quality of life . The quality of life, as assessed by those affected, is significantly impaired by tumor cachexia. Some concomitant symptoms of tumor cachexia lead to the further progression of this syndrome through a self-reinforcing process . The breakdown of muscle proteins is associated with increased energy consumption, which in turn leads to disease progression. Loss of appetite and nausea lead to decreased food intake and also increase the catabolic state. Protein breakdown is essentially limited to the skeletal muscles. The internal organs , on the other hand, are hardly affected by protein breakdown. Here, too, the picture of tumor cachexia differs from that of malnutrition, in which - after the fat reserves have been used up - both skeletal and visceral proteins are broken down. The mass of the liver in cachectic patients can even increase significantly due to increased metabolic activities and the production of acute phase proteins .

diagnosis

A reliably diagnosed cancer is the first prerequisite for the diagnosis of tumor cachexia. The diagnosis of tumor cachexia itself is very difficult in most cases. Weight loss in a cancer patient does not have to be cachexia. A number of psychological factors also play a role, such as obstructions or stenoses in the gastrointestinal tract, which can be a result of tumor growth. Inflammation of the oral mucous membranes ( stomatitis ), dry mouth or fungal infections ( mycoses ) of the oral cavity ( oral thrush ) are possible side effects of cancer that have a negative effect on food intake. Diagnostic and above all therapeutic measures, in particular chemotherapy and radiation therapy , can lead to weight loss due to a decrease in appetite. Even if these influencing factors can be ruled out, a clear diagnosis is often difficult. The most important criteria for making a diagnosis at present are the patient's medical history and physical examination. The body weight before the onset of cancer serves as a reference . Also anthropometric studies can be used for diagnosis of cancer cachexia. In addition to the already mentioned elementary determination of body weight, these are, for example, measuring the circumference of the upper arm or the thickness of a skin fold. With the bioelectrical impedance analysis (BIA) the fat-free mass of the patient can be determined. However, this method is not available in many clinics and has not yet been established as a diagnostic standard for tumor cachexia.

Some laboratory parameters can be used to support the diagnosis. However, the informative value is often very limited due to physiological changes - caused by the underlying malignant disease and any therapeutic measures. The level of human albumin in the serum can be used as the laboratory parameter to support the diagnosis , which is usually lower in the case of tumor cachexia, but can be falsified in the case of liver and kidney dysfunction - for example caused by cancer. The concentration of C-reactive protein (CRP) in the serum can be increased as a result of an acute phase reaction (APR) which is frequently observed in tumor cachexia . Typical markers for an acute phase reaction are transferrin , transthyretin and ceruloplasmin . On the other hand, an acute phase reaction can also occur without cachexia in cancer, so that these markers are not a reliable measure for the diagnosis of “cachexia”. Increased levels of glycerol and catecholamines are often detectable in the blood of the patients . The increased glycerine content is due to the increased breakdown of body fats. Anemia is often an accompanying symptom of tumor cachexia .

At the time of diagnosis of cancer, about 80 percent of patients with tumors of the upper gastrointestinal tract and 60 percent of patients with lung cancer have lost significant weight (over 10 percent in six months). In many cases, the weight loss observed by the patient is the first symptom of the cancer, which - as part of the medical examination - is diagnosed as the cause of the weight loss.

At the time of the diagnosis of “cancer”, up to 50 percent of cancer patients already complain of anorexia in the form of loss of appetite and premature satiety.

Pathogenesis

The new model for the pathogenesis of tumor cachexia

Tumor cachexia arises from a complex interplay of metabolic products of the tumor and messenger substances that are formed in the body of the person affected as a result of the cancer, which has not yet been fully elucidated . These compounds cause a catabolic metabolism , an increased mobilization of fats from the adipose tissue, an increased breakdown and a reduced build-up of proteins in the skeletal muscles as well as an increased energy consumption of the body cells (hypermetabolism).

Tumor cachexia is a chronic inflammatory condition - similar to infection or inflammation or tissue damage. Various pro-inflammatory (pro-inflammatory) cytokines, prostaglandins and factors produced by the tumor itself are overexpressed . These substances intervene directly in the peripheral and central control loops of food intake and metabolism and are also able to trigger muscle atrophy. In addition, there are other factors such as decreased food intake, poor digestion ( maldigestion ) or absorption ( malabsorption ), among other things as a result of the patient's depressed mood . Disorders in taste or the hunger center can also contribute to tumor cachexia. Recurrent bleeding, for example in ulcerating or polypoid gastrointestinal carcinomas , can also lead to an increased loss of proteins in the body.

Cytokines

From a certain critical size of a tumor, tumor necrosis sets in . This means that in the center of the tumor - due to an insufficient supply of blood - tumor cells die. When these tumor cells disintegrate, the tumor necrosis factor TNF-α (formerly known as cachectin in relation to cachexia ) and a number of other cytokines are released . These substances cause functional disorders in hepatocytes (liver cells) in particular and lead to a negative nitrogen balance. The metabolism is switched to catabolism . The cytokines TNF-α, interleukin-1, interleukin-6 and interferon-γ have been identified in various experiments as mediators for the loss of muscle mass in tumor cachexia. In fact, these compounds are in principle capable of causing cachexia. In animal models, for example, the state of cachexia can be established experimentally through the injection of TNF-α. TNF-α causes both a breakdown of body fat and the skeletal muscles. TNF-α directly influences the ubiquitin-proteasome system , the most important intracellular protein degradation mechanism. The formation of reactive oxygen species (ROS) leads to an increased expression of the transcription factor NF-κB . TNF-α lowers protein synthesis through reduced phosphorylation of the eukaryotic translation initiation factor-4E eIF4E , which can thereby bind more strongly to EIF4EBP1 (eukaryotic translation initiation factor 4E binding protein-1) and reduces the active eukaryotic initiation factor 4F ( eIF4F ) . For a long time it was therefore assumed that the cytokines mentioned - and above all TNF-α - are the main cause of tumor cachexia.

In studies of patients with advanced and end-stage cancer, however, it was found that there was no correlation between the concentration of these cytokines and weight loss and anorexia in those affected. Also in cancer patients - in comparison to healthy people - no higher serum concentrations of TNF-α could be detected. In contrast, the plasma levels of these cytokines are significantly increased in patients with cachexia caused by AIDS or septicemia . The fact that cytokines can inhibit the enzyme lipoprotein lipase also speaks in favor of cytokines as a trigger of tumor cachexia . As a result of the inhibition, the adipocytes can not build up any triglycerides (fats) from the lipoproteins in the plasma and consequently cannot store them. Interestingly, however, the total activity of lipoprotein lipase and also the concentration of the corresponding mRNA in adipose tissue in cancer patients are unchanged compared to healthy individuals, while that of hormone-sensitive lipase (HSL) is about twice as high. The inhibition of TNF-α production - as a possible therapeutic approach - does not lead to any improvement in the state of health. Based on these data, it is currently assumed that TNF-α, interleukin-1, interleukin-6 and interferon-γ are in principle capable of causing cachexia - as in AIDS or sepsis, for example - but in the case the pathogenesis of tumor cachexia are obviously not the main factor.

Metabolic products of the tumor

Schematic representation of the interactions between tumor and healthy body tissue in tumor cachexia

Tumors produce catabolic messenger substances. Of the messenger substances identified so far, the proteolysis-inducing factor (PIF) and the lipid-mobilizing factor (LMF) are the most important factors in the pathogenesis of tumor cachexia. These factors cause complex, not yet fully understood, neurohormonal and metabolic changes that can lead to catabolic metabolism and nutritional deficiencies.

Lipid mobilizing factor

In the mid-1990s, a peptide was isolated for the first time from the urine of patients with tumor cachexia , which cannot be detected in cancer patients without weight loss and which is able to induce lipolysis in animal models . The peptide, known as the lipid-mobilizing factor , acts directly on the adipocytes and stimulates lipolysis there via a cAMP- dependent process that is mechanistically similar to that of lipolytic hormones. LMF is a 43  kDa heavy homolog binding zinc to the plasma protein α-2-Glycoprotein ( AZGP1 , also called ZAG). If LMF is injected into mice, the treated animals lose body fat without changing the animals' food intake. ZAG is overexpressed by the univacuolar adipocytes of the white adipose tissue in cachectic mice. Based on the data, it is currently assumed that LMF makes a significant contribution to tumor cachexia in the breakdown of body fat.

Proteolysis-inducing factor

In the mid-1990s, a sulfated glycoprotein with a molar mass of 24 kDa was isolated in mice with MAC16 adenocarcinoma , which leads to tumor cachexia by inducing catabolism in the skeletal muscles. This peptide , later called proteolysis-inducing factor (PIF), was also found in the urine of patients with tumor cachexia. In contrast, it could not be detected in cancer patients without weight loss or in patients with weight loss not initiated by cancer. These results have been confirmed in a number of other tests and studies. The detection of PIF is indicative of weight loss in tumor cachexia. NF-κB mediates the protein synthesis induced by PIF in the skeletal muscles through an increased phosphorylation of eIF2 on its α-subunit. Blocking the PIF receptor or the signal cascade in the skeletal muscles is seen as a potential starting point for future drugs for the treatment of tumor cachexia.

In animal experiments, in addition to the breakdown of the skeletal muscles, an increased production of prostaglandin E2 was demonstrated. In vitro, PIF binds with a very high affinity in the nanomolar range to the sarcolemma of skeletal muscle cells in mice, pigs and humans and to murine myoblasts . PIF is a potential marker for diagnosing tumor cachexia. The role of PIF in human tumor cachexia is not rated equally by all working groups and is the subject of controversial discussions. In some studies, results were obtained that contradict the previous data. Also, no correlation could be found between the presence / absence of PIF and a patient's prognosis.

anorexia

→ see also main article anorexia

For a long time, it was assumed that malnutrition and weight loss in many cancer patients are the sole consequences of anorexia. Obstructions (constrictions, occlusions) in the gastrointestinal tract, pain , side effects of cancer therapy, nausea or changes in gustatory perception (sense of taste) caused by the tumor can cause loss of appetite. Since anorexia can occur in cancer patients even in the absence or treatment of these symptoms , it is believed that changes caused by the tumor cause the loss of appetite. The body mass index (BMI) of cancer patients correlates with the concentration of leptin in the same way as that of healthy comparators . This means that if the BMI value in the serum is high, high leptin concentrations are measured. Leptin is an important messenger substance produced by adipocytes that inhibits the feeling of hunger . Certain cytokines can affect the production of leptin in adipocytes. In advanced cancer patients, the serum concentration of the cytokine interleukin-6 (IL-6) is significantly increased, which leads to a decrease in the blood leptin level in those affected. The survival rate in patients with high IL-6 concentrations, and the resulting particularly low leptin levels, is significantly reduced. In addition to the altered leptin production, the activity of the enzyme fatty acid synthase (FAS) and the melanocyte-stimulating hormone (MSH) obviously play an important role in anorexia.

If the loss of appetite in cancer is treated with medication, for example with substances that stimulate the appetite , or through artificial nutrition ( enteral or parenteral ), there is no improvement in metabolism towards anabolism (building up body mass) and away from catabolism (breaking down body mass ) reached. Measurable increases in weight, which are achieved through the administration of appetite-stimulating substances, are limited to the increase in fatty tissue and the storage of water in the interstitium of the treated cachectic patients. Muscle mass is hardly built up.

In contrast to anorexia, in which the lean body mass (lean mass) is largely retained, in tumor cachexia the skeletal muscles are also broken down. Up to 80 percent of the fatty tissue and skeletal muscles can be lost. For example, in patients with lung cancer who have lost 30 percent of their original body mass due to the disease, the weight loss results from a reduction in adipose tissue by 85 percent and that of skeletal muscle protein by 75 percent.

Anorexia is an additional symptom - very often accompanying tumor cachexia - that is the result of a disturbed appetite signaling pathway and is not responsible for the massive loss of muscle protein in cancer. In the Anglo-Saxon language area, the term Cancer Anorexia-Cachexia Syndrome (CACS) has established itself in place of the term Cancer Cachexia .

Direct influences of the tumor

The old model for the pathogenesis of tumor cachexia

According to the outdated development model of tumor cachexia, it was assumed that the increased energy requirement of the tumor is essentially responsible for this syndrome. This thesis was generally accepted until the 1980s and is still widespread among the population today - but no longer tenable in this form. Larger tumors can cause the affected patient to need more nutrients. However, this increased nutrient requirement is not the cause of tumor cachexia.

Tumor cachexia can occur regardless of the size and extent of the tumor and regardless of metastasis. The risk of developing tumor cachexia is much more dependent on the type of cancer than, for example, on the size of the tumor, the tumor location and the degree of metastasis. Cachexia can be observed in certain tumor types as early as 5 cm³ tumor volume, while large tumors do not trigger cachexia in other carcinomas. This is an indication that cachectic tumors have an altered gene expression that allows the tumor cells to produce lipolytic (fat-degrading) and proteolytic (protein-degrading) proteins that enable cachexia.

In a number of different studies was examined whether the resting energy expenditure ( resting energy expenditure , REE) is increased from cachectic cancer patients. The data are partly contradicting or without a significant cause-effect relationship . In some cases, the studies show an increased energy requirement, partly the exact opposite or an unchanged energy requirement, so that there are currently no reliable findings on this.

Larger tumors can have an additional energy requirement of up to 300  kcal per day. Tumors consume large amounts of glucose , which, due to the anaerobic conditions in the tumor, is broken down into lactate . The lactate is converted back into glucose in the liver in the so-called Cori cycle . This process is very energy-intensive. In healthy people, the proportion of glucose converted via the Cori cycle is around 20 percent, in cachectic patients around 50 percent. This then corresponds to a share of 60 percent of the total lactate production. Although this additional energy requirement is not the cause of tumor cachexia, it is an important aspect of the patient's diet.

Therapy and future therapeutic approaches

There is currently no drug approved by the FDA or the European Commission for the treatment of tumor cachexia. Some dietary supplements , as well as finished medicinal products that are approved for other indications ( off-label use ) , are sometimes used for treatment. Their use is purely palliative . An immediate curative treatment of tumor cachexia is currently not known. A cure is only possible if the cancer underlying the tumor cachexia is eliminated (indirect treatment). This would be the most effective therapy for tumor cachexia. Since tumor cachexia often only occurs in a late stage of cancer, the chances of a cure for the cancer and thus the tumor cachexia are usually very low. In many cases it is no longer possible to cure the underlying disease “cancer” through therapeutic measures. The affected patients are resistant to therapy - "out of therapy ". The main therapeutic goal of tumor cachexia is to significantly improve the quality of life of the affected patient. In addition, the overall survival time should be increased and the body strengthened for tumor therapeutic measures (surgery, chemotherapy , radiation therapy ).

The currently established measures for the treatment of tumor cachexia are inadequate in their effectiveness, so that the success of treatment is very modest. The reasons for this lie on the one hand in the incomplete knowledge about the pathogenesis of tumor cachexia and on the other hand in the multitude of influencing factors on the pathogenesis itself. Due to the latter aspect, it is currently assumed that a single therapy alone - also in the future - will not be the universal one Solution will be. Rather, there is a need to combine several types of treatment. Some oncologists see future cancer therapies combined with therapy for anorexia and tumor cachexia right at the beginning of the diagnosis of "cancer". This promises synergistic effects, which are reflected on the one hand in a better response rate in tumor therapy and on the other hand in a significantly improved quality of life.

Increase in food intake and appetite stimulant

The most obvious therapeutic measure is to increase the patient's food intake. Even if the patient's loss of appetite can be overcome and the nutritional needs are more than met - if necessary through artificial nutrition with a high physiological calorific value - these measures alone do not lead to an increase in lean and anhydrous body mass. The catabolic protein breakdown in the skeletal muscles cannot be stopped or even reversed. The administration of substances that only stimulate the appetite can therefore not prevent the catabolic degradation.

Ghrelin

→ see main article Ghrelin
The ribbon model of preproghrelin

Ghrelin (engl. G rowth H ormone Rel ease In ducing ) is from 28 amino acids existing appetizing peptide hormone . It is formed from the precursor protein preproghrelin - which consists of 117 amino acids - through post-translational modification in the gastric mucosa . It is the only currently identified hormone that circulates in the human body and stimulates appetite. Ghrelin stimulates the release of neuropeptide Y (NPY), which among other things affects hunger and the motility of the gastrointestinal tract.

In preclinical experiments with model organisms , promising results have been obtained in the treatment of cachectic mice with ghrelin. The stimulation of the appetite and an increased food intake could be demonstrated; likewise the building of muscle mass. The positive effects against digestive disorders and vomiting when chemotherapy was carried out at the same time were also surprising .

Ghrelin can be administered subcutaneously or intravenously . It is generally well tolerated. Undesirable side effects are largely unknown. Ghrelin does not stimulate tumor growth. In the animal model and in the first tests on humans, however, ghrelin resistance was found after repeated administration, which could be compensated by higher doses. The mechanism of resistance development is similar to that of insulin resistance. The dangers of diabetes mellitus when taking ghrelin for long periods of time are also discussed.

Ghrelin is still in clinical trials. Proof of effectiveness in humans with the indication tumor cachexia (successful phase III) is still pending. Only then can drug approval and market launch take place.

Megestrol

The structural formula of Megestrol

Megestrol is a sex hormone from the progesterone group . The substance has been shown to have an orexigenic (appetite-stimulating) effect. In 1993, Megestrol was approved by the FDA for the treatment of anorexia, cachexia, or unexplained weight loss in AIDS patients. There is no approval for cancer patients with tumor cachexia. This medicinal product is well tolerated and has few side effects. The mechanism of action for appetite stimulation is still unclear. Megestrol has been shown to increase appetite in patients with tumor cachexia. Weight gain can also be demonstrated. A significant increase in quality of life could not be proven in the studies carried out so far. The survival time could also not be increased compared to patients who received a placebo .

Megestrol is effective in the presence of anorexia without cachexia. In contrast, the weight gain in cachectic patients is essentially due to the increase in adipose tissue and the storage of water in the interstitium. However, the desired effect of increasing skeletal muscle mass does not materialize.

Cannabinoids

The cannabinoids contained in hemp have an appetite-stimulating effect, but have not been able to provide any proof of effectiveness in the treatment of tumor cachexia in controlled studies.

Endocannabinoids have an appetite-stimulating effect in humans. Plant cannabinoids , for example from hemp ( Cannabis sativa ), have been known to have the same effect since ancient times. The appetite-stimulating effect has been proven for Δ9- tetrahydrocannabinol (THC), the main active ingredient of Cannabis sativa , and the partly synthetically produced THC dronabinol . The substance is approved in the United States as a drug for the treatment of anorexia and cachexia in AIDS and as an antiemetic in cancer.

In a large-scale, multicentre, randomized, placebo-controlled, double-blind phase III study with 289 patients over a period of six weeks, however, no difference was found between the THC arm and the placebo arm with regard to appetite, nausea, mood and quality of life. Other comparative studies come to similar results.

Other appetite stimulants or serotonin antagonists have also shown in clinical studies that they cannot stop the progressive weight loss in tumor cachexia.

Inhibition of acute phase proteins or their messenger substances

Acute phase proteins (APP) are mainly produced in the liver and released into the bloodstream in acute or chronic inflammatory reactions. The production of the acute phase proteins is mainly stimulated by the messenger substances tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). C-reactive protein as well as various transport and complement proteins are produced as acute phase proteins . The APPs affect the central nervous system , among other things, and influence appetite, eating habits and metabolism there. Which APP works how and where exactly is not yet fully known and is the subject of current research. The production of APPs is significantly increased especially in tumors in the organs of the pancreas , lungs , esophagus and kidneys . A therapeutic approach to the treatment of tumor cachexia consists in the inhibition of the acute phase proteins or the messenger substances that stimulate the production of APPs in the liver.

Nonsteroidal anti-inflammatory drugs

Compounds with anti-inflammatory effects, such as the nonsteroidal anti-inflammatory drugs (NSAID) ibuprofen or indomethacin , inhibit the acute phase proteins non-specifically. The protein metabolism was positively influenced both in the animal model and in patients with a pancreatic tumor or colorectal cancer . With indomethacin, survival time was significantly increased. In addition to these unspecific approaches to inhibiting acute phase proteins, research is also being carried out into the specific inhibition of individual APPs - or of messenger substances that stimulate the production of the APPs. An example of this is the inhibition of interleukin-6. However, previous attempts have been unsuccessful.

Steroidal anti-inflammatory drugs

Steroidal anti-inflammatory drugs such as the glucocorticoid prednisolone have also shown positive results in clinical studies. Glucocorticoids are often given to treat tumor cachexia. The usually undesirable side effect of weight gain with glucocorticoids is desirable in this application. Prednisolone and dexamethasone significantly increase the appetite of patients and thus improve their quality of life. In addition, cytokines are inhibited via the anti-inflammatory effect. The effect of the glucocorticoids is short-lived and the condition of the skeletal muscles is not improved by the administration of glucocorticoids. In some studies, the overall survival time is significantly increased compared to the placebo administration. The undesirable side effects of glucocorticoids, such as nausea, pain, water retention , weakness and insulin resistance or even osteoporosis and immunosuppression, are so significant that the therapeutic benefit is very questionable and controversial. Glucocorticoids are not well established in the treatment of tumor cachexia.

TNF-α inhibitors (thalidomide)

The structural formula of thalidomide, better known under the brand name Contergan

The approaches to inhibit TNF-α are more promising. Thalidomide - much better known under the brand name Contergan - is a selective antagonist of TNF-α and TGF-β . Positive results were obtained in the animal model as well as in the first clinical studies. For example, the administration of thalidomide resulted in an increase in lean and anhydrous body mass in eight out of ten patients with unresectable esophageal cancer . The breakdown of skeletal muscles was also significantly delayed in patients with pancreatic cancer. However, the survival time of the thalidomide-treated patients - compared to the placebo group - could not be increased, although tumor cachexia is the direct cause of death in a large number of patients with pancreatic cancer. Thalidomide and other inhibitors are being tested in a number of controlled clinical studies for the treatment of tumor cachexia and have yet to prove their therapeutic effectiveness.

Inhibition of proteasome activity

The breakdown of the proteins of the skeletal muscles takes place - regardless of the triggering messenger substance - via the ubiquitin-proteasome system. A therapeutic starting point is therefore to reduce the activity of the proteasome. Several proteasome inhibitors are in clinical trials. The first proteasome inhibitor to be approved in the US and the EU, bortezomib , is effective against multiple myeloma .

3-hydroxy-3-methylbutyric acid

The structural formula of 3-hydroxy-3-methylbutyric acid

3-Hydroxy-3-methylbutyric acid (HMB), usually referred to as β-hydroxy-β-methylbutyric acid or β-hydroxy-β-methylbutyrate, is a metabolic product of the essential amino acid leucine . About 5 percent of the ingested leucine is metabolized to HMB. HMB is offered as a dietary supplement and shows, among other things, anabolic, anti-catabolic and lipolytic effects in the human body . HMB is therefore taken by many bodybuilders and strength or endurance athletes to legally increase muscle mass or performance. In well-trained athletes, however, neither aerobic nor anaerobic performance can be measured through taking HMB.

In the animal model of cachectic mice, it was possible to show that the breakdown of protein is reduced and the build-up of muscle mass is stimulated. If HMB is administered to the test animals together with PIF, which upregulates the ubiquitin proteasome system, the effect of PIF can be completely compensated for by HMB. HMB apparently has a regulating effect on the expression of NF-κB, which is less strongly produced by the cells. The mechanism for building protein mass is via the mTOR receptor, the phosphorylation of which is apparently stimulated by HMB.

A number of clinical studies with healthy volunteers, trained and untrained, have been conducted with HMB. The results are partly contradictory. However, there are conclusive indications that the administration of HMB for tumor cachexia could be an effective future form of therapy. In a study with cachectic patients, an increase in lean body mass was demonstrated by the administration of HMB in combination with the amino acids arginine and glutamine . In a phase III, double-blind, randomized, placebo-controlled study of 472 patients, lean body mass increased in patients who received HMB with arginine and glutamine. However, only 37 percent of all patients completed the study, as a result of which the primary and secondary endpoints could not be achieved and therefore no proof of the effectiveness of HMB for the treatment of tumor cachexia is provided. Further, large-scale studies are necessary to provide evidence of effectiveness.

Bortezomib

→ see main article bortezomib
The structural formula of bortezomib

Bortezomib is a proteasome inhibitor approved for the treatment of multiple myeloma . In clinical studies that aimed to delay or prevent the proteolytic muscle breakdown occurring in tumor cachexia, the compound did not show sufficient effectiveness.

Eicosapentaenoic acid

→ see main article eicosapentaenoic acid
The structural formula of eicosapentaenoic acid

Eicosapentaenoic acid , usually abbreviated as EPA ( Eicosapentaenoic acid ), is a polyunsaturated fatty acid from the class of omega-3 fatty acids . It is one of the main components of certain fish oils, especially fatty fish . EPA has anti-inflammatory properties and is the only dietary supplement that is able to influence the proteasome through various mechanisms . The activity of the proteasome, which breaks up proteins into fragments inside the body's cells, is reduced by EPA. According to the model of action, it should be possible to reduce the breakdown of muscle proteins. In the first studies on patients with pancreatic cancer , cachectic weight loss could be reduced. The EPA was well tolerated by the patients without significant side effects. This success could not be repeated in three subsequent, large-scale, placebo-controlled, randomized double-blind studies . In the “mouse” animal model, no influence of EPA on protein synthesis could be determined.

Other therapy concepts

insulin

→ see main article insulin

The hormone insulin is a potent regulator of protein turnover in the body. The administration of insulin can stimulate the uptake of carbohydrates in cachectic patients. In a clinical study, the patients with various tumor diseases gained body weight, but mainly due to an increased percentage of body fat. The lean body mass remained unchanged. The administration of insulin did not affect tumor growth and the survival time was slightly increased compared to the control group.

Hydrazine sulfate

Hydrazine sulfate was one of the first active ingredients to be used specifically against tumor cachexia. The compound is an inhibitor of gluconeogenesis , i.e. the formation of glucose in the body from non-carbohydrates. Hydrazinium sulfate deactivates the enzyme phosphoenolpyruvate carboxykinase . In the first clinical studies in the treatment of cancer patients at a late stage of the disease, improved glucose tolerance, reduced glucose metabolism, increased uptake of nutrients and a stabilization or even increase in body weight could be determined. The undesirable side effects were minor. The intention at the time to administer hydrazinium sulfate was to start from the hypothesis that uncontrolled glucogenesis was closely related to tumor cachexia.

In later studies it was found that the administration of hydrazine sulfate for the treatment of tumor cachexia is ineffective, making this therapeutic approach obsolete .

Gene therapy

Follistatin and myostatin are two natural proteins that control muscle growth. While follistatin stimulates muscle growth, myostatin does the exact opposite. Both compounds form a control loop that controls muscle growth in mammals . The administration or cellular overexpression of follistatin, or the inhibition of myostatin or the switching off of the gene coding for myostatin , are potential therapeutic approaches for tumor cachexia. With these approaches it may be possible to counteract the reduction in muscle mass caused by tumor cachexia. The first successes were achieved in animal models. This therapeutic approach is still in the preclinical phase and - even with proof of effectiveness - is still many years away from approval as a medicinal product.

forecast

The severity of tumor cachexia correlates inversely with the mean survival time of a cancer patient. This means that the more pronounced the tumor cachexia, the shorter the survival time of the person affected. Basically, tumor cachexia is associated with a poor prognosis for the patient. Weight loss with cancer is an independent prognostic factor. Tumor cachexia increases the likelihood of postoperative complications and also has a negative impact on the success of chemotherapy. Patients with breast cancer without tumor cachexia respond on average 2.5 times better to chemotherapy than patients with tumor cachexia. Even a relatively small loss of body mass of less than five percent, for example, can significantly worsen the prognosis.

There is only a chance of a cure if the cancer underlying the tumor cachexia is cured.

literature

Reference books

  • Angela P. Löser u. a .: Outpatient care for tumor patients . Schlütersche Verlag , Hannover 2000, ISBN 3-87706-479-5 , p. 195 ff .
  • David S. Ettinger (Editor): Supportive Care in Cancer Therapy . Verlag Springer , Luxemburg 2008, ISBN 1-58829-941-4 .
  • Jeffrey A. Norton et al. a .: Surgery: basic science and clinical evidence . Verlag Springer, Luxemburg 2008, ISBN 0-387-30800-8 , pp. 2123 ff .
  • Giovanni Mantovani: Cachexia and wasting: a modern approach . Verlag Springer, Luxemburg 2006, ISBN 88-470-0471-3 .
  • Karl G. Hofbauer u. a .: Pharmacotherapy of cachexia . Verlag Auerbach, Leipzig 2006, ISBN 0-8493-3379-2 .

Review article

  • M. Bossola et al. a .: Skeletal muscle in cancer cachexia: the ideal target of drug therapy. In: Curr Cancer Drug Targets 8, 2008, pp. 285-298, PMID 18537552
  • A. Jatoi: Weight loss in patients with advanced cancer: effects, causes, and potential management. In: Curr Opin Support Palliat Care 2, 2008, pp. 45-48, PMID 18685394
  • S. Al-Majid and H. Waters: The biological mechanisms of cancer-related skeletal muscle wasting: the role of progressive resistance exercise. In: Biol Res Nurs 10, 2008, pp. 7-20, PMID 18705151
  • ML Hardy: Dietary supplement use in cancer care: help or harm. In: Hematol Oncol Clin North Am 22, 2008, pp. 581-617, PMID 18638690
  • B. Seruga et al. a .: Cytokines and their relationship to the symptoms and outcome of cancer. In: Nat Rev Cancer 8, 2008, pp. 887-899, PMID 18846100
  • N. Bennani-Baiti and MP Davis: Cytokines and cancer anorexia cachexia syndrome. In: Am J Hosp Palliat Care 25, 2008, pp. 407-411, PMID 18403577
  • KD Hall and VE Baracos: Computational modeling of cancer cachexia. In: Curr Opin Clin Nutr Metab Care 11, 2008, pp. 214-221, PMID 18403915
  • M. Bossola et al. a .: Novel treatments for cancer cachexia. In: Expert Opin Investig Drugs 16, 2007, pp. 1241-1253, PMID 17685872
  • A. Lelbach et al. a .: Current perspectives of catabolic mediators of cancer cachexia. In: Med Sci Monit 13, 2007, RA168-173, PMID 17767131
  • JL Ryan et al. a .: Mechanisms of cancer-related fatigue. In: Oncologist 12, 2007, pp. 22-34, PMID 17573453
  • JM Argilés u. a .: Mechanisms to explain wasting of muscle and fat in cancer cachexia. In: Curr Opin Support Palliat Care 1, 2007, pp. 293-298, PMID 18685378
  • MS Boddaert u. a .: On our way to targeted therapy for cachexia in cancer? In: Curr Opin Oncol 18, 2006, pp. 335-340, PMID 16721127
  • JM Argilés u. a .: Cytokines as mediators and targets for cancer cachexia. In: Cancer Treat Res 130, 2006, pp. 199-217, PMID 16610709
  • JM Argilés u. a .: Mediators involved in the cancer anorexia-cachexia syndrome: past, present, and future. In: Nutrition 21, 2005, pp. 977-985, PMID 16043325
  • JM Argilés u. a .: Cytokines in the pathogenesis of cancer cachexia. In: Curr Opin Clin Nutr Metab Care 6, 2003, pp. 401-406, PMID 12806213
  • ME Martignoni u. a .: Cancer cachexia. In: Mol Cancer 2, 2003, p. 26, PMID 14613583
  • H. Rubin: Cancer cachexia: its correlations and causes. In: PNAS 100, 2003, pp. 5384-5389, PMID 12702753
  • B. Gagnon and E. Bruera: A review of the drug treatment of cachexia associated with cancer. In: Drugs 55, 1998, pp. 675-688, PMID 9585863

Web links

Individual evidence

  1. The classification in C80 is valid according to ICD-10 (WHO version) from 2006. Deviating from this, tumor cachexia is classified in R64 (cachexia) in the ICD-10-GM version from 2009 . The same applies to the German coding guidelines of 2005.
  2. a b c d e f g h M. J. Tisdale: Cachexia in cancer patients. In: Nat Rev Cancer 2, 2002, pp. 862-871, PMID 12415256 .
  3. J. Inagaki et al. a .: Causes of death in cancer patients. In: Cancer 33, 1974, pp. 568-573, PMID 4591273 .
  4. RD Lohberg u. a .: The lethal phenotype of cancer: the molecular basis of death due to malignancy. ( Memento of April 8, 2010 on the Internet Archive ) In: CA Cancer J Clin 57, 2007, pp. 225-241, PMID 17626119 (review).
  5. ^ AM Lindsey: Cancer cachexia: Effects of the disease and its treatment. In: Semin Oncol Nurs 2, 1986, pp. 19-29, PMID 3513270 .
  6. R. Truig -Burfeind: WAHRIG foreign words dictionary. Bertelsmann Lexikon Verlag, 2007, ISBN 3-577-09030-8 .
  7. T. Ohnuma: Complications of Cancer and Its Treatment. ( Memento of the original from August 31, 2004 in the Internet Archive ) Info: The archive link was inserted automatically and has not yet been checked. Please check the original and archive link according to the instructions and then remove this notice. (PDF; 486 kB) In: Holland-Frei Cancer Medicine , 6th Edition, DW Kufe u. a. (Editors), BC Decker Inc, Hamilton, Ontario, 2003, ISBN 1-55009-213-8 , Section 38: Complication of Cancer and its Treatment , Chapter 144: Cancer Anorexia and Cachexia. Pp. 2425-2441. ( Complete works ). @1@ 2Template: Webachiv / IABot / cancer.yu.ac.kr
  8. a b c KC Fearon: Cancer cachexia: developing multimodal therapy for a multidimensional problem. In: Eur J Cancer 44, 2008, pp. 1124-1132, PMID 18375115 (review).
  9. J. Springer et al. a .: The need for a standardized definition for cachexia in chronic illness. In: Nat Clin Pract Endocrinol Metab 2, 2006, pp. 416-417, PMID 16932326 .
  10. KC Fearon et al. a .: Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. In: American Journal of Clinical Nutrition 83, 2006, pp. 1345-1350, PMID 16762946 (Review).
  11. M. Lainscak et al. a .: How does cachexia influence survival in cancer, heart failure and other chronic diseases? In: Curr Opin Support Palliat Care 1, 2007, pp. 299-305, PMID 18685379 (review).
  12. a b c K. M. Fox et al. a .: Estimation of Cachexia among Cancer Patients Based on Four Definitions. In: J Oncol 2009: 693458, PMID 19587829 .
  13. a b W. J. Evans et al. a .: Cachexia: a new definition. In: Clinical Nutrition 27, 2008, pp. 793-799, PMID 18718696 .
  14. a b A. Laviano et al. a .: Therapy Insight: cancer anorexiacachexia syndrome when all you can eat is yourself. In: Nature Clinical Practice Oncology 2, 2005, pp. 158-165, PMID 16264909 (review).
  15. a b c D. E. Rivadeneira: Nutritional support of the cancer patient. ( September 7, 2008 memento on the Internet Archive ) In: CA Cancer J Clin 48, 1998, pp. 69-80, PMID 9522822 (review).
  16. a b c J. A. Palesty and SJ Dudrick: What we have learned about cachexia in gastrointestinal cancer. In: Dig Dis 21, 2003, pp. 198-213, PMID 14571093 (review).
  17. G. Ollenschläger : Nutritional problems in palliative medicine. In: Textbook of Palliative Medicine E. Aulbert and D. Zech (eds.), Verlag Schattauer, 1997, ISBN 3-7945-2361-X , pp. 556-565.
  18. L. Stepp and TS Pakiz: anorexia and cachexia in advanced cancer. In: Nurs Clin North Am 36, 2001, pp. 735-744, PMID 11726350 (Review).
  19. a b c d E. Bruera: ABC of palliative care: anorexia, cachexia, and nutrition. In: BMJ 315, 1997, pp. 1219-1222, PMID 9393230 (review).
  20. ^ S. Warren: The immediate causes of death in cancer. In: American Journal of Medical Science 184, 1932, pp. 610-616.
  21. NJ Espat et al. a .: Cytokine-mediated alterations in host metabolism prevent nutritional repletion in cachectic cancer patients. In: J Surg Oncol 58, 1995, pp. 77-82, PMID 7844987 (review).
  22. a b c G. Uomo u. a .: Anorexia-Cachexia Syndrome in Pancreatic Cancer: Recent Development in Research and Management. In: J Pancreas 7, 2006, pp. 157-162, PMID 16525199 (review).
  23. a b c C. Deans and SJ Wigmore: Systemic inflammation, cachexia and prognosis in patients with cancer. In: Curr Opin Clin Nutr Metab Care 8, 2005, pp. 265-269, PMID 15809528 .
  24. a b c d e f g A. Inui: Cancer anorexia-cachexia syndrome: current issues in research and management. ( July 1, 2010 memento on the Internet Archive ) In: CA Cancer J Clin 52, 2002, pp. 72-91, PMID 11929007 (review).
  25. a b c d e f K. CH Fearon and AGW Moses: Cancer cachexia. In: Int J of Cardiology 85, 2002, pp. 73-81, PMID 12163211 .
  26. a b c d e f g h i j k l m n S. Zimmer: Phenotypic characterization of weight loss and changes in appetite in cancer patients. (PDF; 466 kB) Dissertation, Philipps University Marburg, 2007.
  27. S. Blanke: Influence of a nutritive oral supportive therapy on the nutritional status of tumor patients in a radiation therapy department. Dissertation, Medical Faculty of the Charité, 2007.
  28. a b K. CH Fearon: The mechanisms and treatment of weight loss in cancer. In: Proc Nutr Soc 51, 1992, pp. 251-265, PMID 1438334 (review).
  29. KF Giordano and A. Jatoi: The cancer anorexia / weight loss syndrome: therapeutic challenges. In: Current Oncology Reports 7, 2005, pp. 271-276, PMID 15946586 (Review).
  30. a b J. M. Argiles et al. a .: Muscle wasting in cancer and aging: cachexia versus sarcopenia. In: Advances in Gerontology 18, 2006, pp. 39-54, PMID 16676797 (Review).
  31. ^ P. O'Gorman et al. a .: Longitudinal study of weight, appetite, performance status, and inflammation in advanced gastrointestinal cancer. In: Nutr Cancer 35, 1999, pp. 127-129, PMID 10693165 .
  32. a b c W. D. Dewys et al. a .: Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. In: Am J Med 69, 1980, pp. 491-497, PMID 7424938 .
  33. H. Iwagaki et al. a .: Cancer cachexia and depressive states: a neuro-endocrine-immunological disease? In: Acta medica Okayama 51, 1997, pp. 233-236, PMID 9284972 .
  34. DR Brown et al. a .: Weight loss is not associated with hyperleptinemia in humans with pancreatic cancer. In: J Clin Endocrinol Metab 86, 2001, pp. 162-166, PMID 11231995 .
  35. a b c N. Mac Donald et al. a .: Understanding and managing cancer cachexia. In: J Am Coll Surg 197, 2003, pp. 143-161, PMID 12831935 .
  36. DL Marks u. a .: Role of Central Melanocortin System in Cachexia. In: Cancer Research 61, 2001, pp. 1432-1438, PMID 11245447 .
  37. a b E. G. Berenstein and Z. Ortiz: megestrol acetate for the treatment of anorexia-cachexia syndrome. In: Cochrane Database Syst Rev 2, 2005, CD004310, PMID 15846706 (Review).
  38. P. Ravasco et al. a: Cancer: disease and nutrition are key determinants of patients' quality of life. In: Support Care Cancer 12, 2004, pp. 246-252 PMID 14997369 .
  39. K. Fearon et al. a .: Pancreatic Cancer as a Model: Inflammatory Mediators, Acute Phase Response and Cancer Cachexia. In: World J Surgery 23, 1999, pp. 584-588, PMID 10227928 (Review).
  40. M. Tisdale: Biomedicine: Protein Loss in Cancer Cachexia. In: Science 289, 2000, pp. 2293-2295, PMID 11041796 .
  41. G. Zurcher: Anorectic Syndrome. In: Z Gastroenterology 40, 2002, pp. 71-75, PMID 11930295 .
  42. a b R. Meier: Pathogenesis of Tumorkachexie. (PDF; 110 kB) In: Swiss Journal for Nutritional Medicine 4, 2007.
  43. a b M. J. Tisdale: Mechanisms of cancer cachexia. In: Physiol Rev 89, 2009, pp. 381-410, PMID 19342610 (review).
  44. W. Böckler u. a .: pathology. Urban & Fischer Verlag, 2008, ISBN 3-437-42382-7 , p. 211.
  45. ^ AS Khan and S. Malik: Cachectin / tumor necrosis factor. In: J Pak Med Assoc 46, 1996, pp. 137-140, PMID 8991374 (Review).
  46. MD Barber et al. a .: Disordered Metabolic Response with Cancer and its Management. In: World Journal of Surgery 24, 2000, 24, pp. 681-689, PMID 10773120 .
  47. A. Roessner: General pathology and foundations of special pathology. Urban & Fischer Verlag, 2008, ISBN 3-437-41541-7 , p. 253.
  48. ^ H. Dreger: Suppression of the hypertrophy of cardiac myocytes by inhibition of the ubiquitin-proteasome system. Dissertation, Medical Faculty Charité of the Humboldt University Berlin, 2003.
  49. a b c M. J. Tisdale: Catabolic mediators of cancer cachexia. In: Curr Opin Support Palliat Care 2, 2008, pp. 256-261, PMID 19069310 (review).
  50. a b M. Stroud: Thalidomide and cancer cachexia: old problem, new hope? In: Gut 54, 2005, pp. 447-448, PMID 15753523 (Review).
  51. EJ Ramos et al. a .: Cancer anorexia-cachexia syndrome: cytokines and neuropeptides. In: Curr Opin Clin Nutr Metab Care 7, 2004, pp. 427-434, PMID 15192446 (review).
  52. M. Maltoni et al. a .: Serum levels of tumor necrosis factor alpha and other cytokines do not correlate with weight loss and anorexia in cancer patients. In: Support Care Cancer 5, 1997, pp. 130-135, PMID 9069613 .
  53. a b M. P. Thompson et al. a .: Increased expression of the mRNA for the hormone-sensitive lipase in adipose tissue of cancer patients. In: Biochim Biophys Acta 1180, 1993, pp. 236-242, PMID 8422428 .
  54. CJ Horvath et al. a .: Effect of simian immunodeficiency virus infection on tumor necrosis factor-a production by alveolar macrophages. In: Lab Invest 65, 1991, pp. 280-286, PMID 1890808 .
  55. A. Waage u. a .: Detection of tumor necrosis factor-like cytotoxicity in serum from patients with septicemia but not from untreated cancer patients. In: Scand J Immunol 24, 1986, pp. 739-743, PMID 3798026 .
  56. ^ MJ Tisdale: Proteolysis-Inducing Factor in Cancer Cachexia. In: Cachexia and Wasting: A Modern Approach. Verlag Springer, 2006, pp. 483-488, doi: 10.1007 / 978-88-470-0552-5 .
  57. ^ MJ Tisdale: Biology of cachexia. In: J Natl Cancer Inst 89, 1997, pp. 1763-1773, PMID 9392617 (Review).
  58. a b c d M. J. Tisdale: Molecular pathways leading to cancer cachexia. In: Physiology 20, 2005, pp. 340-348, PMID 16174873 (review).
  59. a b P. T. Todorov and a .: Purification and characterization of a tumor lipid-mobilizing factor. In: Cancer Res 58, 1998, pp. 2353-2358, PMID 9622074 .
  60. a b T. M. McDevitt et al. a .: Purification and characterization of a lipid-mobilizing factor associated with cachexia-inducing tumors in mice and humans. In: Cancer Res 55, 1995, pp. 1458-1463, PMID 7882353 .
  61. B. Fröhlich u. a .: Influence of lipid-mobilizing factor (LMF) on the development of tumor cachexia in vivo. In: Z Gastroenterol 43, 2005, pp. 227-228.
  62. E. Aulbert: Textbook of Palliative Medicine. Verlag Schattauer, 2008, ISBN 3-7945-2361-X , p. 309.
  63. ^ S. Khan and MJ Tisdale: Catabolism of adipose tissue by a tumor-produced lipid-mobilizing factor. In: Int J Cancer 80, 1999, pp. 444-447, PMID 9935188 .
  64. ^ A. Laviano: Lipid Mobilizing Factor in Cancer Cachexi. In: Cachexia and Wasting: A Modern Approach. Verlag Springer, 2006, pp. 489-493, doi: 10.1007 / 978-88-470-0552-5 .
  65. C. Bing et al. a .: Zinc-alpha2-glycoprotein, a lipid mobilizing factor, is expressed in adipocytes and is up-regulated in mice with cancer cachexia. In: PNAS 101, 2004, pp. 2500-2505, PMID 14983038 .
  66. PT Todorov et al. a .: Characterization of a cancer cachectic factor. In: Nature 379, 1996, pp. 739-742, PMID 8602222 .
  67. PT Todorov et al. a .: Structural analysis of a tumor-produced sulfated glycoprotein capable of initiating muscle protein degradation. In: J Biol Chem 272, 1997, pp. 12279-12288, PMID 9139670 .
  68. PT Todorov et al. a .: Induction of muscle protein degradation and weight loss by a tumor product. In: Cancer Res 56, 1996, pp. 1256-1261, PMID 8640810 .
  69. ML Williams et al. a .: The relationship between a urinary cachectic factor and weight loss in advanced cancer patients. In: Cancer Invest 22, 2004, pp. 866-870, PMID 15641484 .
  70. R. Cabal-Manzano et al. a .: Proteolysis-inducing factor is expressed in tumors of patients with gastrointestinal cancers and correlates with weight loss. In: Br J Cancer 84, 2001, pp. 1599-1601, PMID 11401311 .
  71. PT Todorov et al. a .: Role of a proteolysis-inducing factor (PIF) in cachexia induced by a human melanoma (G361). In: Br J Cancer 80, 1999, pp. 1734-1737, PMID 10468289 .
  72. SM Wyke and MJ Tisdale: NF-kappaB mediates proteolysis inducing factor induced protein degradation and expression of the ubiquitin-proteasome system in skeletalmuscle. In: Br J Cancer 92, 2005, pp. 711-721, PMID 15714207 .
  73. MJ Lorite et al. a .: Mechanism of muscle protein degradation induced by a cancer cachectic factor. In: Br J Cancer 78, 1998, pp. 850-856, PMID 9764574 .
  74. MJ Lorite et al. a .: Induction of muscle protein degradation by a tumor factor. In: Br J Cancer 76, 1997, pp. 1035-1040, PMID 9376263 .
  75. P. Cariuk et al. a .: Induction of cachexia in mice by a product isolated from the urine of cachectic cancer patients. In: Br J Cancer 76, 1997, pp. 606-613, PMID 9303359 .
  76. PT Todorov et al. a .: Identification and characterization of a membrane receptor for proteolysis-inducing factor on skeletal muscle. In: Cancer Res 67, 2007, pp. 11419-11427, PMID 18056470 .
  77. N. Teich u. a .: The presence of the proteolysis-inducing factor in urine does not predict the malignancy of a pancreatic tumor. In: BMC Gastroenterol 5, 2005, p. 20, PMID 15969757 .
  78. J. Ockenga et al. a .: tumor anorexia - tumor cachexia in gastroenterology: standards and visions. In: Z Gastroenterol 40, 2002, pp. 929-936, doi: 10.1055 / s-2002-35411 .
  79. BM Wieland: Is there a human homologue to the murine proteolysis-inducing factor? In: Clin Cancer Res 13, 2007, pp. 4984-4992, PMID 17785548 .
  80. ^ MJ Tisdale: Cancer anorexia and cachexia. In: Nutrition 17, 2001, pp. 438-442, PMID 11377146 (Review).
  81. a b c S. Priepke: Food preferences in patients with gastrointestinal tumors. Dissertation, Humboldt University Berlin, Charité University Hospital, 2006.
  82. G. Mantovani et al. a .: Serum values ​​of proinflammatory cytokines are inversely correlated with serum leptin levels in patients with advanced stage cancer at different sites. In: J Mol Med 79, 2001, pp. 406-414, PMID 11466563 .
  83. G. Mantovani et al. a .: Serum Levels of Leptin and Proinflammatory Cytokines in Patients with advanced-stage Cancer at different sites. In: J Mol Med 78, 2000, pp. 554-561, PMID 11199328 .
  84. ^ F. Rossi Fanelli and A. Laviano: Cancer anorexia: a model for the understanding and treatment of secondary anorexia. In: International Journal of Cardiology 85, 2002, pp. 67-72, PMID 12163210 .
  85. ^ S. Forbes et al. a .: Integrated control of appetite and fat metabolism by the leptin-proopiomelanocortin pathway. In: PNAS 98, 2001, pp. 4233-4237, PMID 11259669 .
  86. ^ A b D. Heber and N. Tchekmedyian: Pathophysiology of cancer: hormonal and metabolic abnormalities. In: Oncology 49, 1992, pp. 28-31, PMID 1461623 (Review).
  87. K. Nelson: The Cancer-Anorexia-Cachexia Syndrome. In: Seminars in Oncology 27, 2000, pp. 64-69, PMID 10697022 .
  88. ^ O. Selberg: Causes and characteristics of the tumor cachexia perspectives of a nutritional medical treatment. In: Ernährungsumschau 47, 2000, pp 298-303.
  89. a b C. L. Loprinzi u. a .: Body composition changes in patients who gain weight while receiving megestrol acetate. In: J Clin Oncol 11, 1993, pp. 152-154, PMID 8418227 .
  90. R. Patarca-Montero: Handbook of cancer-related fatigue. Haworth Press Inc, 2004, ISBN 0-7890-2168-4 , p. 106.
  91. a b J. M. Arbeit u. a .: Resting energy expenditure in controls and cancer patients with localized and diffuse disease. In: Ann Surg 199, 1984, pp. 292-298, PMID 6703790 .
  92. ^ A. Theologides: Cancer cachexia. In: Cancer 43, 1979, pp. 2004-2012, PMID 376104 (review).
  93. ^ H. Munro: Tumor-host competition for nutrients in the cancer patient. In: J Am Diet Assoc 71, 1977, pp. 380-384, PMID 908798 .
  94. DT Dempsey et al. a .: Energy expenditure in malnourished gastrointestinal cancer patients. In: Cancer 53, 1984, pp. 1265-1273, PMID 6692317 .
  95. ^ DM Russell et al. a .: Effects of total parenteral nutrition and chemotherapy on the metabolic derangements in small cell lung cancer. In: Cancer Res 44, 1984, pp. 1706-1711, PMID 6322985 .
  96. I. Warnold u. a .: Energy balance and body composition in cancer patients. In: Cancer Res 38, 1978, pp. 1801-1807, PMID 647689 .
  97. DT Hansell et al. a .: The relationship between resting energy expenditure and weight loss in benign and malignant disease. In: Ann Surg 203, 1986, pp. 240-245, PMID 3082302 .
  98. CP Holroyde u. a .: Altered glucose metabolism in metastatic carcinoma. In: Cancer Res 35, 1975, pp. 3710-3714, PMID 1192429 .
  99. M. Lainscak et al. a .: Cachexia: common, deadly, with an urgent need for precise definition and new therapies. In: The American Journal of Cardiology 101, 2008, pp. 8-10, PMID 18514632 .
  100. P. Geels et al. a .: Palliative effect of chemotherapy: objective tumor response is associated with symptom improvement in patients with metastatic breast cancer. In: J Clin Oncol 18, 2000, pp. 2395-2405, PMID 10856099 .
  101. M. Muscaritoli: Therapy of muscle wasting in cancer: what is the future? In: Curr Opin Clin Nutr Metab Care 7, 2004, pp. 459-466, PMID 15192450 .
  102. ^ A b c G. Mantovani, C. Madeddu: Cancer cachexia: medical management. In: Support Care Cancer [electronic publication before printing], 2009, PMID 19688225 .
  103. G. Mantovani et al. a .: Randomized phase III clinical trial of five different arms of treatment for patients with cancer cachexia: interim results. In: Nutrition 24, 2008, pp. 305-313, PMID 18262758 .
  104. a b c d e f M. Bossola u. a .: Novel treatments for cancer cachexia. In: Expert Opin Investig Drugs 16, 2007, pp. 1241-1253. (Review).
  105. a b Z. H. Khan u. a .: Oesophageal cancer and cachexia: the effect of short-term treatment with thalidomide on weight loss and lean body mass. In: Aliment Pharmacol Ther 17, 2003, pp. 677-682, PMID 12641516 .
  106. T. Hanada et al. a .: Anti-cachectic effect of ghrelin in nude mice bearing human melanoma cells . In: Biochem Biophys Res Commun 301, 2003, pp. 275-279, PMID 12565855 .
  107. a b W. Wang u. a .: Effects of ghrelin on anorexia in tumor-bearing mice with eicosanoid-related cachexia. In: Int J Oncol 28, 2006, pp. 1393-1400, PMID 16685441 .
  108. MD DeBoer: Emergence of ghrelin as a treatment for cachexia syndromes. In: Nutrition 24, 2008, pp. 806-814, PMID 18725076 (review).
  109. MD DeBoer u. a .: Ghrelin treatment causes increased food intake and retention of lean body mass in a rat model of cancer cachexia. In: Endocrinology 148, 2007, pp. 3004-3012, PMID 17347304 .
  110. YL Liu et al. a .: Ghrelin alleviates cancer chemotherapy-associated dyspepsia in rodents. In: Cancer Chemother Pharmacol 58, 2006, pp. 326-333, PMID 16435157 .
  111. JA Rudd u. a .: Anti-emetic activity of ghrelin in ferrets exposed to the cytotoxic anti-cancer agent cisplatin. In: Neurosci Lett 392, 2006, pp. 79-83, PMID 16182445 .
  112. a b F. Strasser u. a .: Safety, tolerability and pharmacokinetics of intravenous ghrelin for cancer-related anorexia / cachexia: a randomized, placebo-controlled, double-blind, double-crossover study. In: British Journal of Cancer 98, 2008, pp. 300-308, PMID 18182992 .
  113. JM Garcia et al. a .: Active ghrelin levels and active to total ghrelin ratio in cancer-induced cachexia. ( Memento of the original from December 29, 2010 in the Internet Archive ) Info: The archive link was inserted automatically and has not yet been checked. Please check the original and archive link according to the instructions and then remove this notice. In: J Clin Endocrinol Metab 90, 2005, pp. 2920-2926, PMID 15713718 . @1@ 2Template: Webachiv / IABot / jcem.endojournals.org
  114. WW Cheung and RH Mak: Ghrelin and its analogues as therapeutic agents for anorexia and cachexia in end-stage renal disease. In: Kidney Int 76, 2009, pp. 135-137, PMID 19564855 .
  115. TA Splinter: cachexia and cancer: a clinician's view. In: Ann Oncol 3, 1992, pp. 25-27, PMID 1382553 .
  116. AP Lopez et al. a .: Systematic review of megestrol acetate in the treatment of anorexia-cachexia syndrome. In: J Pain Sympt Manag 27, 2004, pp. 360-369, PMID 15050664 (Review).
  117. ^ W. Leśniak et al. a .: Effects of megestrol acetate in patients with cancer anorexia-cachexia syndrome - a systematic review and meta-analysis. In: Pol Arch Med Wewn 118, 2008, pp. 636-644, PMID 19140567 (review).
  118. T. Yavuzsen et al. a .: Systematic review of the treatment of cancer-associated anorexia and weight loss. In: J Clin Oncol 23, 2005, pp. 8500-8511, PMID 16293879 (review).
  119. ^ R. Mechoulam (Editor): Cannabinoids as Therapeutic Agents. CRC Press, 1984. ISBN 0-8493-5772-1 .
  120. ^ E. Fride: Cannabinoids and feeding: the role of the endogenous cannabinoid system as a trigger for newborn suckling. In: J Cannabis Ther 2, 2002, pp. 51-62.
  121. E. Fride et al. a .: Endocannabinoids and food intake: newborn suckling and appetite regulation. In: Adulthood Exp Biol Med 230, 2005, pp. 225-234, PMID 15792943 (Review).
  122. EM Berry and R. Mechoulam: Tetrahydrocannabinol and endocannabinoids in feeding and appetite. In: Pharmacol Ther 95, 2002, pp 185-190, PMID 12182965 (review).
  123. F. Strasser et al. a .: Comparison of orally administered cannabis extract and delata-9-tetrahydrocannabinol in treating patients with cancer-related anorexia-cachexia syndrome. A multicenter, phase III, randomized, double-blind, placebo-controlled clinical trial from the Cannabis-In-Cachexia-Study-Group. In: J Clin Oncol 24, 2006, pp. 3394-3400, PMID 16849753 .
  124. A. Jatoi et al. a .: Dronabinol versus combination therapy for cancer-associated anorexia: a North Central Cancer Treatment study. In: J Clin Oncol 20, 2002, pp. 567-573, PMID 11786587 .
  125. MJ Tisdale: Clinical anticachexia treatments. In: Nutr Clin Pract 21, 2006, pp. 168-174, PMID 16556927 (Review).
  126. JR Nicholson et al. a .: Peripheral administration of a melanocortin 4-receptor inverse agonist prevents loss of lean body mass in tumor-bearing mice. In: J Pharmacol Exp Ther 317, 2006, pp. 771-777, PMID 16436498 .
  127. ^ DO McCarthy et al. a .: Indomethacin and ibuprofen preserve gastrocnemius muscle mass in mice bearing the colon-26 adenocarcinoma. In: Res Nurs Health 27, 2004, pp. 174-184, PMID 15141370 .
  128. M. Trikha et al. a .: Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: a review of the rational and clinical evidence. In: Clin Cancer Res 9, 2003, pp. 4653-4665, PMID 14581334 .
  129. a b K. Lundholm u. a .: Anti-inflammatory treatment may prolong survival in undernourished patients with metastatic solid tumors. In: Cancer Res 54, 1994, pp. 5602-5606, PMID 7923204 .
  130. a b S. von Haehling u. a .: Cachexia: a therapeutic approach beyond cytokine antagonism. In: Int Jour Of Cardiology 85, 2002, pp. 173-183, PMID 12163222 (Review).
  131. V. Eleutherakis-Papaiakovou u. a .: Thalidomide in cancer medicine. In: Ann Oncol 15,2004, pp. 1151-1160, PMID15277253 (Review).
  132. E. Bruera et al. a .: Thalidomide in patients with cachexia due to terminal cancer: preliminary report. In: Ann Oncol 10, 1999, pp. 857-859, PMID 10470435
  133. E. Esposito and S. Cuzzocrea: TNF-alpha as a therapeutic target in inflammatory diseases, ischemia-reperfusion injury and trauma. In: Curr Med Chem 16, 2009, pp. 3152-3167, PMID 19689289 .
  134. a b K. H. Liu u. a .: Thalidomide attenuates tumor growth and preserves fast-twitch skeletal muscle fibers in cholangiocarcinoma rats. In: Surgery 143, 2008, pp. 375-383, PMID 18291259 .
  135. D. Tassinari et al. a .: Thalidomide in the treatment of cancer cachexia. In: J Palliat Care 24, 2008, pp. 187-189, PMID 18942570 .
  136. JN Gordon et al. a .: Thalidomide in the treatment of cancer cachexia: a randomized placebo controlled trial. In: Gut 54, 2005, pp. 540-545, PMID 15753541 .
  137. ^ MJ Tisdale: The ubiquitin-proteasome pathway as a therapeutic target for muscle wasting. In: J Support Oncol 3, 2005, pp. 209-217, PMID 15915823 (review).
  138. a b c d G. J. Wilson et al. a .: Effects of beta-hydroxy-beta-methylbutyrate (HMB) on exercise performance and body composition across varying levels of age, sex, and training experience: A review. In: Nutr Metab (Lond) 5, 2008, 1, PMID 18173841 (Review).
  139. E. Jowko et al. a .: Creatine and β-hydroxy-β-methylbutyrate (HMB) additively increase lean body mass and muscle strength during a weight-training program. In: Nutr 17, 2001, pp. 558-566, doi: 10.1016 / S0899-9007 (01) 00540-8 PMID 11448573 .
  140. AE Knitter et al. a .: Effects of β-hydroxy-β-methylbutyrate on muscle damage after a prolonged run. In: J Appl Physiol 89, 2000, pp. 1340-1344, PMID 11007567 .
  141. ^ PM Gallagher et al. a .: β-hydroxy-β-methylbutyrate ingestion, part I: Effects on strength and fat free mass. In: Med Sci Sports Exerc 32, 2000, pp. 2109-2115, doi: 10.1097 / 00005768-200012000-00022 PMID 11128859 .
  142. DM O'Connor and MJ Crowe: Effects of beta-hydroxy-beta-methylbutyrate and creatine monohydrate supplementation on the aerobic and anaerobic capacity of highly trained athletes. In: J Sports Med Phys Fitness 43, 2003, pp. 64-68, PMID 12629464 .
  143. T. Palisin and JJ Stacy: Beta-hydroxy-beta-methylbutyrate and its use in athletics. In: Curr Sports Med Rep 4, 2005, pp. 220-223, PMID 16004832 (review).
  144. ^ MJ Tisdale and SA Beck: Cancer cachexia. In: Int J Pancreatol 7, 1990, pp. 141-150, PMID 2081920 (review).
  145. HJ Smith et al. a .: Attenuation of proteasome-induced proteolysis in skeletal muscle by beta-hydroxy-beta-methylbutyrate in cancer-induced muscle loss. In: Cancer Res 65, 2005, pp. 277-283, PMID 15665304 .
  146. HJ Smith et al. a .: Mechanism of the attenuation of proteolysis-inducing factor stimulated protein degradation in muscle by β-hydroxy-β-methylbutyrate. In: Cancer Res 64, 2004, pp. 8731-8735, PMID 15574784 .
  147. H. L Eley et al. a .: Signaling pathways initiated by beta-hydroxy-beta-methylbutyrate to attenuate the depression of protein synthesis in skeletal muscle in response to cachectic stimuli. In: Am J Physiol Endocrinol Metab 293, 2007, pp. E923-31, PMID 17609254 .
  148. EA Nunes et al. a .: Beta-hydroxy-beta-methylbutyrate supplementation reduces tumor growth and tumor cell proliferation ex vivo and prevents cachexia in Walker 256 tumor-bearing rats by modifying nuclear factor-kappaB expression. In: Nutr Res 28, 2008, pp. 487-493, PMID 19083450 .
  149. HL Eley et al. a .: Attenuation of depression of muscle protein synthesis induced by lipopolysaccharide, tumor necrosis factor, and angiotensin II by beta-hydroxy-beta-methylbutyrate. In: Am J Physiol Endocrinol Metab 295, 2008, pp. E1409-1416, PMID 18854427 .
  150. ^ Studies Which Support the Efficacy of HMB supplementation in Varying Populations. from PMID 18173841 .
  151. ^ Studies Which do not Support the Efficacy of HMB Supplementation in Varying Populations. from PMID 18173841 .
  152. PE May et al. a .: Reversal of cancer-related wasting using oral supplementation with a combination of beta-hydroxy-beta-methylbutyrate, arginine and glutamine. In: Am J Surg 181, 2002, pp. 471-479, PMID 11975938 .
  153. L. Berk: A randomized, double-blind, placebo-controlled trial of a beta-hydroxyl beta-methyl butyrate, glutamine, and arginine mixture for the treatment of cancer cachexia (RTOG 0122). In: Support Care Cancer 16, 2008, pp. 1179-1188, PMID 18293016 .
  154. C. Madeddu, G. Mantovani: An update on promising agents for the treatment of cancer cachexia. In: Curr Opin Support Palliat Care 3, 2009, pp. 258-262 PMID 19667995 (Review).
  155. ^ AS Whitehouse et al. a .: Mechanism of attenuation of skeletal muscle protein catabolism in cancer cachexia by eicosapentaenoic acid. In: Cancer Res 61, 2001, pp. 3604-3609, PMID 11325828 .
  156. MD Barber et al. a .: The effect of an oral nutritional supplement enriched with fish oil on weight-loss in patients with pancreatic cancer. In: Br J Cancer 81, 1999, pp. 80-86, PMID 10487616 .
  157. SJ Wigmore et al. a .: Effect of oral eicosapentaenoic acid on weight loss in patients with pancreatic cancer. In: Nutr Cancer 36, 2000, pp. 177-184, PMID 10890028 .
  158. A. Jatoi et al. a .: An eicosapentaenoic acid supplement versus megestrol acetate versus both in patients with cancer-associated wasting: a North Central Cancer Treatment Group and National Cancer Institute of Canada collaborative efforts. In: J Clin Oncol 22, 2004, pp. 2469-2476, PMID 15197210 .
  159. KC Fearon et al. a .: Double-blind, placebo controlled, randomized study of eicosapentaenoic acid diester in patients with cancer cachexia. In: J Clin Oncol 24, 2006, pp. 3401-3407, PMID 16849754 .
  160. KC Fearon et al. a .: 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 randomized double blind trial. In: Gut 52, 2003, pp. 1479-1486, PMID 12970142 .
  161. C. Persson et al. a .: Impact of fish oil and melatonin on cachexia in patients with advanced gastrointestinal cancer: a randomized pilot study. In: Nutrition 21, 2005, pp. 170-178, PMID 15723745 .
  162. HJ Smith: Effect of eicosapentaenoic acid, protein and amino acids on protein synthesis and degradation in skeletal muscle of cachectic mice. In: Br J Cancer 91, 2004, pp. 408-412, PMID 15213711 .
  163. MJ Heslin et al. a .: Effect of systemic hyperinsulinemia in cancer patients. In: Cancer Res 52, 1992, pp. 3845-3850, PMID 1617658 .
  164. K. Lundholm et al. a .: Insulin Treatment in Cancer Cachexia: Effects on Survival, Metabolism, and Physical Functioning. In: Clinical Cancer Research 13, 2007, pp. 2699-2706, PMID 17473202 .
  165. ^ J. Gold: Anabolic profiles in late-stage cancer patients responsive to hydrazine sulfate. In: Nutr Cancer 3, 1981, pp. 13-19, PMID 7050921 .
  166. B. Grubbs et al. a .: Total parenteral nutrition and inhibition of gluconeogenesis on tumor-host responses. In: Oncology 36, 1979, pp. 216-223, PMID 113715 .
  167. J. Gold: Hydrazine sulfate: a current perspective. In: Nutr Cancer 9, 1987, pp. 59-66, PMID 3104888 (Review).
  168. RT Chlebowski et al. a .: Influence of hydrazine sulfate on abnormal carbohydrate metabolism in cancer patients with weight loss. In: Cancer Res 44, 1984, pp. 857-861, PMID 6692384 .
  169. IL Cameron: Effect of total parenteral nutrition on tumor-host responses in rats. In: Cancer Treat Rep 65, 1981, pp. 93-99, PMID 6809328 .
  170. ^ SJ Lee: Quadrupling Muscle Mass in Mice by Targeting TGF-β Signaling Pathways. In: PLoS ONE 2, 2007, e789, doi: 10.1371 / journal.pone.0000789 , PMID 17726519 .
  171. JM Argiles et al. a .: Cancer cachexia: the molecular mechanisms. In: Int J Biochem Cell Biol 35, 2003, p. 405409, PMID 12565701 (review).
  172. ^ P. Schlag and C. Decker-Baumann: Strategies and needs for nutritional support in cancer surgery. In: Recent Results Cancer Res 121, 1991, pp. 45-52, PMID 1907017 (Review).
  173. F. Dombrowski et al. a .: General pathology . Urban & Fischer Verlag, 2004, ISBN 3-437-21290-7 , p. 19.
This article was added to the list of excellent articles on December 15, 2009 in this version .