Commentary
Cachexia [Greek origin, Etymology: kakos (bad) and hexes (condition)] is a complex metabolic syndrome associated with underlying illness and characterized by loss of muscle mass with or without loss of body fat. This syndrome can be linked to anorexia, inflammation, insulin resistance, and raised protein turnover [1]. Cachexia is typically found to be associated with severe disorders such as chronic heart/renal failure, cancer, and autoimmune diseases. This condition results in deprived nutrition and decreased skeletal muscle mass [2,3]. Cachexia, a prevalent disorder in Western countries, mostly eventuates in cancer patients (30%) [4]. Moreover, the international consensus defines cancer-associated cachexia (CAC) as a complex condition characterized by a continuous loss of muscle mass (sometimes fat), which cannot be reversed entirely with standard nutrition support. It results from a combination of reduced food intake and altered metabolism [5].
Systemic inflammation is a crucial characteristic of CAC. Although several non-steroidal anti-inflammatory drugs (NSAIDs) have been proposed as potential therapy for CAC, none of them have consistently demonstrated efficacy [6]. We conducted a research study published as “R-ketorolac ameliorates cancer-associated cachexia and prolongs survival of tumor-bearing mice," which explored the effects of the R-enantiomer of ketorolac (RK), a non-steroidal anti-inflammatory drug (NSAID), on CAC and survival rates in tumor-bearing mice. CAC is a chronic condition that causes muscle and fat loss, systemic inflammation, and a significant reduction in the quality of life and life expectancy of cancer patients. This study included 10- to 11-week-old mice inoculated with C26 or CHX207 cancer cells to induce CAC or vehicle control (phosphate buffered saline, PBS). The onset of cachexia was confirmed when body weight loss of ≥ 5% was achieved within 2–3 consecutive days (mild cachexia). Upon induction of cachexia, mice were administered either 2 mg/kg of RK or 12 gm/kg of anamorelin/PBS (vehicle) as control daily by oral gavage. The body weight, food intake, and tumor size were monitored throughout the study. Blood samples were withdrawn at the end of the study, and tissues were collected for further analysis. Immune cell abundance, COX activity, muscle tissues, and muscle fiber size were analyzed with a flow cytometer, fluorometric kit, RT-PCR/western blotting, and hematoxylin/eosin staining, respectively.
The result of this study demonstrated significant improvement in survival rates in mice treated with RK compared to untreated mice. The 10-day survival rate for C26 tumor-bearing mice was 10% in untreated mice, whereas it was found to be a 100% survival rate (P = 0.0009) in the RK-treated mice. Similarly, chemotherapy alone led to a 10% survival rate 14 days after treatment initiation. However, when RK and cyclophosphamide were administered together, all mice survived (P = 0.0001). RK-treated tumor-bearing mice demonstrated less body weight loss than untreated tumor-bearing mice. Moreover, a protective effect against muscle and fat wasting was observed in RK-treated tumor-bearing mice. RK was found to significantly reduce systemic levels of interleukin-6 (IL-6), a pro-inflammatory cytokine associated with CAC, in both C26 and CHX207 tumor-bearing mice. Additionally, RK protects against cancer-induced T-lymphopenia, further supporting its role in modulating the immune response in CAC. The beneficial effects of RK in ameliorating CAC appeared to depend on the presence of T-cells, as the treatment was ineffective in thymus-deficient nude mice. The study emphasizes that RK’s beneficial effects operate independently of COX inhibition, setting it apart from other NSAIDs with similar anti-inflammatory functions but possessing significant side effects. The study concludes that RK significantly enhances survival and alleviates CAC-associated symptoms in tumor-bearing mice. RK benefits by reducing systemic IL-6 levels and preventing T-lymphopenia. These findings indicate that RK could be considered a promising therapeutic option in managing CAC, thereby warranting further investigation and potential clinical trials to explore the underlying mechanisms further.
Rethinking CAC: An Immune Disorder Beyond Metabolism
CAC is a multifactorial complex syndrome defined by the loss of skeletal muscle mass, often accompanied by fat loss, which eventually leads to reduced functionality and quality of life. This condition manifests due to elevated systemic inflammation and catabolic signals, suppressing protein synthesis and promoting muscle breakdown [7]. Despite the crucial role of inflammation-driven involvement of immunoregulation in cachexia, the immunological aspect extensively involved in the cachectic progression remains poorly explored [8]. The data focusing on the impact of the immune system on cachexia initiation are currently limited. However, while we do not understand the mechanisms of cachexia fully as yet, it is quite clear that its onset is driven by the inflammatory processes initiated by the underlying illness.
Cancer is characterized by malfunctioning of the immune system and inflammation. This disruption of immune tolerance is a crucial feature of cancer cells, and failure of this homeostasis state may result in autoimmunity. Overproduction of cytokines and autoantibodies against different organs is a common feature of advanced malignancy and autoimmune disorders. These cytokines, overproduced in the tumor microenvironment, interact with muscle cells and result in muscle wasting. Notably, IL-1β, IL-6, and TNF-α cytokines have been associated with CAC (summarized in Table 1) [9].
Cytokine |
Produced by |
Organs affected by cancer |
Model (Trials) |
Effects in cachexia |
Reference |
IL-1α |
Macrophages and endothelial cells |
Pancreas, breast, colorectal |
Mice and rats |
Lipolysis, anorexia, weight loss, suppression of hunger |
[54-56] |
IL-1β |
Macrophages |
Gastric, breast, lung, and several other |
Mice and human |
Anorexia, weight loss, sarcopenia, weakness, impaired mitochondrial metabolism |
[57-59] |
IL6 |
Activated macrophages |
Colorectal, pancreas, and several others |
Mice and human |
Adipose wasting, skeletal muscle loss, affects gut and liver tissue, impaired mitochondrial metabolism |
[55,56,59] |
TNFα |
Activated macrophages, CD4+, neutrophils, mast cells, eosinophils and neurons |
Pancreas, lungs, colon, and several other |
Mice and human |
Anorexia, muscle and adipose wasting, insulin resistance, increased energy expenditure, impaired mitochondrial metabolism |
[60-62] |
GDF15 |
Adipocytes, Macrophages, Endothelial cells, Vascular smooth muscle cells, Cardiomyocytes, and Trophoblastic cells |
Lungs, pancreas, colorectal, prostate, and several others |
Mice |
energy metabolism, anorexia, muscle atrophy, adipose tissue depletion, bone loss, anemia |
[56,63,64] |
IFNγ |
Activated T and NK cells |
Pancreas, lung, colon, prostate |
Mice and rats |
Loss of body weight, reduced appetite, atrophy of adipose tissue |
[56,65,66] |
IL-1α: Interleukin-1 Alpha; IL-1β: Interleukin-1 Beta; IL-6: Interleukin-6; TNFα: Tumour Necrosis Factor Alpha; GDF15: Growth Differentiation Factor 15; INFγ: Interferon Gamma |
Both immune and non-immune cells exhibit abundant expression of these inflammatory mediators once their pattern-recognition receptors are activated by damage-associated molecular patterns and/or pathogen-associated molecular patterns [8,10,11]. Cytokines activate JAK-STAT (Janus kinase/signal transducer and activator of transcription) and NF-κB signaling pathways as well as downstream transcriptional regulation can induce several catabolic pathways in muscles and adipose tissue including mitochondrial dysfunction, a burst of reactive oxygen species (ROS), and pathways like NLRP3 (nucleotide-binding domain, leucine-rich–containing family, pyrin domain–containing-3), JNK (Jun N-terminal kinase), MAPK (mitogen-activated protein kinase), PI3K (phosphatidylinositol-3 kinase) and NF-κB leading to inflammation [8,10,12-14]. According to a study conducted by Lima et al., in patients with colorectal CAC, the tumors exhibited elevated levels of inflammatory cytokines (IFN-α and IL-8), growth factors (such as granulocyte-macrophage colony-stimulating factor and epidermal growth factor), and deposits of actin and collagen [15]. Another study reported elevated levels of growth differentiation factor 15 (GDF15) in several malignant tumors and was found to be associated with cachexia-inducing weight loss [16]. Several cytokines are induced in a context-dependent manner in cachexia and may contribute to its development. However, further research is imperative to elucidate the role of pro and anti-inflammatory cytokines in the pathophysiology of cachexia [5].
Studies on the association between immune cell function and cachexia are limited. However, recent studies have suggested the involvement of multiple immune cell types, such as macrophages, neutrophils, T cells, and myeloid-derived suppressor cells (MDSCs), in the initiation and progression of cachexia [5]. The pancreatic ductal adenocarcinoma (PDAC) mouse model of CAC demonstrated that the increased number of microglial cells in the brain caused the depletion of neutrophils (the number of neutrophils is directly proportional to the degree of cachexia), thereby giving protection against severe cachexia [17]. Further, macrophages are involved in protection against lipolysis in adipose tissue, whereas in muscle tissue, an increased number of macrophages leads to a higher degree of atrophy, which is primarily affected by polarization of macrophages [18,19]. The number of MDSCs in the spleen and bone marrow positively correlates with cachexia [20,21]. A negative correlation was observed between the abundance of CD4+FOXP3+ T cells and cachexia, while CD8+ T cells exhibited a negative correlation with cachexia [22,23].
Immunometabolism plays a pivotal role in cachexia, as the condition depletes the body’s energy reserves, thereby potentially triggering systemic inflammation, immune cell activation, and metabolic disturbances [5,8]. Cancer cells can induce metabolic reprogramming of immune cells, resulting in systemic changes in metabolism, further inducing the transition from pro-inflammatory to immunosuppressive responses [24,25]. Recent studies suggest that activated immune cells mediate the activity of several signaling molecules, such as p38 MAPK, STAT3, etc., resulting in the promotion of cancer cachexia [26-30]. Several preclinical studies have suggested overexpression of cytokines (IL-6 and IL8), increased NLR, and changes in the TGFβ pathway during CAC progression, indicating that immune disorders are associated with CAC [9,31-34].
Research shows that immune responses in cachexia usually trigger tissue catabolism. Immunometabolic crosstalk during cachexia induces adipose tissue and muscle catabolism, producing fatty acids and amino acids, respectively [5]. Adipose tissue lipolysis increases levels of NEFAs (nonesterified fatty acids) and glycerol, which are utilized by other organs [35,36]. In vitro studies have shown that treating adipocyte and hepatocyte cell lines with saturated fatty acids activates Toll-like receptor 4 (TLR4) signaling, further activating NF-κB and resulting in increased TNF and IL-6 production [37]. The studies suggest that adipose tissue lipolysis and NEFA release carry out activation of TLR4, further aggravating tissue catabolism [38,39]. Further, alterations in amino acid profiles have also been observed in the tumor microenvironment. Immune cells and tumor cells compete for available amino acids to support their high anabolic demands, which modulates the activation status of immune cells [40]. For example, glutamine is primarily taken up by tumor cells, limiting its availability for T cells [41]. Comprehensive profiling and dissection of systemic and local amino acid levels in cachexia will contribute to a better understanding of their pathophysiological effect on inflammation, metabolism, and disease outcomes [5].
As stated earlier, cachexia is characterized by muscle atrophy and adipose tissue depletion. The depletion of skeletal and cardiac muscle mass is due to three cellular processes downstream of FOXO (forkhead box O) and NF-κB (nuclear factor kappa B) signaling, viz., proteasomal degradation, autophagy, and reduced myocyte regeneration [42-45]. Cachexia is responsible for increased lipolysis with suppression of liquid uptake and lipogenesis causing depletion of lipid droplets of adipose tissue [46,47]. The changes in the adipose tissue are known to occur through either transcriptional modulation or ATGL-mediated and HSL-mediated lipolysis [47].
The intricate link between immune responses and cachexia remains unexplored and yet to be unraveled. Therefore, future studies on cachexia should integrate immunology and the use of tools and knowledge from evolutionary medicine and systems biology [48]. Moreover, clinical studies encompassing both animal models and human trials are also crucial for further validation. Immunomodulation in cachexia, specifically in the context of CAC, has been largely overlooked in research. Therefore, more rigorous studies on immune cell function will substantially enhance our understanding of the role of immunomodulation in tissue metabolism during cachexia [5]. Though numerous pre-clinical studies have indicated that tumor-induced immune disorder promotes the development of CAC, direct evidence demonstrating immunomodulation to the onset of CAC symptoms is still lacking [9]. However, research indicates that inhibiting cytokines and immunomodulation can reduce CAC progression, suggesting that these pathways may be considered an effective strategy for treating CAC [9].
Similarly, our study revealed that RK-mediated immunomodulation in a mouse model improved T-lymphopenia and decreased systemic IL-6 concentrations, thereby ameliorating cachexia and increasing the survival rate in cachexigenic tumor-bearing mice undergoing chemotherapy. Hence, our study highlights and emphasizes the benefit of RK in patients suffering from CAC [49].
Immune Disorder may be the Root Cause of Cancer Death
Cancer cachexia, generally an overlooked condition in cancer patients, is often associated with immune system disturbances. In response to cancer, immune cells release inflammatory molecules in CAC, including interleukins, members of the growth factor family, and tumor necrosis factor [50]. Particularly in cancer cachexia, substances secreted by the tumor and tumor-induced immune responses followed by an altered metabolic state have been suggested to be deeply involved in its pathogenesis [30]. The study by Riccardi et al., compared plasma profiles of CAC and weight-stable cancer (WSC) patients. The study demonstrated alterations in plasma concentrations of markers of systemic inflammation in cancer cachexia patients. Higher levels of pro-inflammatory cytokines like IL-6, tumor necrosis factor-alpha (TNF-α), and IL-8 were reported [51]. In another study by Mangano et al. [52], TNF-α directly leads to muscle wasting by activating the ubiquitin-proteasome system. In cancer cachexia, it disrupts carbohydrate, protein, and fat metabolism. TNF-α also increases gluconeogenesis, breaks down fat and proteins, and reduces the synthesis of proteins, lipids, and glycogen. Another study by Shukla et al. [19], highlighted the role of macrophages in muscle wasting in preclinical trials. It demonstrated that reducing macrophages lessened systemic inflammation and muscle wasting in mice with pancreatic tumors. Inhibiting macrophage-driven STAT3 activation or macrophage-derived interleukin-1 alpha or interleukin-6 reduced muscle fiber atrophy.
Pre-clinical and clinical studies have confirmed increased levels of proinflammatory cytokines like IL-6, IL-8, and TNFα, as well as immune markers (NLR) in patients with CAC. Hence, immunomodulatory mechanisms involved in cachexia may impact the anti-neoplastic immune response [9]. Immunomodulation could be a potential anti-cancer therapy to constrain CAC mechanisms. RK, a new immunomodulator, was found to reverse the cachexia weight loss and substantially elevated the survival rate in CAC mice. This immunomodulator opens new avenues for research in immunomodulation and may hold an answer to long pending questions, i.e., why do patients with cancer die? [53]. Our study provides an answer that late-stage cancer patients could die from immune disorder-induced implications like cachexia, multiple organ failure, etc. This study revealed that RK-mediated immunomodulation can significantly reverse cancer cachexia and enhance the survival rate. Hence, the study proposes using RK in treating patients suffering from CAC [49]. If the role of this new immunomodulator, i.e., RK, holds true in clinical trials, it would change the paradigm of late-stage cancer care. Our ongoing trials (NCT05336266) support the role of RK in treating patients with CAC.
References
2. Baba MR, Buch SA. Revisiting Cancer Cachexia: Pathogenesis, Diagnosis, and Current Treatment Approaches. Asia Pac J Oncol Nurs. 2021 Aug 27;8(5):508-18.
3. Nishikawa H, Goto M, Fukunishi S, Asai A, Nishiguchi S, Higuchi K. Cancer cachexia: Its mechanism and clinical significance. Int J Mol Sci. 2021 Aug 6;22(16):8491.
4. Anker MS, Holcomb R, Muscaritoli M, von Haehling S, Haverkamp W, Jatoi A, et al. Orphan disease status of cancer cachexia in the USA and the European Union: a systematic review. J Cachexia Sarcopenia Muscle. 2019 Feb 1;10(1):22-34.
5. Baazim H, Antonio-Herrera L, Bergthaler A. The interplay of immunology and cachexia in infection and cancer. Nat Rev Immunol. 2022 May;22(5):309-21.
6. Solheim TS, Fearon KC, Blum D, Kaasa S. Non-steroidal anti-inflammatory treatment in cancer cachexia: a systematic literature review. Acta Oncol. 2013 Jan;52(1):6-17.
7. Setiawan T, Sari IN, Wijaya YT, Julianto NM, Muhammad JA, Lee H, et al. Cancer cachexia: molecular mechanisms and treatment strategies. J Hematol Oncol. 2023 May 22;16(1):54.
8. Wu Q, Liu Z, Li B, Liu YE, Wang P. Immunoregulation in cancer-associated cachexia. J Adv Res. 2024 Apr;58:45-62.
9. Zhang L, Bonomi PD. Immune System Disorder and Cancer-Associated Cachexia. Cancers (Basel). 2024 Apr 27;16(9):1709.
10. Man SM, Jenkins BJ. Context-dependent functions of pattern recognition receptors in cancer. Nat Rev Cancer. 2022 Jul;22(7):397-413.
11. Wu Q, Zhang H, Sun S, Wang L, Sun S. Extracellular vesicles and immunogenic stress in cancer. Cell Death Dis. 2021 Oct 1;12(10):894.
12. Li C, Wu Q, Li Z, Wang Z, Tu Y, Chen C, et al. Exosomal microRNAs in cancer-related sarcopenia: Tumor-derived exosomal microRNAs in muscle atrophy. Exp Biol Med (Maywood). 2021 May;246(10):1156-66.
13. Wu QL, Zhao Y, Zhang YN, Yang Y. High sensitive applied load measurement using optical fiber tapered-loop probe with SPR effect. Opt Laser Technol. 2019;114:95-102.
14. Wu Q, Li B, Li J, Sun S, Yuan J, Sun S. Cancer-associated adipocytes as immunomodulators in cancer. Biomark Res. 2021 Jan 7;9(1):2.
15. Lima JDCC, Simoes E, de Castro G, Morais MRPT, de Matos-Neto EM, Alves MJ, et al. Tumour-derived transforming growth factor-β signalling contributes to fibrosis in patients with cancer cachexia. J Cachexia Sarcopenia Muscle. 2019 Oct 1;10(5):1045-59.
16. Siddiqui JA, Pothuraju R, Khan P, Sharma G, Muniyan S, Seshacharyulu P, et al. Pathophysiological role of growth differentiation factor 15 (GDF15) in obesity, cancer, and cachexia. Cytokine Growth Factor Rev. 2022 Apr;64:71-83.
17. Burfeind KG, Zhu X, Norgard MA, Levasseur PR, Huisman C, Michaelis KA, et al. Microglia in the hypothalamus respond to tumor-derived factors and are protective against cachexia during pancreatic cancer. Glia. 2020 Jul 1;68(7):1479-94.
18. Erdem M, Möckel D, Jumpertz S, John C, Fragoulis A, Rudolph I, et al. Macrophages protect against loss of adipose tissue during cancer cachexia. J Cachexia Sarcopenia Muscle. 2019 Oct 1;10(5):1128-42.
19. Shukla SK, Markov SD, Attri KS, Vernucci E, King RJ, Dasgupta A, et al. Macrophages potentiate STAT3 signaling in skeletal muscles and regulate pancreatic cancer cachexia. Cancer Lett. 2020 Aug 1;484:29-39.
20. Cuenca AG, Cuenca AL, Winfield RD, Joiner DN, Gentile L, Delano MJ, et al. Novel Role for Tumor-Induced Expansion of Myeloid-Derived Cells in Cancer Cachexia. The Journal of Immunology. 2014 Jun 15;192(12):6111-9.
21. Khaled YS, Ammori BJ, Elkord E. Increased levels of granulocytic myeloid-derived suppressor cells in peripheral blood and tumor tissue of pancreatic cancer patients. J Immunol Res. 2014:879897.
22. Narsale A, Moya R, Ma J, Anderson LJ, Wu D, Garcia JM, et al. Cancer-driven changes link T cell frequency to muscle strength in people with cancer: a pilot study. J Cachexia Sarcopenia Muscle. 2019 Aug;10(4):827-43.
23. Olguín JE, Fernández J, Salinas N, Juárez I, Rodriguez-Sosa M, Campuzano J, et al. Adoptive transfer of CD4+Foxp3+ regulatory T cells to C57BL/6J mice during acute infection with Toxoplasma gondii down modulates the exacerbated Th1 immune response. Microbes Infect. 2015 Aug;17(8):586-95.
24. Biswas SK. Metabolic Reprogramming of Immune Cells in Cancer Progression. Immunity. 2015 Sep 15;43(3):435-49.
25. Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Vol. 21, Nat Rev Cancer. 2021 Aug;21(8):481-99.
26. Ding H, Zhang G, Sin KWT, Liu Z, Lin RK, Li M, et al. Activin A induces skeletal muscle catabolism via p38β mitogen-activated protein kinase. J Cachexia Sarcopenia Muscle. 2017 Apr 1;8(2):202-12.
27. Li Y, Schwartz RJ, Waddell ID, Holloway BR, Reid MB. Skeletal muscle myocytes undergo protein loss and reactive oxygen‐mediated NF‐κB activation in response to tumor necrosis factorα. The FASEB Journal. 1998 Jul;12(10):871-80.
28. LI YP, LECKER SH, CHEN Y, WADDELL ID, GOLDBERG AL, REID MB. TNF‐α increases ubiquitin‐conjugating activity in skeletal muscle by up‐regulating UbcH2/E2 20k. The FASEB Journal. 2003 Jun;17(9):1048-57.
29. Li YP, Chen Y, John J, Moylan J, Jin B, Mann DL, et al. TNF‐α acts via p38 MAPK to stimulate expression of the ubiquitin ligase atrogin1/MAFbx in skeletal muscle. The FASEB Journal. 2005 Mar;19(3):362-70.
30. Rausch V, Sala V, Penna F, Porporato PE, Ghigo A. Understanding the common mechanisms of heart and skeletal muscle wasting in cancer cachexia. Oncogenesis. 2021 Jan 8;10(1):1.
31. Alves MJ, Figuerêdo RG, Azevedo FF, Cavallaro DA, Neto NIP, Lima JDC, et al. Adipose tissue fibrosis in human cancer cachexia: The role of TGFβ pathway. BMC Cancer. 2017 Mar 14; 17(1):190.
32. Han J, Lu C, Meng Q, Halim A, Yean TJ, Wu G. Plasma concentration of interleukin-6 was upregulated in cancer cachexia patients and was positively correlated with plasma free fatty acid in female patients. Nutr Metab (Lond). 2019 Nov 15;16:80.
33. Jesus J de CR de, Murari AS de P, Radloff K, Moraes RCM de, Figuerêdo RG, Pessoa AFM, et al. Activation of the Adipose Tissue NLRP3 Inflammasome Pathway in Cancer Cachexia. Front Immunol. 2021 Sep 23;12:729182.
34. Zhang Q, Song MM, Zhang X, Ding JS, Ruan GT, Zhang XW, et al. Association of systemic inflammation with survival in patients with cancer cachexia: results from a multicentre cohort study. J Cachexia Sarcopenia Muscle. 2021 Dec;12(6):1466-76.
35. Fukawa T, Yan-Jiang BC, Min-Wen JC, Jun-Hao ET, Huang D, Qian CN, et al. Excessive fatty acid oxidation induces muscle atrophy in cancer cachexia. Nat Med. 2016 Jun 1;22(6):666-71.
36. Liu KJ, Drucker Y, Jarad J. Hepatic glycerol metabolism in tumorous rats: a 13C nuclear magnetic resonance study. Cancer Res. 1995 Feb 15;55(4):761-6.
37. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest. 2006 Nov;116(11):3015-25.
38. Henriques F, Lopes MA, Franco FO, Knobl P, Santos KB, Bueno LL, et al. Toll-Like Receptor-4 Disruption Suppresses Adipose Tissue Remodeling and Increases Survival in Cancer Cachexia Syndrome. Sci Rep. 2018 Dec 21;8(1):18024.
39. Zhang G, Liu Z, Ding H, Miao H, Garcia JM, Li YP. Toll-like receptor 4 mediates Lewis lung carcinoma-induced muscle wasting via coordinate activation of protein degradation pathways. Sci Rep. 2017 May 23;7(1):2273.
40. Lemos H, Huang L, Prendergast GC, Mellor AL. Immune control by amino acid catabolism during tumorigenesis and therapy. Nat Rev Cancer. 2019 Mar;19(3):162-175.
41. Hirayama A, Kami K, Sugimoto M, Sugawara M, Toki N, Onozuka H, et al. Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 2009 Jun 1;69(11):4918-25.
42. Aversa Z, Pin F, Lucia S, Penna F, Verzaro R, Fazi M, et al. Autophagy is induced in the skeletal muscle of cachectic cancer patients. Sci Rep. 2016 Jul 27;6:30340.
43. Bossola M, Marzetti E, Rosa F, Pacelli F. Skeletal muscle regeneration in cancer cachexia. Clin Exp Pharmacol Physiol. 2016 May 1;43(5):522-7.
44. Penna F, Ballarò R, Martinez-Cristobal P, Sala D, Sebastian D, Busquets S, et al.Autophagy Exacerbates Muscle Wasting in Cancer Cachexia and Impairs Mitochondrial Function. J Mol Biol. 2019 Jul 12;431(15):2674-86.
45. Sandri M. Protein breakdown in cancer cachexia. Semin Cell Dev Biol. 2016 Jun;54:11-9.
46. Sun X, Feng X, Wu X, Lu Y, Chen K, Ye Y. Fat Wasting Is Damaging: Role of Adipose Tissue in Cancer-Associated Cachexia. Front Cell Dev Biol. 2020 Feb 12;8:33.
47. Zechner R, Zimmermann R, Eichmann TO, Kohlwein SD, Haemmerle G, Lass A, et al. FAT SIGNALS--lipases and lipolysis in lipid metabolism and signaling. Cell Metab. 2012 Mar 7;15(3):279-91.
48. Gluckman P, Beedle A, Buklijas T, Low F, Hanson M. Principles of Evolutionary Medicine. 2nd Edn. Oxford University Press; 2016.
49. Chrysostomou SE, Eder S, Pototschnig I, Mayer AL, Derler M, Mussbacher M, et al. R-ketorolac ameliorates cancer-associated cachexia and prolongs survival of tumor-bearing mice. J Cachexia Sarcopenia Muscle. 2024 Apr 1;15(2):562-74.
50. Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers [Internet]. 2018;4(1):17105.
51. Riccardi DMDR, das Neves RX, de Matos-Neto EM, Camargo RG, Lima JDCC, Radloff K, et al. Plasma Lipid Profile and Systemic Inflammation in Patients With Cancer Cachexia. Front Nutr. 2020 Jan 31;7:4.
52. Mangano GD, Fouani M, D’amico D, Di Felice V, Barone R. Cancer‐Related Cachexia: The Vicious Circle between Inflammatory Cytokines, Skeletal Muscle, Lipid Metabolism and the Possible Role of Physical Training. Int J Mol Sci. 2022 Mar 10;23(6):3004.
53. Boire A, Burke K, Cox TR, Guise T, Jamal-Hanjani M, Janowitz T, et al. Why do patients with cancer die? Nat Rev Cancer. 2024 Aug;24(8):578-89.
54. McDonald JJ, McMillan DC, Laird BJA. Targeting IL-1α in cancer cachexia: a narrative review. Curr Opin Support Palliat Care. 2018 Dec;12(4):453-9.
55. Miller J, Laird BJA, Skipworth RJE. The immunological regulation of cancer cachexia and its therapeutic implications. Journal of Cancer Metastasis and Treatment. 2019;5:68.
56. Rounis K, Makrakis D, Gioulbasanis I, Ekman S, De Petris L, Mavroudis D, et al. Cancer Cachexia and Antitumor Immunity: Common Mediators and Potential Targets for New Therapies. Life (Basel). 2022 Jun 12;12(6):880.
57. Zhang D, Zheng H, Zhou Y, Tang X, Yu B, Li J. Association of IL-1beta gene polymorphism with cachexia from locally advanced gastric cancer. BMC Cancer. 2007 Mar 14;7:45.
58. Scheede-Bergdahl C, Watt HL, Trutschnigg B, Kilgour RD, Haggarty A, Lucar E, et al. Is IL-6 the best pro-inflammatory biomarker of clinical outcomes of cancer cachexia? Clin Nutr. 2012 Feb;31(1):85-8.
59. Rébé C, Ghiringhelli F. Interleukin-1β and Cancer. Cancers (Basel). 2020 Jul 4;12(7):1791.
60. Braun TP, Zhu X, Szumowski M, Scott GD, Grossberg AJ, Levasseur PR, et al. Central nervous system inflammation induces muscle atrophy via activation of the hypothalamic-pituitary-adrenal axis. Journal of Experimental Medicine. 2011 Nov 21;208(12):2449-63.
61. Peyta L, Jarnouen K, Pinault M, Coulouarn C, Guimaraes C, Goupille C, et al. Regulation of hepatic cardiolipin metabolism by TNFα: Implication in cancer cachexia. Biochim Biophys Acta Mol Cell Biol Lipids. 2015 Nov 1;1851(11):1490-500.
62. Siddiqui JA, Pothuraju R, Jain M, Batra SK, Nasser MW. Advances in cancer cachexia: Intersection between affected organs, mediators, and pharmacological interventions. Biochim Biophys Acta Rev Cancer. 2020 Apr;1873(2):188359.
63. Cao Z, Scott AM, Hoogenraad NJ, Osellame LD. Mediators and clinical treatment for cancer cachexia: a systematic review. JCSM Rapid Commun. 2021 Jul;4(2):166-86.
64. Ling T, Zhang J, Ding F, Ma L. Role of growth differentiation factor 15 in cancer cachexia (Review). Oncol Lett. 2023 Sep 13;26(5):462.
65. Mah AY, Cooper MA. Metabolic regulation of natural killer cell IFN-γ production. Crit Rev Immunol. 2016;36(2):131-47.
66. Jorgovanovic D, Song M, Wang L, Zhang Y. Roles of IFN-γ in tumor progression and regression: A review. Biomark Res. 2020 Sep 29;8:49.