Cancer cachexia: traditional therapies and novel molecular mechanism-based approaches to treatment

Nagi B Kumar, Aslam Kazi, Tiffany Smith, Theresa Crocker, Daohai Yu, Richard R Reich, Kiran Reddy, Sally Hastings, Martine Exterman, Lodovico Balducci, Kyle Dalton, Gerold Bepler, Nagi B Kumar, Aslam Kazi, Tiffany Smith, Theresa Crocker, Daohai Yu, Richard R Reich, Kiran Reddy, Sally Hastings, Martine Exterman, Lodovico Balducci, Kyle Dalton, Gerold Bepler

Abstract

The complex syndrome of cancer cachexia (CC) that occurs in 50% to 80% cancer patients has been identified as an independent predictor of shorter survival and increased risk of treatment failure and toxicity, contributing to the mortality and morbidity in this population. CC is a pathological state including a symptom cluster of loss of muscle (skeletal and visceral) and fat, manifested in the cardinal feature of emaciation, weakness affecting functional status, impaired immune system, and metabolic dysfunction. The most prominent feature of CC is its non-responsiveness to traditional treatment approaches; randomized clinical trials with appetite stimulants, 5-HT3 antagonists, nutrient supplementation, and Cox-2 inhibitors all have failed to demonstrate success in reversing the metabolic abnormalities seen in CC. Interventions based on a clear understanding of the mechanism of CC, using validated markers relevant to the underlying metabolic abnormalities implicated in CC are much needed. Although the etiopathogenesis of CC is poorly understood, studies have proposed that NFkB is upregulated in CC, modulating immune and inflammatory responses induce the cellular breakdown of muscle, resulting in sarcopenia. Several recent laboratory studies have shown that n-3 fatty acid may attenuate protein degradation, potentially by preventing NFkB accumulation in the nucleus, preventing the degradation of muscle proteins. However, clinical trials to date have produced mixed results potentially attributed to timing of interventions (end stage) and utilizing outcome markers such as weight which is confounded by hydration, cytotoxic therapies, and serum cytokines. We propose that selective targeting of proteasome activity with a standardized dose of omega-3-acid ethyl esters, administered to cancer patients diagnosed with early stage CC, in addition to a standard intervention with nutritionally adequate diet and appetite stimulants, will alter metabolic abnormalities by downregulating NFkB, preventing the breakdown of myofibrillar proteins and resulting in increasing serum protein markers, lean body mass, and functional status.

Figures

Figure 1
Figure 1
Proteasome inhibitory activity in serum samples of 14 patients with early symptoms of cancer cachexia treated with n-3 fatty acid supplements.

References

References and Recommended Reading Papers of particular interest, published recently, have been highlighted as: •Of importance ••Of major importance

    1. Ross PJ, Ashley S, Norton A, et al. Do patients with weight loss have a worse outcome when undergoing chemotherapy for lung cancers? Br J Cancer. 2004;90:1905–1911. doi: 10.1038/sj.bjc.6601781.
    1. Jatoi A. Weight loss in patient with advanced cancer: effects, causes, and potential management. Curr Opin Support Palliat Care. 2008;2(1):45–48. doi: 10.1097/SPC.0b013e3282f4b734.
    1. Evans WJ, Morley JE, Argilés J, et al. Cachexia: a new definition. Clin Nutr. 2008;27(6):793–799. doi: 10.1016/j.clnu.2008.06.013.
A classic manuscript that proposes the definition and comprehensive discussion of the CC syndrome.
    1. Behl D, Jatoi A. Pharmacological options for advanced cancer patients with loss of appetite and weight. Expert Opin Pharmacother. 2007;8(8):1085–1090. doi: 10.1517/14656566.8.8.1085.
    1. Hall W, Christie M, Currow D. Cannabinoids and cancer: causation, remediation, and palliation. Lancet Oncol. 2005;6(1):35–42.
    1. Maltoni M, Fabbri L, Nanni O, et al. Serum levels of tumor necrosis factor alpha and other cytokines do not correlate with weight loss and anorexia in cancer patients. Support Care Cancer. 1997;5(2):130–135. doi: 10.1007/BF01262570.
    1. Barton BE. IL-6-like cytokines and cancer cachexia: consequences of chronic inflammation. Immunol Res. 2001;23(1):1–58. doi: 10.1385/IR:23:1:41.
    1. Mantovani G, Maccio A, Mura L, et al. Serum levels of leptin and proinflammatory cytokines in patients with advanced-stage cancer at different sites. J Mol Med. 2000;78(10):554–561. doi: 10.1007/s001090000137.
    1. Deans DA, Wigmore SJ, Gilmour H, et al. Expression of the proteolysis-inducing factor core peptide mRNA is upregulated in both tumour and adjacent normal tissue in gastro-esophageal malignancy. Br J Cancer. 2008;98(1):242. doi: 10.1038/sj.bjc.6604117.
    1. Wigmore SJ, Todorov PT, Barber MD, et al. Characteristics of patients with pancreatic cancer expressing a novel cancer cachectic factor. Br J Surg. 2000;87(1):53–58. doi: 10.1046/j.1365-2168.2000.01317.x.
    1. Tisdale MJ. Catabolic mediators of CC. Curr Opin Support Palliat Care. 2008;2(4):256–261. doi: 10.1097/SPC.0b013e328319d7fa.
This review is comprehensive review that compares the catabolic actions of tumour necrosis factor-alpha (TNF-alpha) and proteolysis-inducing factor (PIF) and their involvement in human cancer cachexia.
    1. Deans C, Wigmore SJ. Systemic inflammation, cachexia and prognosis in patients with cancer. Curr Opin Clin Nurs Metal Care. 2005;8(3):265–269. doi: 10.1097/01.mco.0000165004.93707.88.
This excellent review summarizes current thinking relating to inflammation, cachexia and prognosis in cancer patients, with particular emphasis on studies relating to recent therapeutic advances
    1. Ciechanover A, Orian A, Schwartz AL. The ubiquitin-mediated proteolytic pathway: mode of action and clinical implications. J Cell Biochem Suppl. 2000;34:40–51. doi: 10.1002/(SICI)1097-4644(2000)77:34+<40::AID-JCB9>;2-6.
This article reviews the enzymatic cascade involved in ubiquitin-mediated degradation, describes some of the structural motifs identified by the conjugating machinery, and summarize recent developments in the involvement of the system in the pathogenesis of selected disease states, including CC.
    1. Ghosh S, Karin M. Missing pieces in the NF-kappaB puzzle. Cell. 2002;109(Suppl):S81–S96. doi: 10.1016/S0092-8674(02)00703-1.
    1. Chen S, Fribley A, Wang CY. Potentiation of tumor necrosis factor-mediated apoptosis of oral squamous cell carcinoma cells by adenovirus-mediated gene transfer of NF-kappaB inhibitor. J Dent Res. 2002;81:98–102. doi: 10.1177/154405910208100203.
    1. Mitch WE, Price SR. Transcription factors and muscle cachexia: is there a therapeutic target? Lancet. 2001;357(9258):734–735. doi: 10.1016/S0140-6736(00)04177-5.
    1. Hochstrasser M. Origin and function of ubiquitin-like proteins. Nature. 2009;458(7237):422–429. doi: 10.1038/nature07958.
    1. Voges D, Zwickl P, Baumeister W. The 26S proteasome: a molecular machine designed for controlled proteolysis. Annu Rev Biochem. 1999;68:1015–1068. doi: 10.1146/annurev.biochem.68.1.1015.
    1. Loprinzi CL, Michalak JC, Schaid DJ, et al. Phase III evaluation of four doses of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia. J Clin Oncol. 1993;11(4):762–767.
    1. Lynch GS, Schertzer JD, Ryall JG. Therapeutic approaches for muscle wasting disorders. Pharmacol Ther. 2007;113(3):461–487. doi: 10.1016/j.pharmthera.2006.11.004.
    1. Karcic E, Philpot C, Morley JE. Treating malnutrition with megestrol acetate: literature review and review of our experience. J Nutr Health Aging. 2002;6(3):191–200.
    1. Morley JE. Orexigenic and anabolic agents. Clin Geriatr Med. 2002;18(4):853–866. doi: 10.1016/S0749-0690(02)00036-8.
    1. Femia RA, Goyette RE. The science of megestrol acetate delivery: potential to improve outcomes in cachexia. BioDrugs. 2005;19(3):179–187. doi: 10.2165/00063030-200519030-00004.
    1. Walsh D, Nelson KA, Mahmoud FA. Established and potential therapeutic applications of cannabinoids in oncology. Support Care Cancer. 2003;11(3):137–143.
    1. Ovesen L, Allingstrup L, Hannibal J, et al. Effect of dietary counseling on food intake, body weight, response rate, survival, and quality of life in cancer patients undergoing chemotherapy: a prospective, randomized study. J Clin Oncol. 1993;11:2043–2049.
    1. Deans DA, Tan BH, Wigmore SJ, et al. The influence of systemic inflammation, dietary intake and stage of disease on rate of weight loss in patients with gastro-esophageal cancer. Br J Cancer. 2009;100(1):63–69. doi: 10.1038/sj.bjc.6604828.
    1. Riechelmann RP, Burman D, Tannock IF, et al. Phase II trial of mirtazapine for cancer-related cachexia and anorexia. Am J Hosp Palliat Care. 2010;27(2):106–110. doi: 10.1177/1049909109345685.
    1. Kast RE, Foley KF. Cancer chemotherapy and cachexia: mirtazapine and olanzapine as 5-HT3 antagonists with good antinausea effects. Eur J Cancer Care (Engl). 2007;16(4):351–354. doi: 10.1111/j.1365-2354.2006.00760.x.
    1. Mantovani G, Madeddu C. Cyclooxygenase-2 inhibitors and antioxidants in the treatment of cachexia. Curr Opin Support Palliat Care. 2008;2(4):275–281. doi: 10.1097/SPC.0b013e32830f47e4.
    1. Bozzetti F, Mariani L. Defining and classifying CC: a proposal by SCRINIO Working Group. JPEN J Parenter Enteral Nutr. 2009;33(4):361–367. doi: 10.1177/0148607108325076.
    1. Blum B, Omlin A, Fearon K, et al. European Palliative Care Research Collaborative. Evolving classification systems for CC: ready for clinical practice? Support Care Cancer. 2010;18(3):273–279. doi: 10.1007/s00520-009-0800-6.
    1. McMillan DC. Systemic inflammation, nutritional status and survival in patients with cancer. Curr Opin Clin Nutr Metab Care. 2009;12(3):223–226. doi: 10.1097/MCO.0b013e32832a7902.
    1. Morley JE. Anorexia and weight loss in older persons. J Gerontol A Biol Sci Med Sci. 2003;58(2):131–137.
    1. Bosaeus I. Nutritional support in multimodal therapy for cancer cachexia. Support Care Cancer. 2008;16(5):447–451. doi: 10.1007/s00520-007-0388-7.
This review provides a pragmatic approach to the treatment of cachexia, stating the goal must be to stabilise cachexia and prevent or delay further decline and established the need to evaluate strategies to counteract both hypermetabolism and reduced dietary intake and the importance of improving not only survival but function and quality of life of cancer patients.
    1. Couch M, Lai V, Cannon T, et al. Cancer cachexia syndrome in head and neck cancer patients: part I. Diagnosis, impact on quality of life and survival, and treatment. Head Neck. 2007;29(4):401–411. doi: 10.1002/hed.20447.
    1. Fearon KC, von Meyenfeldt M, Moses AG, et al. Effect of a protein and energy dense n-3 fatty acid enriched oral supplement on weight and lean tissue in CC: a randomized double blind trial. Gut. 2003;52(10):1479–1486. doi: 10.1136/gut.52.10.1479.
    1. Fearon KC. Cancer cachexia: developing multimodal therapy for a multidimensional problem. Eur J Cancer. 2008;44(8):1124–1132. doi: 10.1016/j.ejca.2008.02.033.
This review provides a comprehensive evidence-base and makes a case for multimodal approaches for the design of complex intervention studies in the management of cancer cachexia.
    1. Al-Majid S, Waters H. The biological mechanisms of cancer-related skeletal muscle wasting: the role of progressive resistance exercise. Biol Res Nurs. 2008;10(1):7–20. doi: 10.1177/1099800408317345.
This article discusses several cancer-related skeletal muscle wasting mechanisms and proposes how physical activity might attenuate muscle wasting by counteracting some of these mechanisms.
    1. Karcic E, Philpot C, Morley JE. Treating malnutrition with megestrol acetate: literature review and review of our experience. J Nutr Health Aging. 2002;6(3):191–200.
    1. De Petrocellis L, Melck D, Bisogno T, Di Marzo V. Endocannabinoids and fatty acid amides in cancer, inflammation and related disorders. Chem Phys Lipids. 2000;108(1-2):191–209. doi: 10.1016/S0009-3084(00)00196-1.
    1. United States Department of Health & Human Services, Statistics, (USDHHS), March 4, 2002
    1. Fearon KC, Van Meyenfeldt MF, Moses AG, et al. Effect of a protein and energy dense N-3 fatty acid enriched oral supplement on loss of weight and lean tissue in CC: a randomised double blind trial. Gut. 2003;52(10):1479–1486. doi: 10.1136/gut.52.10.1479.
    1. Dewey A, Baughan C, Dean T, et al. Eicosapentaenoic acid (EPA, an omega-3 fatty acid from fish oils) for the treatment of cancer cachexia. Cochrane Database Syst Rev. 2007;24(1):CD004597.
    1. Hamerman D. Molecular-based therapeutic approaches in treatment of anorexia of aging and cancer cachexia. J Gerontol A Biol Sci Med Sci. 2002;57(8):M511–M518.
    1. Mantovani G, Macciò A, Madeddu C, et al. Randomized phase III clinical trial of five different arms of treatment for patients with cancer cachexia: interim results. Nutrition. 2008;24(4):305–313.
This is a phase III randomized study which includes all the agents currently used to treat CC and examines several safety and efficacy endpoints of cancer cachexia: lean body mass (LBM), resting energy expenditure (REE), fatigue; and relevant secondary endpoints: appetite, quality of life, grip strength, Glasgow Prognostic Score (GPS) and proinflammatory cytokines.
    1. Wigmore SJ, Ross JA, Falconer JS, et al. The effect of polyunsaturated fatty acids on the progress of cachexia in patients with pancreatic cancer. Nutrition. 1996;12(1 Suppl):S27–S305.

Source: PubMed

3
Subscribe