Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter Open Access August 9, 2011

Melanocortin system in cancer-related cachexia

  • Julie Bienertová-Vašků EMAIL logo , Petr Bienert , Dalibor Valík and Anna Vašků
From the journal Open Medicine

Abstract

The melanocortin system plays a pivotal role in the regulation of appetite and energy balance. It was recognized to play an important role in the development of cancer-related cachexia, a debilitating condition characterized by progressive body wasting associated with anorexia, increased resting energy expediture and loss of fat as well as lean body mass that cannot be simply prevented or treated by adequate nutritional support.

The recent advances in understanding of mechanisms underlying cancer-related cachexia led to consequent recognition of the melanocortin system as an important potential therapeutic target. Several molecules have been made available for animal experiments, including those with oral bioavailability, that act at various checkpoints of the melanocortin system and that might confer singificant benefits for the patients suffering from cancer-related cachexia. The application of melanocortin 4 receptor antagonists/agouti-related peptide agonists has been however restricted to animal models and more pharmacological data will be necessary to progress to clinical trials on humans. Still, pharmacological targeting of the melanocortin system seem to represent an elegant and promising way of treatment of cancer-related cachexia.

[1] Argilés J.M., Moore-Carrasco R., Busquets S., López-Soriano F.J. Catabolic mediators as targets for cancer cachexia. Drug Discov Today., 2003, 18, 838–844 http://dx.doi.org/10.1016/S1359-6446(03)02826-510.1016/S1359-6446(03)02826-5Search in Google Scholar

[2] Tisdale M.J. Cachexia in cancer patients. Nat Rev Cancer., 2002, 11, 862–871 http://dx.doi.org/10.1038/nrc92710.1038/nrc927Search in Google Scholar PubMed

[3] DeWys W.D., Weight loss and nutritional abnormalities in cancer patients: incidence, severity and significance. In: Clinics in Oncology, Calman K.C., Fearon K.C.H. (Eds.) London: Saunders, vol. 5, no. 2, p. 251–261., 1986 Search in Google Scholar

[4] Laviano A, Meguid MM, Inui A, Muscaritoli M, Rossi-Fanelli F. Therapy insight: Cancer anorexia-cachexia syndrome—when all you can eat is yourself. Nat Clin Pract Oncol., 2005, 3, 158–165 http://dx.doi.org/10.1038/ncponc011210.1038/ncponc0112Search in Google Scholar PubMed

[5] Evans W.J., Morley J.E., Argiles J., Bales C., Baracos V., et al. Cachexia: A new definition. Clin Nutr 27., 2008, 793–799 http://dx.doi.org/10.1016/j.clnu.2008.06.01310.1016/j.clnu.2008.06.013Search in Google Scholar PubMed

[6] Tisdale M.J. Are tumoral factors responsible for host tissue wasting in cancer cachexia? Future Oncol., 2010, 4, 503–513 http://dx.doi.org/10.2217/fon.10.2010.2217/fon.10.20Search in Google Scholar PubMed

[7] Fearon K.C.H., Voss A.S., Hustend D.S. Definition of cancer cachexia: effect of weight loss, reduced food intake and systemic inflammation on functional status and prognosis. Am J Clin Nutr., 2006, 83, 1345–1350 10.1093/ajcn/83.6.1345Search in Google Scholar PubMed

[8] DeWys W.D. Weight loss and nutritional abnormalities in cancer patients: incidence, severity and significance. In: Clinics in Oncology, edited by Calman KC and Fearon KCH. London: Saunders, 1986, vol. 5, no. 2, p. 251–261 Search in Google Scholar

[9] Bing C., Brown M., King P., Collins P., Tisdale M.J., Williams G. Increased gene expression of brown fat uncoupling protein (UCP)1 and skeletal muscle UCP2 and UCP3 in MAC16-induced cancer cachexia. Cancer Res. 2000, 9, 2405–2410 10.1042/cs098001PaSearch in Google Scholar

[10] Kulstad R., Schoeller D.A. The energetics of wasting diseases. Curr Opin Clin Nutr Metab Care., 2007, 4, 488–493 10.1097/MCO.0b013e3281e38942Search in Google Scholar PubMed

[11] Bennani-Baiti N., Davis M.P.. Cytokines and cancer anorexia cachexia syndrome. Am J Hosp Palliat Care. 2008, 5, 407–411 http://dx.doi.org/10.1177/104990910831551810.1177/1049909108315518Search in Google Scholar PubMed

[12] Yavuzsen T., Davis M.P., Walsh D., LeGrand S., Lagman R. Systematic review of the treatment of cancer-associated anorexia and weight loss. J Clin Oncol., 2005, 23, 8500–8511 http://dx.doi.org/10.1200/JCO.2005.01.801010.1200/JCO.2005.01.8010Search in Google Scholar PubMed

[13] DeBoer M.D. Update on melanocortin interventions for cachexia: progress toward clinical application. Nutrition., 2010, 2,146–151 http://dx.doi.org/10.1016/j.nut.2009.07.00310.1016/j.nut.2009.07.003Search in Google Scholar PubMed PubMed Central

[14] Scarlett J.M., Marks D.L. The use of melanocortin antagonists in cachexia of chronic disease. Expert Opin Investig Drugs., 2005, 14,1233–1239 http://dx.doi.org/10.1517/13543784.14.10.123310.1517/13543784.14.10.1233Search in Google Scholar PubMed

[15] Marks D.L., Ling N., Cone R.D. Role of the central melanocortin system in cachexia. Cancer Res 2001;61:1432–1438 Search in Google Scholar

[16] Wisse B.E., Frayo R.S., Schwartz M.W., Cummings D.E. Reversal of cancer anorexia by blockade of central melanocortin receptors in rats. Endocrinology., 2001,142, 3292–3301 http://dx.doi.org/10.1210/en.142.8.329210.1210/en.142.8.3292Search in Google Scholar

[17] Tung Y.C., Yeo G.S. Central melanocortin signaling regulates cholesterol. Nat Neurosci., 2010, 7, 779–780 http://dx.doi.org/10.1038/nn0710-77910.1038/nn0710-779Search in Google Scholar PubMed

[18] Stewart P.M., Boulton A., Kumar S., Clark P.M., Shackleton C.H.. Cortisol metabolism in human obesity: impaired cortisone—>cortisol conversion in subjects with central adiposity. J Clin Endocrinol Metab., 1999, 3, 1022–1027 http://dx.doi.org/10.1210/jc.84.3.102210.1210/jc.84.3.1022Search in Google Scholar

[19] Cone R.D. Anatomy and regulation of the central melanocortin system. Nat Neurosci., 2005, 5, 571–578 http://dx.doi.org/10.1038/nn145510.1038/nn1455Search in Google Scholar PubMed

[20] Whitaker K.W., Reyes T.M. Central blockade of melanocortin receptors attenuates the metabolic and locomotor responses to peripheral interleukin-1beta administration. Neuropharmacology., 2008, 54, 509–520 http://dx.doi.org/10.1016/j.neuropharm.2007.10.01410.1016/j.neuropharm.2007.10.014Search in Google Scholar PubMed PubMed Central

[21] Ellacott K.L., Halatchev I.G., Cone R.D. Interactions between gut peptides and the central melanocortin system in the regulation of energy homeostasis. Peptides., 2006, 2, 340–349 http://dx.doi.org/10.1016/j.peptides.2005.02.03110.1016/j.peptides.2005.02.031Search in Google Scholar PubMed

[22] Marks D.L., Cone R.D. The role of the melanocortin-3 receptor in cachexia. Ann N Y Acad Sci., 2003, 994, 258–266 http://dx.doi.org/10.1111/j.1749-6632.2003.tb03188.x10.1111/j.1749-6632.2003.tb03188.xSearch in Google Scholar PubMed

[23] Cone R.D. Studies on the physiological functions of the melanocortin system. Endocr Rev., 2006, 7, 736–749 10.1210/er.2006-0034Search in Google Scholar PubMed

[24] Butler A.A., Marks D.L., Fan W., Kuhn C.M., Bartolome M., Cone R.D. Melanocortin-4 receptor is required for acute homeostatic responses to increased dietary fat. Nat Neurosci., 2001, 6, 605–611 http://dx.doi.org/10.1038/8842310.1038/88423Search in Google Scholar PubMed

[25] Tung Y.C., Piper S.J., Yeung D., O’Rahilly S., Coll A.P. A comparative study of the central effects of specific proopiomelancortin (POMC)-derived mela nocortin peptides on food intake and body weight in pomc null mice. Endocrinology., 2006, 12, 5940–5947 http://dx.doi.org/10.1210/en.2006-086610.1210/en.2006-0866Search in Google Scholar PubMed PubMed Central

[26] Coll A.P. Effects of pro-opiomelanocortin (POMC) on food intake and body weight: mechanisms and therapeutic potential? Clin Sci (Lond)., 2007, 4, 171–182 10.1042/CS20070105Search in Google Scholar PubMed

[27] Jackson P.J., Yu B., Hunrichs B., Thompson D.A., Chai B., Gantz I., Millhauser G.L. Chimeras of the agouti-related protein: insights into agonist and antagonist selectivity of melanocortin receptors. Peptides., 2005, 10, 1978–1987 http://dx.doi.org/10.1016/j.peptides.2004.12.03610.1016/j.peptides.2004.12.036Search in Google Scholar PubMed

[28] Fan W., Boston B.A., Kesterson R.A., Hruby V.J., Cone R.D. Role of melanocortinergic neurons in feeding and the agouti obesity syndrome. Nature, 1997, 385,165–168 http://dx.doi.org/10.1038/385165a010.1038/385165a0Search in Google Scholar PubMed

[29] Ollmann M.M., Wilson B.D., Yang Y.K., Kerns J.A., Chen Y., Gantz I., et al. Antagonism of central melanocortin receptors in vitro and in vivo by agoutirelated protein. Science,1997, 278,135–138 http://dx.doi.org/10.1126/science.278.5335.13510.1126/science.278.5335.135Search in Google Scholar PubMed

[30] Farooqi I.S., Keogh J.M., Yeo G.S., Lank E.J., Cheetham T., O’Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med., 2003, 348, 1085–1095 http://dx.doi.org/10.1056/NEJMoa02205010.1056/NEJMoa022050Search in Google Scholar PubMed

[31] Olszewski P.K., Wickwire K., Wirth M.M., Levine A.S., Giraudo S.Q. Agouti-related protein: appetite or reward? Ann N Y Acad Sci., 2003, 994,187–191 http://dx.doi.org/10.1111/j.1749-6632.2003.tb03179.x10.1111/j.1749-6632.2003.tb03179.xSearch in Google Scholar PubMed

[32] Hagan M.M., Rushing P.A., Benoit S.C., Woods S.C., Seeley R.J. Opioid receptor involvement in the effect of AgRP — (83–132) on food intake and food selection. Am J Physiol Regul Integr Comp Physiol., 2001, 3,:R814–R821 10.1152/ajpregu.2001.280.3.R814Search in Google Scholar PubMed

[33] Butler A.A., Cone R.D. Knockout studies defining different roles for melanocortin receptors in energy homeostasis. Ann N Y Acad Sci., 2003, 994, 240–245. http://dx.doi.org/10.1111/j.1749-6632.2003.tb03186.x10.1111/j.1749-6632.2003.tb03186.xSearch in Google Scholar PubMed

[34] Ste Marie L., Miura G.I., Marsh D.J., Yagaloff K., Palmiter R.D. A metabolic defect promotes obesity in mice lacking melanocortin-4 receptors. Proc Natl Acad Sci U S A., 2000,97,12339–12344 http://dx.doi.org/10.1073/pnas.22040949710.1073/pnas.220409497Search in Google Scholar

[35] Stütz A.M., Morrison C.D., Argyropoulos G. The agouti-related protein and its role in energy homeostasis. Peptides., 2005, 10,1771–1781 http://dx.doi.org/10.1016/j.peptides.2004.12.02410.1016/j.peptides.2004.12.024Search in Google Scholar

[36] Ilnytska O., Argyropoulos G. The role of the Agouti-Related Protein in energy balance regulation. Cell Mol Life Sci., 2008, 17, 2721–2731 http://dx.doi.org/10.1007/s00018-008-8104-410.1007/s00018-008-8104-4Search in Google Scholar

[37] Qian S., Chen H., Weingarth D., Trumbauer ME., Novi D.E., Guan X. et al. Neither Agouti-Related Protein nor Neuropeptide Y Is Critically Required for the Regulation of Energy Homeostasis in Mice. Mol. Cell. Biol., 2002, 22, 5027–5035 http://dx.doi.org/10.1128/MCB.22.14.5027-5035.200210.1128/MCB.22.14.5027-5035.2002Search in Google Scholar

[38] Wortley K.E., Anderson K.D., Yasenchak J., Murphy A., Valenzuela D., Diano S. et al. Agoutirelated protein-deficient mice display an age-related lean phenotype. Cell. Metab., 2005, 2, 421–427 http://dx.doi.org/10.1016/j.cmet.2005.11.00410.1016/j.cmet.2005.11.004Search in Google Scholar

[39] Vaughan C.H., Moore M.C., Haskell-Luevano C., Rowland N.E. Meal patterns and foraging in melanocortin receptor knockout mice. Physiol Behav., 2005, 1, 129–133 http://dx.doi.org/10.1016/j.physbeh.2004.10.01610.1016/j.physbeh.2004.10.016Search in Google Scholar

[40] Koegler F.H., Schaffhauser R.O., Mynatt R.L., York D.A., Bray G.A. Macronutrient diet intake of the lethal yellow agouti (Ay/a) mouse. Physiol Behav., 1999, 5, 809–812 http://dx.doi.org/10.1016/S0031-9384(99)00104-310.1016/S0031-9384(99)00104-3Search in Google Scholar

[41] Cheung W.W., Kuo H.J., Markison S., Chen C., Foster A.C., Marks D.L. et al. Peripheral administration of the melanocortin-4 receptor antagonist NBI-12i ameliorates uremia-associated cachexia in mice. J Am Soc Nephrol., 2007, 9, 2517–2524 http://dx.doi.org/10.1681/ASN.200609102410.1681/ASN.2006091024Search in Google Scholar PubMed

[42] Bowe D.D., Scarlett J.M., Basra A.K., Steiner R.A., Marks D.L. Blockade of central melanocortin signaling promotes accumulation of lean body mass in rodent models of chronic heart failure. J Investig Med., 2007, 55:S77 10.1097/00042871-200701010-00018Search in Google Scholar

[43] Basra A.K., Scarlett J.M., Bowe D.D., Steiner R.A., Marks D.L. Central melanocortin blockade attenuates cardiac cachexia in a rat model of chronic heart failure. J Investig Med., 2008, 56, 229–230 Search in Google Scholar

[44] Nicholson J.R., Kohler G., Schaerer F., Senn C., Weyermann P., Hofbauer K.G. Peripheral administration of a melanocortin 4-receptor inverse agonist prevents loss of lean body mass in tumor-bearing mice. J Pharmacol Exp Ther., 2006, 2, 771–777 http://dx.doi.org/10.1124/jpet.105.09772510.1124/jpet.105.097725Search in Google Scholar PubMed

[45] Weyermann P., Dallmann R., Magyar J., Anklin C., Hufschmid M., Dubach-Powell J. et al. Orally available selective melanocortin-4 receptor antagonists stimulate food intake and reduce cancer-induced cachexia in mice. PLoS One., 2009; 4(3), e4774 http://dx.doi.org/10.1371/journal.pone.000477410.1371/journal.pone.0004774Search in Google Scholar PubMed PubMed Central

[46] Tung Y.C., Piper S.J., Yeung D., O’Rahilly S. and Coll A.P. A comparative study of the central effects of specific proopiomelancortin (POMC)-derived melanocortin peptides on food intake and body weight in pomc null mice. Endocrinology 2006, 147, 5940–5947 http://dx.doi.org/10.1210/en.2006-086610.1210/en.2006-0866Search in Google Scholar PubMed PubMed Central

[47] Hoggard N., Rayner D.V., Johnston S.L., Speakman J.R. Peripherally administered [Nle4,D-Phe7]-alpha-melanocyte stimulating hormone increases resting metabolic rate, while peripheral agouti-related protein has no effect, in wild type C57BL/6 and ob/ob mice. J Mol Endocrinol 2004; 33, 693–703 http://dx.doi.org/10.1677/jme.1.0163210.1677/jme.1.01632Search in Google Scholar PubMed

[48] Markison S., Foster A.C., Chen C., Brookhart G.B., Hesse A., Hoare S.R. The regulation of feeding and metabolic rate and the prevention of murine cancer cachexia with a small-molecule melanocortin-4 receptor antagonist. Endocrinology., 2005, 146(6),2766–2773 http://dx.doi.org/10.1210/en.2005-014210.1210/en.2005-0142Search in Google Scholar PubMed

[49] Santhera Pharmaceuticals (Switzerland) AG (2008) Preparation of imidazopyridines as melanocortin-4 receptor antagonists. WO 2008/116665 A1 Search in Google Scholar

[50] Santhera Pharmaceuticals (Switzerland) AG (2009) Subsituted heteroarylpiperidine derivatives as melanocortin-4 receptor modulators. WO 2009/010299 A1 Search in Google Scholar

[51] Joppa M.A., Ling N., Chen C., Gogas K.R., Foster A.C., Markison S. Central administration of peptide and small molecule MC4 receptor antagonists induce hyperphagia in mice and attenuate cytokine-induced anorexia. Peptides., 2005, 26(11), 2294–2301 http://dx.doi.org/10.1016/j.peptides.2005.03.00210.1016/j.peptides.2005.03.002Search in Google Scholar PubMed

[52] Chen C., Tucci F.C., Jiang W., Tran J.A., Fleck B.A., Hoare S.R. Pharmacological and pharmacokinetic characterization of 2-piperazine-alpha-isopropyl benzylamine derivatives as melanocortin-4 receptor antagonists. Bioorg Med Chem., 2008,16(10), 5606–5618 http://dx.doi.org/10.1016/j.bmc.2008.03.07210.1016/j.bmc.2008.03.072Search in Google Scholar PubMed

[53] Vos T.J., Caracoti A., Che J.L., Dai M., Farrer C.A., Forsyth N.E., et al. Identification of 2-[2-[2-(5-bromo-2 — methoxyphenyl)-ethyl]-3-fluorophenyl]-4,5-dihydro-1H-imidazole (ML00253764), a small molecule melanocortin 4 receptor antagonist that effectively reduces tumor-induced weight loss in a mouse model. J Med Chem., 2004,47(7),1602–164 http://dx.doi.org/10.1021/jm034244g10.1021/jm034244gSearch in Google Scholar PubMed

[54] Cheung W.W., Kuo H.J., Markison S., Chen C., Foster A.C., Marks D.L. et al. Peripheral administration of the melanocortin-4 receptor antagonist NBI-12i ameliorates uremia-associated cachexia in mice. J Am Soc Nephrol., 2007, 18(9), 2517–2524 http://dx.doi.org/10.1681/ASN.200609102410.1681/ASN.2006091024Search in Google Scholar PubMed

[55] Nijenhuis W.A., Oosterom J., Adan R.A.. AgRP(83–132) acts as an inverse agonist on the human-melanocortin-4 receptor. Mol Endocrinol., 2001, 15(1), 164–171 http://dx.doi.org/10.1210/me.15.1.16410.1210/me.15.1.164Search in Google Scholar

[56] Oosterom J., Garner K.M., den Dekker W.K., Nijenhuis W.A., Gispen W.H., Burbach J.P, et al. Common requirements for melanocortin-4 receptor selectivity of structurally unrelated melanocortin agonist and endogenous antagonist, Agouti protein. J Biol Chem., 2001, 276(2), 931–936 http://dx.doi.org/10.1074/jbc.M00726120010.1074/jbc.M007261200Search in Google Scholar PubMed

[57] Adan R.A., Tiesjema B., Hillebrand J.J., la Fleur S.E., Kas M.J., de Krom M. The MC4 receptor and control of appetite. Br J Pharmacol., 2006, 149(7), 815–827 http://dx.doi.org/10.1038/sj.bjp.070692910.1038/sj.bjp.0706929Search in Google Scholar

[58] Joseph C.G., Bauzo R.M., Xiang Z., Shaw A.M., Millard W.J., Haskell-Luevano C. Elongation studies of the human agouti-related protein (AGRP) core decapeptide (Yc[CRFFNAFC]Y) results in antagonism at the mouse melanocortin-3 receptor. Peptides., 2003, 24(2), 263–270 http://dx.doi.org/10.1016/S0196-9781(03)00030-510.1016/S0196-9781(03)00030-5Search in Google Scholar

[59] Fu L.Y., van den Pol A.N. Agouti-related peptide and MC3/4 receptor agonists both inhibit excitatory hypothalamic ventromedial nucleus neurons. J Neurosci., 2008, 28(21), 5433–5449 http://dx.doi.org/10.1523/JNEUROSCI.0749-08.200810.1523/JNEUROSCI.0749-08.2008Search in Google Scholar PubMed PubMed Central

[60] Tisdale M.J. Clinical anticachexia treatments. Nutr Clin Pract., 2006, 21(2), 168–174 http://dx.doi.org/10.1177/011542650602100216810.1177/0115426506021002168Search in Google Scholar PubMed

[61] Rigas J.R., Schuster M., Orlov S.V., Milovanovic B., Prabhash K., Smith J.T. and the ALD518 study group. Affect of ALD518, a humanized anti-IL-6 antibody, on lean body mass loss and symptoms in patients with advanced non-small cell lung cancer (NSCLC): Results of a phase II randomized, double-blind safety and efficacy trial. J Clin Oncol 28 (1534). http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=74&abstractID=50646 Search in Google Scholar

[62] Steiner M.S., Barnette K.G., Hancock M.L., Dodson S.T., Rodriguez D., Morton R.A.; GTx, Inc., Memphis, TN (June 2010). „Effect of GTx-024, a selective androgen receptor modulator (SARM), on stair climb performance and quality of life (QOL) in patients with cancer cachexia”. J Clin Oncol 28 (1534) http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=74&abstractID=52947 Search in Google Scholar

[63] Bhattacharyya G.S., Julka P.K., Bondarde S., Naik R., Ranade A., Bascomb N. et al. (June 2010). „Phase II study evaluating safety and efficacy of coadministering propranolol and etodolac for treating cancer cachexia”. J Clin Oncol 28 (1534). http://www.asco.org/ASCOv2/Meetings/Abstracts?&vmview=abst_detail_view&confID=74&abstractID=49474 Search in Google Scholar

[64] Zhou X., Wang J.L., Lu J., Song Y., Kwak K.S., Jiao Q. et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell., 2010, 142(4), 531–543. http://dx.doi.org/10.1016/j.cell.2010.07.01110.1016/j.cell.2010.07.011Search in Google Scholar PubMed

[65] Trials for cachexia treatment. Available at: http://clinicaltrials.gov/ct2/results?term¼cachexia&pg¼4. Accessed September 1, 2010 Search in Google Scholar

Published Online: 2011-8-9
Published in Print: 2011-10-1

© 2011 Versita Warsaw

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.

Downloaded on 24.4.2024 from https://www.degruyter.com/document/doi/10.2478/s11536-011-0057-6/html
Scroll to top button