Skip to main content

How to Succeed in Marketing Marine Natural Products for Nutraceutical, Pharmaceutical and Cosmeceutical Markets

  • Chapter
  • First Online:
Grand Challenges in Marine Biotechnology

Abstract

The marine ecosystem shelters a vast number of macro- and microorganisms that have developed unique metabolic skills to survive in diverse and hostile habitats. These survival strategies often result in the biosynthesis of an array of secondary metabolites with specific activities and functions in the cellular context. Several metabolites can give origin to high-value commercial products for nutraceutical, pharmaceutical and cosmeceutical markets, among others. This chapter outlines those industries’ paths for marketing marine natural products (MNPs), from discovery and development up to final product marketing. Focus is given on compounds that successfully reached the market and, particularly, the approaches employed by the nutraceutical, pharmaceutical and cosmeceutical companies that succeeded in marketing those products. Some key failures in each market segment are analysed, allowing lessons to be learned and key hurdles to be avoided in MNP development. The main challenges faced during MNP programs are assessed and mapped in the market funnel of common product development routes. Suggestions to surpass these challenges are provided, in order to improve market entry success rates of highly promising marine bioactives in current pipelines, highlighting what can be applied to novel and/or ongoing MNP development programs.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 169.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Costello MJ, May RM, Stork NE (2013) Can we name Earth’s species before they go extinct? Science 339:413–416

    Article  PubMed  CAS  Google Scholar 

  2. McGinn AP (1999) Safeguarding the health of oceans, vol 145. Worldwatch Institute, Danvers

    Google Scholar 

  3. Devries DJ, Hall MR (1994) Marine biodiversity as a source of chemical diversity. Drug Dev Res 33(2):161–173

    Article  CAS  Google Scholar 

  4. Leal MC, Puga J, Serôdio J, Gomes NCM, Calado R (2012) Trends in the discovery of new marine natural products from invertebrates over the last two decades – where and what are we bioprospecting? PLoS One 7(1):e30580. https://doi.org/10.1371/journal.pone.0030580.t003

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Leal MC, Munro MHG, Blunt JW et al (2013) Biogeography and biodiscovery hotspots of macroalgal marine natural products. Nat Prod Rep 30:1380–1390

    Article  PubMed  CAS  Google Scholar 

  6. Montaser R, Luesch H (2011) Marine natural products: a new wave of drugs? Future Med Chem 3(12):1475–1489. https://doi.org/10.4155/fmc.11.118

    Article  PubMed  CAS  Google Scholar 

  7. Martins A, Vieira H, Gaspar H, Santos S (2014) Marketed marine natural products in the pharmaceutical and cosmeceutical industries: tips for success. Mar Drugs 12:1066–1101

    Article  PubMed  PubMed Central  Google Scholar 

  8. Newman DJ, Cragg GM (2012) Natural products as sources of new drugs over the 30 years from 1981 to 2010. J Nat Prod 75(3):311–335. https://doi.org/10.1021/np200906s

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Imhoff JF, Labes A, Wiese J (2011) Bio-mining the microbial treasures of the ocean: new natural products. Biotechnol Adv 29(5):468–482. https://doi.org/10.1016/j.biotechadv.2011.03.001

    Article  PubMed  CAS  Google Scholar 

  10. National Research Council (2002) Marine biotechnology in the twenty-first century: problems, promise and products. National Academies Press, Washington, DC

    Google Scholar 

  11. Munro M, Blunt JW, Dumdei E et al (1999) The discovery and development of marine compounds with pharmaceutical potential. J Biotechnol 70:15–25

    Article  PubMed  CAS  Google Scholar 

  12. Leal MC, Madeira C, Brandao CA, Puga J, Calado R (2012) Bioprospecting of marine invertebrates for new natural products – a chemical and zoogeographical perspective. Molecules 17(8):9842–9854. https://doi.org/10.3390/molecules17089842

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  13. Leal MC, Hilário A, Munro HG, Blunt JW, Calado R (2016) Natural products discovery needs improved taxonomic and geographic information. Nat Prod Rep 33:747–750

    Article  PubMed  CAS  Google Scholar 

  14. Lederer E, Teissier G, Huttrer C (1940) The isolation and chemical composition of calliactine, pigment of the sea anemone “Sargatia parasitica” (Calliactis effoeta). Bull Soc Chim Fr 7:608–615

    CAS  Google Scholar 

  15. MarinLit: A database of the marine natural products literature (2016)

    Google Scholar 

  16. Skropeta D (2008) Deep-sea natural products. Nat Prod Rep 25(6):1131–1166. https://doi.org/10.1039/b808743a

    Article  PubMed  CAS  Google Scholar 

  17. Kinghorn AD, Chin Y-W, Swanson SM (2009) Discovery of natural product anticancer agents from biodiverse organisms. Curr Opin Drug Discov Dev 12(2):189–196

    CAS  Google Scholar 

  18. WoRMS Editorial Board (2016) World register of marine species. http://www.marinespecies.org at VLIZ. Accessed 19 Dec 2016. doi:10.14284/170

  19. Haefner B (2003) Drugs from the deep: marine natural products as drug candidates. Drug Discov Today 8(12):536–544. https://doi.org/10.1016/S1359-6446(03)02713-2

    Article  PubMed  CAS  Google Scholar 

  20. Leal MC, Sheridan C, Osinga R et al (2014) Marine microorganism-invertebrate assemblages: perspectives to solve the “supply problem” in the initial steps of drug discovery. Mar Drugs 12:3929–3952

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Pettit GR, Fujii Y, Hasler JA, Schmidt JM (1982) Isolation and characterization of palystatins A–D. J Nat Prod 45:4

    Google Scholar 

  22. Kim S-K (ed) (2012) Marine cosmeceuticals: trends and prospects. CRC Press, Taylor & Francis Group, Boca Raton, FL

    Google Scholar 

  23. Hurst D, Børresen T, Almesjö L, De Raedemaecker F, Bergseth S (2016) Marine biotechnology strategic research and innovation roadmap: insights to the future direction of European marine biotechnology, 1st edn. Marine Biotechnology ERA-NET, Oostende

    Google Scholar 

  24. Gomez-Ordonez E, Jimenez-Escrig A, Ruperez P (2010) Dietary fibre and physico-chemical properties of several edible seaweeds from the North western Spanish coast. Food Res Int (9):2289–2294

    Article  CAS  Google Scholar 

  25. Plaza M, Cifuentes A, Ibanez E (2008) In the search of new functional food ingredients from algae. Trends Food Sci Technol 19(1):31–39. https://doi.org/10.1016/j.tifs.2007.07.012

    Article  CAS  Google Scholar 

  26. Jimenez-Escrig A, Sanchez-Muniz FJ (2000) Dietary fibre from edible seaweeds: chemical structure, physicochemical properties and effects on cholesterol metabolism. Nutr Res 20(4):585–598. https://doi.org/10.1016/S0271-5317(00)00149-4

    Article  CAS  Google Scholar 

  27. Ruperez P, Saura-Calixto F (2001) Dietary fibre and physicochemical properties of edible Spanish seaweeds. Eur Food Res Technol 212:349–354

    Article  CAS  Google Scholar 

  28. Hu GP, Yuan J, Sun L et al (2011) Statistical research on marine natural products based on data obtained between 1985 and 2008. Mar Drugs 9:514–525

    Article  PubMed  PubMed Central  Google Scholar 

  29. Shahidi F (2009) Nutraceuticals and functional foods: whole versus processed foods. Trends Food Sci Technol 20:376–387

    Article  CAS  Google Scholar 

  30. Shahidi F, Zhong Y (2005) Marine mammal oils. In: Bailey’s industrial oil and fat products, 6th edn. Wiley, New York, pp 259–278

    Chapter  Google Scholar 

  31. Foundation ES (2010) Marine biotechnology: a new vision and strategy for Europe Marine Board. ESF Position Paper n° 15:96

    Google Scholar 

  32. Afonso C, Costa S, Cardoso C et al (2015) Evaluation of the risk/benefit associated to the consumption of raw and cooked farmed meagre based on the bioaccessibility of selenium, eicosapentaenoic acid and docosahexaenoic acid, total mercury, and methylmercury determined by an in vitro digestion model. Food Chem 170:249–256. https://doi.org/10.1016/j.foodchem.2014.08.044

    Article  PubMed  CAS  Google Scholar 

  33. Kaur G, Cameron-Smith D, Garg M, Sinclair AJ (2011) Docosapentaenoic acid (22:5n-3): a review of its biological effects. Prog Lipid Res 50(1):28–34. https://doi.org/10.1016/j.plipres.2010.07.004

    Article  PubMed  CAS  Google Scholar 

  34. Bandarra NM, Batista I, Nunes ML (2011) In: Chen G (ed) Fatty acids: types, roles and health effects. Nova Science, Hauppauge, NY (ebook)

    Google Scholar 

  35. Byelashov OA, Sinclair AJ, Kaur G (2015) Dietary sources, current intakes, and nutritional role of omega-3 docosapentaenoic acid. Lipid Technol 27(4):79–82. https://doi.org/10.1002/lite.201500013

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Lordan S, Ross RP, Stanton C (2011) Marine bioactives as functional food ingredients: potential to reduce the incidence of chronic diseases. Mar Drugs 9(6):1056–1100. https://doi.org/10.3390/md9061056

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Ngo DH, Vo TS, Ngo DN, Wijesekara I, Kim SK (2012) Biological activities and potential health benefits of bioactive peptides derived from marine organisms. Int J Biol Macromol 51(4):378–383. https://doi.org/10.1016/j.ijbiomac.2012.06.001

    Article  PubMed  CAS  Google Scholar 

  38. Shukla S (2016) Therapeutic importance of peptides from marine source: a mini review. Ind J Geo-Mar Sci 45:1422–1431

    Google Scholar 

  39. Suleria HA, Osborne S, Masci P, Gobe G (2015) Marine-based nutraceuticals: an innovative trend in the food and supplement industries. Mar Drugs 13(10):6336–6351. https://doi.org/10.3390/md13106336

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Mozaffarian D, Rimm EB (2006) Fish intake, contaminants, and human health: evaluating the risks and the benefits. JAMA 296(15):1885–1899. https://doi.org/10.1001/jama.296.15.1885

    Article  PubMed  CAS  Google Scholar 

  41. Balk EM, Lichtenstein AH, Chung M, Kupelnick B, Chew P, Lau J (2006) Effects of omega-3 fatty acids on serum markers of cardiovascular disease risk: a systematic review. Atherosclerosis 189(1):19–30. https://doi.org/10.1016/j.atherosclerosis.2006.02.012

    Article  PubMed  CAS  Google Scholar 

  42. Calder PC (2006) n-3 Polyunsaturated fatty acids, inflammation, and inflammatory diseases. Am J Clin Nutr 83(6 Suppl):1505S–1519S

    Article  PubMed  CAS  Google Scholar 

  43. Chapkin RS, Davidson LA, Ly L, Weeks BR, Lupton JR, McMurray DN (2007) Immunomodulatory effects of (n-3) fatty acids: putative link to inflammation and colon cancer. J Nutr 137(1 Suppl):200S–204S

    Article  PubMed  CAS  Google Scholar 

  44. Sun H, Berquin IM, Edwards IJ (2005) Omega-3 polyunsaturated fatty acids regulate syndecan-1 expression in human breast cancer cells. Cancer Res 65(10):4442–4447. https://doi.org/10.1158/0008-5472.CAN-04-4200

    Article  PubMed  CAS  Google Scholar 

  45. Brasky TM, Darke AK, Song X et al (2013) Plasma phospholipid fatty acids and prostate cancer risk in the SELECT trial. J Natl Cancer Inst 105(15):1132–1141. https://doi.org/10.1093/jnci/djt174

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Uauy R, Dangour AD (2006) Nutrition in brain development and aging: role of essential fatty acids. Nutr Rev 64(5 Pt 2):S24–S33; discussion S72–S91

    Article  Google Scholar 

  47. Peet M, Stokes C (2005) Omega-3 fatty acids in the treatment of psychiatric disorders. Drugs 65(8):1051–1059

    Article  PubMed  CAS  Google Scholar 

  48. EFSA Panel on Dietetic Products NaAN (2012) Scientific opinion related to the tolerable upper intake level of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA). EFSA J 10(7):48

    Google Scholar 

  49. Barrow CJ, Nolan C, MacLead M, Santosa S, Holub BJ (2009) Bioequivalence of encapsulated and microencapsulated fish-oil supplementation. J Funct Foods 1:38–43

    Article  CAS  Google Scholar 

  50. Chen Q, McGillivray D, Wen J, Zhong F, Quek S (2013) Co-encapsulation of fish oil with phytosterol esters and limonene by milk proteins. J Food Eng 117:505–512

    Article  CAS  Google Scholar 

  51. Chatterjee S, Judeh ZMA (2016) Microencapsulation of fish oil. Lipid Technol 28:13–15

    Article  CAS  Google Scholar 

  52. Wanasundara UN, Wanasundara J, Shahidi F (2002) Omega-3 fatty acid concentrates: a review of production technologies. In: Alasalvarand C, Taylor T (eds) Seafoods: quality, technology and nutraceutical applications. Springer, New York, pp 157–174

    Chapter  Google Scholar 

  53. (BIOHAZ) EPoCH (2010) Scientific opinion on fish oil for human consumption. Food hygiene, including rancidity. EFSA J 8(10):1874

    Article  CAS  Google Scholar 

  54. Beer C (2011) Krill: the ocean’s gold. Nutraceuticals World. http://www.nutraceuticalsworld.com/contents/view_features/2011-10-03/krill-the-oceans-gold

  55. Corporation (U.S.) (2006) https://www.mordorintelligence.com/industry-reports/global-nutraceuticals-market-industrygclid=CMOdrsr2lNECFQafGwodDugLyw. Accessed Dec 2016

  56. Moloughney S (2011) Diving deep into the marine nutraceuticals market – consumers continue to ride the ocean’s waves to better health. Nutraceuticals World. October, p 44–52. http://www.nutraceuticalsworld.com/contents/view_features/2011-10-03/diving-deep-into-the-marine-nutraceuticals-market

  57. Lee CM, Barrow CJ, Kim S, Miyashita K, Shahidi F (2012) Global trends in marine nutraceuticals. Food Technol 65:23–31

    Google Scholar 

  58. Shahidi F (2003) Nutraceuticals and bioactives from seafood byproducts. In: Bechtel PJ (ed) Advances in seafood byproducts: 2002 Conference proceedings Alaska Sea Grant College Program. University of Alaska Fairbanks, Fairbanks, p 566

    Google Scholar 

  59. Kim SK, Mendis E (2006) Bioactive compounds from marine processing byproducts – a review. Food Res Int 39:383–393

    Article  CAS  Google Scholar 

  60. Kim SK, Wijesekara I (2010) Development and biological activities of marine-derived bioactive peptides: a review. J Funct Foods 2:1–9

    Article  CAS  Google Scholar 

  61. FAO (2016) The state of world fisheries and aquaculture 2016. Contributing to food security and nutrition for all, Rome

    Google Scholar 

  62. Nunes ML, Bandarra NM, Batista I (2011) Health benefits associated with seafood consumption. In: Alasalvar C, Shahidi F, Miyashita K, Wanasundara U (eds) Handbook of seafood quality, safety and health applications, vol 122. Blackwell, Oxford, pp 876–883

    Google Scholar 

  63. Bernstein AM, Sun Q, Hu FB, Stampfer MJ, Manson JE, Willett WC (2010) Major dietary protein sources and risk of coronary heart disease in women. Circulation 122(9):876–883. https://doi.org/10.1161/CIRCULATIONAHA.109.915165

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Tremblay F, Lavigne C, Jacques H, Marette A (2003) Dietary cod protein restores insulin-induced activation of phosphatidylinositol 3-kinase/Akt and GLUT4 translocation to the T-tubules in skeletal muscle of high-fat-fed obese rats. Diabetes 52(1):29–37

    Article  PubMed  CAS  Google Scholar 

  65. Paddon-Jones D, Westman E, Mattes RD, Wolfe RR, Astrup A, Westerterp-Plantenga M (2008) Protein, weight management, and satiety. Am J Clin Nutr 87(5):1558S–1561S

    Article  PubMed  CAS  Google Scholar 

  66. Hultin HO, Kristinsson HG, Lanier TC, Park JW (2005) Process for recovery of functional proteins by pH shifts. In: Park JW (ed) Surimi and surimi seafood. CRC Press, Boca Raton, FL, pp 107–139

    Chapter  Google Scholar 

  67. Gehring CK, Gigliotti JC, Moritz JS, Tou JC, Jaczynski J (2011) Functional and nutritional characteristics of proteins and lipids recovered by isoelectric processing of fish by-products and low-value fish: a review. Food Chem 124:422–431

    Article  CAS  Google Scholar 

  68. Hultin HO, Kelleher SD (1999) Process for isolating a protein composition from a muscle source and protein composition. US Patent 6,005,073

    Google Scholar 

  69. Hultin HO, Kelleher SD (2000) High efficiency alkaline protein extraction. US Patent 6,136,959

    Google Scholar 

  70. Jamilah B, Harvinder KG (2002) Properties of gelatins from skins of fish – black tilapia (Oreochromis mossambicus) and red tilapia (Oreochromis nilotica). J Food Chem 77:81–84

    Article  CAS  Google Scholar 

  71. Karim AA, Bhat R (2009) Fish gelatin: properties, challenges, and prospects as an alternative to mammalian gelatins. Food Hydrocoll 23:563–576

    Article  CAS  Google Scholar 

  72. Aspmo SI, Horn SJ, Eijsink VGH (2005) Enzymatic hydrolysis of Atlantic cod (Gadus morhua L.) viscera. Process Biochem 40:1957–1966

    Article  CAS  Google Scholar 

  73. Batista I, Pires C, Teixeira B, Nunes ML (2014) Hidrolisados proteicos com atividade biológica: uma alternativa para a valorização de subprodutos de pescado. Boletim de Biotecnologia 2ª Série(5):37–39

    Google Scholar 

  74. Guérard F, Dufossé L, De La Broise D, Binet A (2001) Enzymatic hydrolysis of proteins from yellowfin tuna (Thunnus albacares) wastes using alcalase. J Mol Catal B Enzym 11:1051–1059

    Article  Google Scholar 

  75. Kristinsson HG, Rasco BA (2000) Biochemical and functional properties of Atlantic salmon (Salmo salar) muscle proteins hydrolyzed with various alkaline proteases. J Agric Food Chem 48(3):657–666

    Article  PubMed  CAS  Google Scholar 

  76. Pedersen B (1994) Removing of bitterness from protein hydrolysates. Food Technol 45(10):96–98

    Google Scholar 

  77. Nilsang S, Lertsiri S, Suphantharika M, Assavanig A (2005) Optimization of enzymatic hydrolysis of fish soluble concentrate by commercial proteases. J Food Eng 70:571–578

    Article  Google Scholar 

  78. Benjakul S, Yarnpakdee S, Senphan T, Halldorsdottir SM, Kristinsson HG (2014) Fish protein hydrolysates: production, bioactivities and applications. In: Kristinsson HG (ed) Antioxidants and functional components in aquatic foods, 1st edn. Matil Ltd, Reykjavik, Iceland, pp 237–283

    Chapter  Google Scholar 

  79. Guerard F, Decourcelle N, Sabourin C et al (2010) Recent developments of marine ingredients for food and nutraceutical applications: a review. J Sci Hal Aquat 2:21–27

    Google Scholar 

  80. Cardoso C, Nunes ML (2013) Improved utilization of fish waste, discards, and by-products and low-value fish towards food and health products. In: Galvez RP, Bergé J-P (eds) Utilization of fish waste. Taylor & Francis Group, Boca Raton, FL, pp 26–58

    Chapter  Google Scholar 

  81. Vermeirssen V, Van Camp J, Verstraete W (2004) Bioavailability of angiotensin I converting enzyme inhibitory peptides. Br J Nutr 92(3):357–366

    Article  PubMed  CAS  Google Scholar 

  82. López-Fandiño R, Otte J, Van Camp J (2006) Physiological, chemical and technological aspects of milk-protein-derived peptides with antihypertensive and ACE-inhibitory activity. Int Dairy J 16:1277–1293

    Article  CAS  Google Scholar 

  83. Murray BA, FitzGerald RJ (2007) Angiotensin converting enzyme inhibitory peptides derived from food proteins: biochemistry, bioactivity and production. Curr Pharm Des 13(8):773–791

    Article  PubMed  CAS  Google Scholar 

  84. López Expoósito I, Recio I (2006) Antibacterial activity of peptides and folding variants from milk proteins. Int Dairy J 16:1294–1305

    Article  CAS  Google Scholar 

  85. Mendis E, Rajapakse N, Byun HG, Kim SK (2005) Investigation of jumbo squid (Dosidicus gigas) skin gelatine peptides for their in vitro antioxidant effects. Life Sci 77:2166–2178

    Article  PubMed  CAS  Google Scholar 

  86. Sarmadi BH, Ismail A (2010) Antioxidative peptides from food proteins: a review. Peptides 31(10):1949–1956. https://doi.org/10.1016/j.peptides.2010.06.020

    Article  PubMed  CAS  Google Scholar 

  87. Erdmann K, Cheung BW, Schroder H (2008) The possible roles of food-derived bioactive peptides in reducing the risk of cardiovascular disease. J Nutr Biochem 19(10):643–654. https://doi.org/10.1016/j.jnutbio.2007.11.010

    Article  PubMed  CAS  Google Scholar 

  88. Shahidi F, Janak Kamil YVA (2001) Enzymes from fish and aquatic invertebrates and their application in the food industry. Trends Food Sci Technol 12:435–464

    Article  Google Scholar 

  89. Bourseau PL, Vandanjon P, Jaouen M et al (2009) Fractionation of fish protein hydrolysates by ultrafiltration and nanofiltration: impact on peptidic populations. Desalination 244:303–320

    Article  CAS  Google Scholar 

  90. Picot L, Ravallec R, Fouchereau-Peron M et al (2010) Impact of ultrafiltration and nanofiltration of an industrial fish protein hydrolysate on its bioactive properties. J Sci Food Agric 90(11):1819–1826. https://doi.org/10.1002/jsfa.4020

    Article  PubMed  CAS  Google Scholar 

  91. Dutta PK, Dutta J, Tripathi VS (2004) Chitin and chitosan: chemistry, properties and applications. J Scientif Indus Res 63:20–31

    CAS  Google Scholar 

  92. Kamil J, Jeon YJ, Shahidi F (2002) Antioxidant activity of chitosans of different viscosity in cooked comminuted flesh of herring (Clupea harengus). Food Chem 79:69–77

    Article  CAS  Google Scholar 

  93. Park PJ, Je JY, Kim SK (2003) Free radical scavenging activity of chitooligosaccharides by electron spin resonance spectrometry. J Agric Food Chem 51(16):4624–4627. https://doi.org/10.1021/jf034039+

    Article  PubMed  CAS  Google Scholar 

  94. Rosa R, Nunes ML (2008) Crustáceos: Exploração, bioquímica, conservação e aproveitamento de sub-produtos. Publicações Avulsas do Instituto de Investigação das Pescas e do Mar, IPIMAR, Lisboa, Portugal

    Google Scholar 

  95. Chandy T, Sharma CP (1990) Chitosan–as a biomaterial. Biomater Artif Cells Artif Organs 18(1):1–24

    Article  PubMed  CAS  Google Scholar 

  96. Kadam SU, Prabhasankar P (2010) Marine foods as functional ingredients in bakery and pasta products. Food Res Int 43:1975–1980

    Article  Google Scholar 

  97. Hayes M, Carney B, Slater J, Bruck W (2008) Mining marine shellfish wastes for bioactive molecules: chitin and chitosan–Part B: applications. Biotechnol J 3(7):878–889. https://doi.org/10.1002/biot.200800027

    Article  PubMed  CAS  Google Scholar 

  98. Dawczynski C, Schubert R, Jahreis G (2007) Amino acids, fatty acids, and dietary fibre in edible seaweed products. Food Chem 103:891–899

    Article  CAS  Google Scholar 

  99. Mohapatra BR, Bapuji M, Sree A (2003) Production of industrial enzymes (amylase, carboxymethylcellulase and protease) by bacteria isolated from marine sedentary organizms. Acta Biotechnol 23:75–84

    Article  CAS  Google Scholar 

  100. O’Sullivan L, Murphy B, McLoughlin P et al (2010) Prebiotics from marine macroalgae for human and animal health applications. Mar Drugs 8(7):2038–2064. https://doi.org/10.3390/md8072038

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Devillé C, Gharbi M, Dandrifosse G, Peulen O (2007) Study on the effects of laminarin, a polysaccharide from seaweed, on gut characteristics. J Sci Food Agric 87:1717–1725

    Article  CAS  Google Scholar 

  102. Courtois J (2009) Oligosaccharides from land plants and algae: production and applications in therapeutics and biotechnology. Curr Opin Microbiol 12(3):261–273. https://doi.org/10.1016/j.mib.2009.04.007

    Article  PubMed  CAS  Google Scholar 

  103. Wang Y (2009) Prebiotics: present and future in food science and technology. Food Res Int 42:8–12

    Article  CAS  Google Scholar 

  104. Holdt SL, Kraan S (2011) Bioactive compounds in seaweed: functional food applications and legislation. J Appl Phycol 23:543–597

    Article  CAS  Google Scholar 

  105. Kim SK, Li YX (2011) Medicinal benefits of sulfated polysaccharides from sea vegetables. Adv Food Nutr Res 64:391–402. https://doi.org/10.1016/B978-0-12-387669-0.00030-2

    Article  PubMed  CAS  Google Scholar 

  106. Ciferri O (1983) Spirulina, the edible microorganism (algae, single-cell protein). Microbiol Rev 47:551–578

    PubMed  PubMed Central  CAS  Google Scholar 

  107. Lesser MP (2006) Oxidative stress in marine environments: biochemistry and physiological ecology. Annu Rev Physiol 68:253–278. https://doi.org/10.1146/annurev.physiol.68.040104.110001

    Article  PubMed  CAS  Google Scholar 

  108. Kidd P (2011) Astaxanthin, cell membrane nutrient with diverse clinical benefits and anti-aging potential. Altern Med Rev 16(4):355–364

    PubMed  Google Scholar 

  109. Yang Y, Kim B, Lee JY (2013) Astaxanthin structure, metabolism, and health benefits. J Hum Nutr Food Sci 1(1003):1–11

    Google Scholar 

  110. Pashkow FJ, Watumull DG, Campbell CL (2008) Astaxanthin: a novel potential treatment for oxidative stress and inflammation in cardiovascular disease. Am J Cardiol 101(10A):58D–68D. https://doi.org/10.1016/j.amjcard.2008.02.010

    Article  PubMed  CAS  Google Scholar 

  111. Guerin M, Huntley ME, Olaizola M (2003) Haematococcus astaxanthin: applications for human health and nutrition. Trends Biotechnol 21(5):210–216. https://doi.org/10.1016/S0167-7799(03)00078-7

    Article  PubMed  CAS  Google Scholar 

  112. Matsumoto RL, Bastos DH, Mendonca S et al (2009) Effects of mate tea (Ilex paraguariensis) ingestion on mRNA expression of antioxidant enzymes, lipid peroxidation, and total antioxidant status in healthy young women. J Agric Food Chem 57(5):1775–1780. https://doi.org/10.1021/jf803096g

    Article  PubMed  CAS  Google Scholar 

  113. Heo SJ, Park EJ, Lee KW, Jeon YJ (2005) Antioxidant activities of enzymatic extracts from brown seaweeds. Bioresour Technol 96(14):1613–1623. https://doi.org/10.1016/j.biortech.2004.07.013

    Article  PubMed  CAS  Google Scholar 

  114. Cofrades S, Lopez-Lopez I, Bravo L et al (2010) Nutritional and antioxidant properties of different brown and red Spanish edible seaweeds. Food Sci Technol Int/Ciencia y tecnologia de los alimentos internacional 16(5):361–370. https://doi.org/10.1177/1082013210367049

    Article  PubMed  CAS  Google Scholar 

  115. Li Y, Qian ZJ, Ryu B, Lee SH, Kim MM, Kim SK (2009) Chemical components and its antioxidant properties in vitro: an edible marine brown alga, Ecklonia cava. Bioorg Med Chem 17(5):1963–1973. https://doi.org/10.1016/j.bmc.2009.01.031

    Article  PubMed  CAS  Google Scholar 

  116. Artan M, Li Y, Karadeniz F, Lee SH, Kim MM, Kim SK (2008) Anti-HIV-1 activity of phloroglucinol derivative, 6,6′-bieckol, from Ecklonia cava. Bioorg Med Chem 16(17):7921–7926. https://doi.org/10.1016/j.bmc.2008.07.078

    Article  PubMed  CAS  Google Scholar 

  117. Kong CS, Kim JA, Yoon NY, Kim SK (2009) Induction of apoptosis by phloroglucinol derivative from Ecklonia cava in MCF-7 human breast cancer cells. Food Chem Toxicol 47(7):1653–1658. https://doi.org/10.1016/j.fct.2009.04.013

    Article  PubMed  CAS  Google Scholar 

  118. Kim SK, Karadeniz F (2012) Biological importance and applications of squalene and squalane. Adv Food Nutr Res 65:223–233. https://doi.org/10.1016/B978-0-12-416003-3.00014-7

    Article  PubMed  Google Scholar 

  119. Ghimire GP, Lee HC, Sohng JK (2009) Improved squalene production via modulation of the methylerythritol 4-phosphate pathway and heterologous expression of genes from Streptomyces peucetius ATCC 27952 in Escherichia coli. Appl Environ Microbiol 75(22):7291–7293. https://doi.org/10.1128/AEM.01402-09

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Mantzouridou F, Naziri E, Tsimidou MZ (2009) Squalene versus ergosterol formation using Saccharomyces cerevisiae: combined effect of oxygen supply, inoculum size, and fermentation time on yield and selectivity of the bioprocess. J Agric Food Chem 57(14):6189–6198. https://doi.org/10.1021/jf900673n

    Article  PubMed  CAS  Google Scholar 

  121. Copeman LA, Parrish CC (2004) Lipids classes, fatty acids, and sterols in seafood from Gilbert Bay, Southern Labrador. J Agric Food Chem 52:4872–4881

    Article  PubMed  CAS  Google Scholar 

  122. Murphy KJ, Mann NJ, Sinclair AJ (2003) Fatty acid and sterol composition of frozen and freeze-dried New Zealand Green Lipped Mussel (Perna canaliculus) from three sites in New Zealand. Asia Pac J Clin Nutr 12(1):50–60

    PubMed  CAS  Google Scholar 

  123. Rasmussen HE, Blobaum KR, Jesch ED et al (2009) Hypocholesterolemic effect of Nostoc commune var. sphaeroides Kutzing, an edible blue-green alga. Eur J Nutr 48(7):387–394. https://doi.org/10.1007/s00394-009-0025-y

    Article  PubMed  CAS  Google Scholar 

  124. Patridge EV, Gareiss PC, Kinch MS, Hoyer DW (2015) An analysis of original research contributions toward FDA-approved drugs. Drug Discov Today 20(10):1182–1187. https://doi.org/10.1016/j.drudis.2015.06.006

    Article  PubMed  Google Scholar 

  125. de Goeij BECG, Lambert JM (2016) New developments for antibody-drug conjugate-based therapeutic approaches. Curr Opin Immunol 40:14–23. https://doi.org/10.1016/j.coi.2016.02.008

    Article  PubMed  CAS  Google Scholar 

  126. Gerwick WH, Moore BS (2012) Lessons from the past and charting the future of marine natural products drug discovery and chemical biology (vol 19, pg 85, 2012). Chem Biol 19(12):1631. https://doi.org/10.1016/j.chembiol.2012.12.004

    Article  CAS  Google Scholar 

  127. Newman DJ, Cragg GM (2014) Marine-sourced anti-cancer and cancer pain control agents in clinical and late preclinical development. Mar Drugs 12(1):255–278. https://doi.org/10.3390/md12010255

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Mayer AMS (2017) Marine pharmaceuticals: the clinical pipeline. http://marinepharmacology.midwestern.edu/clinPipeline.htm. Accessed 30 May 2017

  129. FDA (2016) Drugs@FDA: FDA approved drug products. http://www.accessdata.fda.gov/scripts/cder/daf/. Accessed 30 Dec 2016

  130. EMA (2016) Medicines. http://wwwemaeuropaeu/ema/indexjsp?curl=pages/includes/medicines/medicines_landing_pagejsp&mid=. Accessed 30 Dec 2016

  131. Lichtman MA (2013) A historical perspective on the development of the cytarabine (7days) and daunorubicin (3days) treatment regimen for acute myelogenous leukemia: 2013 the 40th anniversary of 7+3. Blood Cells Mol Dis 50(2):119–130. https://doi.org/10.1016/j.bcmd.2012.10.005

    Article  PubMed  CAS  Google Scholar 

  132. Shankland KR, Armitage JO, Hancock BW (2012) Non-Hodgkin lymphoma. Lancet 380(9844):848–857. https://doi.org/10.1016/S0140-6736(12)60605-9

    Article  PubMed  Google Scholar 

  133. Cassidy S, Syed BA (2016) Acute myeloid leukaemia drugs market. Nat Rev Drug Discov 15(8):527–528. https://doi.org/10.1038/nrd.2016.140

    Article  PubMed  CAS  Google Scholar 

  134. Mayer AMS, Glaser KB, Cuevas C et al (2010) The odyssey of marine pharmaceuticals: a current pipeline perspective. Trends Pharmacol Sci 31(6):255–265. https://doi.org/10.1016/j.tips.2010.02.005

    Article  PubMed  CAS  Google Scholar 

  135. Chhikara BS, Parang K (2010) Development of cytarabine prodrugs and delivery systems for leukemia treatment. Expert Opin Drug Del 7(12):1399–1414. https://doi.org/10.1517/17425247.2010.527330

    Article  CAS  Google Scholar 

  136. Kripp M, Hofheinz RD (2008) Treatment of lymphomatous and leukemic meningitis with liposomal encapsulated cytarabine. Int J Nanomedicine 3(4):397–401

    PubMed  PubMed Central  CAS  Google Scholar 

  137. National Cancer Institute. In: Clinical Trials (PDQ®). National Cancer Institute. Accessed 1 Nov 2013

    Google Scholar 

  138. Pharma C. Clavis Pharma announces negative outcome of phase III CLAVELA trial with elacytarabine in patients with acute myeloid leukaemia. http://aqualis.no/home. Accessed 1 Nov 2013

  139. Gandhi V, Keating MJ, Bate G, Kirkpatrick P (2006) Nelarabine. Nat Rev Drug Discov 5(1):17–18

    Article  PubMed  CAS  Google Scholar 

  140. Shen W, Kim JS, Kish PE et al (2009) Design and synthesis of vidarabine prodrugs as antiviral agents. Bioorg Med Chem Lett 19(3):792–796. https://doi.org/10.1016/j.bmcl.2008.12.031

    Article  PubMed  CAS  Google Scholar 

  141. Cimino G, Derosa S, Destefano S (1984) Antiviral agents from a Gorgonian, Eunicella-Cavolini. Experientia 40(4):339–340. https://doi.org/10.1007/Bf01952539

    Article  CAS  Google Scholar 

  142. Montgomery J, Hewson K (1969) Nucleosides of 2-Fluoroadenine. J Med Chem 12(3):498–504

    Article  PubMed  CAS  Google Scholar 

  143. Robak P, Robak T (2013) Older and new purine nucleoside analogs for patients with acute leukemias. Cancer Treat Rev 39(8):851–861

    Article  PubMed  CAS  Google Scholar 

  144. Ricci F, Tedeschi A, Morra E, Montillo M (2009) Fludarabine in the treatment of chronic lymphocytic leukemia: a review. Ther Clin Risk Manag 5:187–207

    PubMed  PubMed Central  CAS  Google Scholar 

  145. Lapponi MJ, Rivero CW, Zinni MA, Britos CN, Trelles JA (2016) New developments in nucleoside analogues biosynthesis: a review. J Mol Catal B Enzym 133:218–233

    Article  CAS  Google Scholar 

  146. Hernandez-Ilizaliturri FJ, Czuczman MS (2009) A review of nelarabine in the treatment of T-cell lymphoblastic leukemia/lymphoma. Clin Med Ther 1:CMT.S1954

    Article  Google Scholar 

  147. Cohen MH, Johnson JR, Justice R, Pazdur R (2008) FDA drug approval summary: nelarabine (Arranon(R)) for the treatment of T-Cell lymphoblastic leukemia/lymphoma. Oncologist 13(6):709–714

    Article  PubMed  CAS  Google Scholar 

  148. Bauer A, Bronstrupt M (2014) Industrial natural product chemistry for drug discovery and development. Nat Prod Rep 31(1):35–60. https://doi.org/10.1039/c3np70058e

    Article  PubMed  CAS  Google Scholar 

  149. Esai C (2016) http://www.eisai.com. Accessed 6 Dec 2016

  150. Duggan PJ, Tuck KL (2015) Bioactive mimetics of conotoxins and other venom peptides. Toxins 7(10):4175–4198. https://doi.org/10.3390/toxins7104175

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  151. Nelson L (2004) One slip, and you’re dead…. Nature 429(6994):798–799. https://doi.org/10.1038/429798a

    Article  PubMed  CAS  Google Scholar 

  152. Han TS, Teichert RW, Olivera BM, Bulaj G (2008) Conus venoms – a rich source of peptide-based therapeutics. Curr Pharm Des 14(24):2462–2479. https://doi.org/10.2174/138161208785777469

    Article  PubMed  CAS  Google Scholar 

  153. Oliveira BM (2000) ω-conotoxin MVIIA: from marine snail venom to analgesic drug. In: Fusetani N (ed) Drugs from the sea. Karger, Basel; New York, NY, pp 74–85

    Chapter  Google Scholar 

  154. Bingham JP, Mitsunaga E, Bergeron ZL (2010) Drugs from slugs-past, present and future perspectives of omega-conotoxin research. Chem Biol Interact 183(1):1–18. https://doi.org/10.1016/j.cbi.2009.09.021

    Article  PubMed  CAS  Google Scholar 

  155. Brady RM, Zhang MM, Gable R, Norton RS, Baell JB (2013) De novo design and synthesis of a mu-conotoxin KIIIA peptidomimetic. Bioorg Med Chem Lett 23(17):4892–4895. https://doi.org/10.1016/j.bmcl.2013.06.086

    Article  PubMed  CAS  Google Scholar 

  156. CORDIS (2016) VENOMICS report summary. http://cordis.europa.eu/result/rcn/182082_en.html. Accessed 6 Dec 2016

  157. Bays H (2006) Clinical overview of omacor: a concentrated formulation of omega-3 polyunsaturated fatty acids. Am J Cardiol 98(4A):71i–76i. https://doi.org/10.1016/j.amjcard.2005.12.029

    Article  PubMed  CAS  Google Scholar 

  158. Gluck T, Alter P (2016) Marine omega-3 highly unsaturated fatty acids: from mechanisms to clinical implications in heart failure and arrhythmias. Vasc Pharmacol 82:11–19. https://doi.org/10.1016/j.vph.2016.03.007

    Article  CAS  Google Scholar 

  159. Rupp H (2009) OmacorA (R) (prescription omega-3-acid ethyl esters 90): from severe rhythm disorders to hypertriglyceridemia. Adv Ther 26(7):675–690. https://doi.org/10.1007/s12325-009-0045-2

    Article  PubMed  CAS  Google Scholar 

  160. Koski RR (2008) Omega-3-acid ethyl esters (lovaza) for severe hypertriglyceridemia. Pharm Therap 33(5):271–303

    Google Scholar 

  161. Glueck CJ, Khan N, Riaz M, Padda J, Khan Z, Wang P (2012) Titrating lovaza from 4 to 8 to 12 grams/day in patients with primary hypertriglyceridemia who had triglyceride levels > 500 mg/dl despite conventional triglyceride lowering therapy. Lipids Health Dis 11. doi:https://doi.org/10.1186/1476-511x-11-143

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  162. Backes J, Anzalone D, Hilleman D, Catini J (2016) The clinical relevance of omega-3 fatty acids in the management of hypertriglyceridemia. Lipids Health Dis 15. doi:https://doi.org/10.1186/S12944-016-0286-4

  163. D’Incalci M, Galmarini CM (2010) A review of trabectedin (ET-743): A unique mechanism of action. Mol Cancer Ther 9(8):2157–2163. https://doi.org/10.1158/1535-7163.MCT-10-0263

    Article  PubMed  CAS  Google Scholar 

  164. Larsen AK, Galmarini CM, D’Incalci M (2016) Unique features of trabectedin mechanism of action. Cancer Chemother Pharmacol 77(4):663–671. https://doi.org/10.1007/s00280-015-2918-1

    Article  PubMed  CAS  Google Scholar 

  165. D’Incalci M, Badri N, Galmarini CM, Allavena P (2014) Trabectedin, a drug acting on both cancer cells and the tumour microenvironment. Br J Cancer 111(4):646–650. https://doi.org/10.1038/bjc.2014.149

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  166. Rinehart KL (2000) Antitumor compounds from tunicates. Med Res Rev 20(1):1–27

    Article  PubMed  CAS  Google Scholar 

  167. Molinski TF, Dalisay DS, Lievens SL, Saludes JP (2009) Drug development from marine natural products. Nat Rev Drug Discov 8(1):69–85. https://doi.org/10.1038/nrd2487

    Article  PubMed  CAS  Google Scholar 

  168. Cuevas C, Francesch A (2009) Development of Yondelis (R) (trabectedin, ET-743). A semisynthetic process solves the supply problem. Nat Prod Rep 26(3):322–337. https://doi.org/10.1039/b808331m

    Article  PubMed  CAS  Google Scholar 

  169. Rinehart KL, Holt TG, Fregeau NL et al (1990) Isolation and characterization of the ecteinascidins, potent antitumor compounds from the Caribbean Tunicate Ecteinascidia-Turbinata. Abstr Pap Am Chem S 200:141-ORGN

    Google Scholar 

  170. Wright AE, Forleo DA, Gunawardana GP, Gunasekera SP, Koehn FE, Mcconnell OJ (1990) Antitumor tetrahydroisoquinoline alkaloids from the colonial Ascidian Ecteinascidia-Turbinata. J Org Chem 55(15):4508–4512. https://doi.org/10.1021/Jo00302a006

    Article  CAS  Google Scholar 

  171. Corey EJ, Gin DY, Kania RS (1996) Enantioselective total synthesis of ecteinascidin 743. J Am Chem Soc 118(38):9202–9203. https://doi.org/10.1021/Ja962480t

    Article  CAS  Google Scholar 

  172. Martinez EJ, Corey EJ (2000) A new, more efficient, and effective process for the synthesis of a key pentacyclic intermediate for production of ecteinascidin and phthalascidin antitumor agents. Org Lett 2(7):993–996. https://doi.org/10.1021/Ol0056729

    Article  PubMed  CAS  Google Scholar 

  173. Cuevas C, Perez M, Martin MJ et al (2000) Synthesis of ecteinascidin ET-743 and phthalascidin Pt-650 from cyanosafracin B. Org Lett 2(16):2545–2548. https://doi.org/10.1021/Ol0062502

    Article  PubMed  CAS  Google Scholar 

  174. Velasco IA, De La Calle F, Aparicio PT et al (2004) The gene cluster involved in safracin biosynthesis and its uses for genetic engineering. Google Patents

    Google Scholar 

  175. Menchaca R, Martinez V, Rodriguez A et al (2003) Synthesis of natural ecteinascidins (ET-729, ET-745, ET-759B, ET-736, ET-637, ET-594) from cyanosafracin B. J Org Chem 68(23):8859–8866. https://doi.org/10.1021/jo034547i

    Article  PubMed  CAS  Google Scholar 

  176. Manzanares I, Cuevas C, García-Nieto R, Marco E, Gago F (2001) Advances in the chemistry and pharmacology of ecteinascidins, a promising new class of anti-cancer agents. Curr Med Chem Anticancer Agents 1(3):257–276. https://doi.org/10.2174/1568011013354561

    Article  PubMed  CAS  Google Scholar 

  177. Goodin S, Barbour S, Song J, Berrak E, Cox D (2015) Safety and tolerability of eribulin mesylate in patients with pretreated metastatic breast cancer. Am J Health-Syst Pharm 72(24):2150–2156. https://doi.org/10.2146/ajhp140773

    Article  PubMed  CAS  Google Scholar 

  178. FDA (2016) FDA approves first drug to show survival benefit in liposarcoma. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm483714.htm. Accessed Dec 2016

  179. Bai R, Paull KD, Herald CL, Malspeis L, Pettit GR, Hamel E (1991) Halichondrin-B and homohalichondrin-B, marine natural-products binding in the vinca domain of tubulin – discovery of tubulin-based mechanism of action by analysis of differential cytotoxicity data. J Biol Chem 266(24):15882–15889

    PubMed  CAS  Google Scholar 

  180. Dybdal-Hargreaves NF, Risinger AL, Mooberry SL (2015) Eribulin Mesylate: mechanism of action of a unique microtubule-targeting agent. Clin Cancer Res 21(11):2445–2452. https://doi.org/10.1158/1078-0432.CCR-14-3252

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  181. Bauer A (2016) Story of Eribulin Mesylate: development of the longest drug synthesis. In: Časar Z (ed) Synthesis of heterocycles in contemporary medicinal chemistry, Topics in heterocyclic chemistry, vol 44. Springer, Switzerland, pp 209–270

    Chapter  Google Scholar 

  182. Aicher TD, Buszek KR, Fang FG et al (1992) Total synthesis of halichondrin-B and norhalichondrin-B. J Am Chem Soc 114(8):3162–3164. https://doi.org/10.1021/Ja00034a086

    Article  CAS  Google Scholar 

  183. Towle MJ, Salvato KA, Budrow J et al (2001) In vitro and in vivo anticancer activities of synthetic macrocyclic ketone analogues of halichondrin B. Cancer Res 61(3):1013–1021

    PubMed  CAS  Google Scholar 

  184. Yu MJ, Zheng WJ, Seletsky BM (2013) From micrograms to grams: scale-up synthesis of eribulin mesylate. Nat Prod Rep 30(9):1158–1164. https://doi.org/10.1039/c3np70051h

    Article  CAS  PubMed  Google Scholar 

  185. Liu KKC, Sakya SM, O’Donnell CJ, Flick AC, Ding HX (2012) Synthetic approaches to the 2010 new drugs. Bioorg Med Chem 20(3):1155–1174. https://doi.org/10.1016/j.bmc.2011.12.049

    Article  PubMed  CAS  Google Scholar 

  186. Newland AM, Li JX, Wasco LE, Aziz MT, Lowe DK (2013) Brentuximab vedotin: a CD30-directed antibody-cytotoxic drug conjugate. Pharmacotherapy 33(1):93–104. https://doi.org/10.1002/phar.1170

    Article  PubMed  CAS  Google Scholar 

  187. Gerber HP, Koehn FE, Abraham RT (2013) The antibody-drug conjugate: an enabling modality for natural product-based cancer therapeutics. Nat Prod Rep 30(5):625–639. https://doi.org/10.1039/c3np20113a

    Article  PubMed  CAS  Google Scholar 

  188. Ansell SM (2011) Brentuximab vedotin: delivering an antimitotic drug to activated lymphoma cells. Expert Opin Investig Drugs 20(1):99–105. https://doi.org/10.1517/13543784.2011.542147

    Article  PubMed  CAS  Google Scholar 

  189. Pettit GR, Kamano Y, Herald CL et al (1987) Antineoplastic agents 136. The isolation and structure of a remarkable marine animal antineoplastic constituent – Dolastatin 10. J Am Chem Soc 109(22):6883–6885. https://doi.org/10.1021/Ja00256a070

    Article  CAS  Google Scholar 

  190. Maderna A, Doroski M, Subramanyam C et al (2014) Discovery of cytotoxic Dolastatin 10 analogues with N-terminal modifications. J Med Chem 57(24):10527–10543. https://doi.org/10.1021/jm501649k

    Article  PubMed  CAS  Google Scholar 

  191. Pettit GR, Singh SB, Hogan F et al (1989) Antineoplastic agents 189. The absolute-configuration and synthesis of natural (-)-Dolastatin-10. J Am Chem Soc 111(14):5463–5465. https://doi.org/10.1021/Ja00196a061

    Article  CAS  Google Scholar 

  192. Luesch H, Moore RE, Paul VJ, Mooberry SL, Corbett TH (2001) Isolation of dolastatin 10 from the marine cyanobacterium Symploca species VP642 and total stereochemistry and biological evaluation of its analogue symplostatin 1. J Nat Prod 64(7):907–910. https://doi.org/10.1021/np010049y

    Article  PubMed  CAS  Google Scholar 

  193. Senter PD, Sievers EL (2012) The discovery and development of brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic anaplastic large cell lymphoma. Nat Biotechnol 30(7):631–637. https://doi.org/10.1038/nbt.2289

    Article  PubMed  CAS  Google Scholar 

  194. FDA (2016) ClinicalTrials.gov. https://clinicaltrials.gov/. Accessed 30 Dec 2016

  195. EMA (2016) EU clinical trials register. https://wwwclinicaltrialsregistereu/ctr-search/search. Accessed 30 Dec 2016

  196. Leibbrandt A, Meier C, Konig-Schuster M et al (2010) Iota-Carrageenan is a potent inhibitor of influenza A virus infection. PLoS One 5(12). https://doi.org/10.1371/journal.pone.0014320

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  197. Eccles R, Meier C, Jawad M, Weinmullner R, Grassauer A, Prieschl-Grassauer E (2010) Efficacy and safety of an antiviral Iota-Carrageenan nasal spray: a randomized, double-blind, placebo-controlled exploratory study in volunteers with early symptoms of the common cold. Resp Res 11. https://doi.org/10.1186/1465-9921-11-108

  198. Gonzalez ME, Alarcon B, Carrasco L (1987) Polysaccharides as antiviral agents – antiviral activity of Carrageenan. Antimicrob Agents Chemother 31(9):1388–1393

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  199. Rao MN, Shinnar AE, Noecker LA et al (2000) Aminosterols from the dogfish shark Squalus-acanthias. J Nat Prod 63(5):631–635. https://doi.org/10.1021/Np990514f

    Article  PubMed  CAS  Google Scholar 

  200. Noguchi T, Arakawa O (2008) Tetrodotoxin – distribution and accumulation in aquatic organisms, and cases of human intoxication. Mar Drugs 6(2):220–242. https://doi.org/10.3390/md20080011

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Pettit GR, Herald CL, Doubek DL, Herald DL, Arnold E, Clardy J (1982) Anti-neoplastic agents 86. Isolation and structure of bryostatin-1. J Am Chem Soc 104(24):6846–6848. https://doi.org/10.1021/Ja00388a092

    Article  CAS  Google Scholar 

  202. Martin MJ, Coello L, Fernandez R et al (2013) Isolation and first total synthesis of PM050489 and PM060184, two new marine anticancer compounds. J Am Chem Soc 135(27):10164–10171. https://doi.org/10.1021/ja404578u

    Article  PubMed  CAS  Google Scholar 

  203. Feling RH, Buchanan GO, Mincer TJ, Kauffman CA, Jensen PR, Fenical W (2003) Salinosporamide A: a highly cytotoxic proteasome inhibitor from a novel microbial source, a marine bacterium of the new genus Salinospora. Angew Chem Int Ed 42(3):355. https://doi.org/10.1002/anie.200390115

    Article  CAS  Google Scholar 

  204. Fenical W, Jensen PR, Cheng XC (2000) Halimide, a cytotoxic marine natural product, and derivatives thereof. Google Patents

    Google Scholar 

  205. Kem W, Soti F, Wildeboer K et al (2006) The nemertine toxin anabaseine and its derivative DMXBA (GTS21): chemical and pharmacological properties. Mar Drugs 4(3):255–273. https://doi.org/10.3390/Md403255

    Article  PubMed Central  CAS  Google Scholar 

  206. Thomas NV, Kim SK (2013) Beneficial effects of marine algal compounds in cosmeceuticals. Mar Drugs 11(1):146–164. https://doi.org/10.3390/md11010146

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  207. Kijjoa ASP (2004) Drugs and cosmetics from the sea. Mar Drugs 2:2

    Article  Google Scholar 

  208. Kim YH, Chung CB, Kim JG et al (2008) Anti-wrinkle activity of ziyuglycoside I isolated from a Sanguisorba officinalis root extract and its application as a cosmeceutical ingredient. Biosci Biotechnol Biochem 72(2):303–311. https://doi.org/10.1271/bbb.70268

    Article  PubMed  CAS  Google Scholar 

  209. Raposo MF, de Morais RM, Bernardo de Morais AM (2013) Bioactivity and applications of sulphated polysaccharides from marine microalgae. Mar Drugs 11(1):233–252. https://doi.org/10.3390/md11010233

    Article  PubMed  Google Scholar 

  210. LIPOTEC (2016) LIPOTEC CATALOGUE. In: LIPOTEC. http://www.lipotec.com. Accessed 7 Jan 2017

  211. Wire B (2016) http://www.businesswire.com/news/home/20160323006461/en/Global-Cosmeceuticals-Market-Worth-USD-61-Billion. Accessed 5 Jan 2017

  212. Gupta S, Abu-Ghannam N (2011) Bioactive potential and possible health effects of edible brown seaweeds. Trends Food Sci Technol 22(6):315–326. https://doi.org/10.1016/j.tifs.2011.03.011

    Article  CAS  Google Scholar 

  213. Li Y-X, Wijesekara I, Li Y, Kim S-K (2011) Phlorotannins as bioactive agents from brown algae. Process Biochem 46(12):2219–2224

    Article  CAS  Google Scholar 

  214. Wijesekara I, Pangestuti R, Kim S-K (2011) Biological activities and potential health benefits of sulfated polysaccharides derived from marine algae. Carbohydr Polym 84(1):14–21. https://doi.org/10.1016/j.carbpol.2010.10.062

    Article  CAS  Google Scholar 

  215. Lee JC, Hou MF, Huang HW et al (2013) Marine algal natural products with anti-oxidative, anti-inflammatory, and anti-cancer properties. Cancer Cell Int 13(1):55. https://doi.org/10.1186/1475-2867-13-55

    Article  PubMed  PubMed Central  Google Scholar 

  216. SEPPIC (2016) SEPPIC CATALOGUE. In: SEPPIC GROUP. http://www.seppic.com/products-@/3701/view-3708-category.html. Accessed 6 Jan 2016

  217. CODIF (2016) DERMOCHLORELLA-D. In: CODIF. http://www.codif-tn.com/en/principesactifs/dermochlorella-d/. Accessed 6 Jan 2017

  218. Stolz P, Obermayer B (2005) Manufacturing microalgae for skin care. Cosmet Toilet 120:7

    Google Scholar 

  219. GREENSEA (2016) An unlimited mine of innovation. In: GREENSEA. http://greensea.fr/en/active-ingredients. Accessed 6 Jan 2017

  220. ALGENIST (2016) Patented breakthrough ingredient. In: ALGENIST. https://algenist.borderfree.com/-ingredient. Accessed 6 Jan 2017

  221. FRUTAROM (2016) Frutarom. https://products.frutarom.com/apex/f?p=105:1:0. Accessed 6 Jan 2017

  222. Chi Z, Fang Y (2005) Exopolysaccharides from marine bacteria. J Ocean Univ China 4(1):67–74. https://doi.org/10.1007/s11802-005-0026-2

    Article  CAS  Google Scholar 

  223. Nichols CA, Guezennec J, Bowman JP (2005) Bacterial exopolysaccharides from extreme marine environments with special consideration of the southern ocean, sea ice, and deep-sea hydrothermal vents: a review. Mar Biotechnol (New York, NY) 7(4):253–271. https://doi.org/10.1007/s10126-004-5118-2

    Article  CAS  Google Scholar 

  224. Cambon-Bonavita MA, Raguenes G, Jean J, Vincent P, Guezennec J (2002) A novel polymer produced by a bacterium isolated from a deep-sea hydrothermal vent polychaete annelid. J Appl Microbiol 93(2):310–315

    Article  PubMed  CAS  Google Scholar 

  225. Raguenes G, Christen R, Guezennec J, Pignet P, Barbier G (1997) Vibrio diabolicus sp. nov., a new polysaccharide-secreting organism isolated from a deep-sea hydrothermal vent polychaete annelid, Alvinella pompejana. Int J Syst Bacteriol 47(4):989–995. https://doi.org/10.1099/00207713-47-4-989

    Article  PubMed  CAS  Google Scholar 

  226. Raguenes G, Pignet P, Gauthier G et al (1996) Description of a new polymer-secreting bacterium from a deep-sea hydrothermal vent, Alteromonas macleodii subsp. fijiensis, and preliminary characterization of the polymer. Appl Environ Microbiol 62(1):67–73

    PubMed  PubMed Central  CAS  Google Scholar 

  227. Raguenes GH, Peres A, Ruimy R et al (1997) Alteromonas infernus sp. nov., a new polysaccharide-producing bacterium isolated from a deep-sea hydrothermal vent. J Appl Microbiol 82(4):422–430

    Article  PubMed  CAS  Google Scholar 

  228. Rougeaux HG, Guezennec J, Carlson RW, Kervarec N, Pichon R, Talaga P (1999) Structural determination of the exopolysaccharide of Pseudoalteromonas strain HYD 721 isolated from a deep-sea hydrothermal vent. Carbohydr Res 315:12

    Article  Google Scholar 

  229. Vincent P, Pignet P, Talmont F et al (1994) Production and characterization of an exopolysaccharide excreted by a deep-sea hydrothermal vent bacterium isolated from the Polychaete Annelid Alvinella pompejana. Appl Environ Microbiol 60(11):4134–4141

    PubMed  PubMed Central  CAS  Google Scholar 

  230. Weiner R, Langille S, Quintero E (1995) Structure, function and immunochemistry of bacterial exopolysaccharides. J Ind Microbiol 15(4):339–346

    Article  PubMed  CAS  Google Scholar 

  231. Desbruyeres DL, Laubier L (1980) Alvinella pompejana gen. sp. nov., aberrant Ampharetidae from East Pacific Rise hydrothermal vents. Oceanol Acta 3:267–274

    Google Scholar 

  232. Le Costaouec T, Cerantola S, Ropartz D et al (2012) Structural data on a bacterial exopolysaccharide produced by a deep-sea Alteromonas macleodii strain. Carbohydr Polym 90(1):49–59. https://doi.org/10.1016/j.carbpol.2012.04.059

    Article  PubMed  CAS  Google Scholar 

  233. Thibodeau AT (2006) The applications and functions of new exopolysaccharide “Deepsane” from the deepest oceans. Fragr J 34:7

    Google Scholar 

  234. Potts BC, Faulkner DJ, Jacobs RS (1992) Phospholipase A2 inhibitors from marine organisms. J Nat Prod 55(12):1701–1717

    Article  PubMed  CAS  Google Scholar 

  235. Day DR, Jabaiah S, Jacobs RS, Little RD (2013) Cyclodextrin formulation of the marine natural product pseudopterosin A uncovers optimal pharmacodynamics in proliferation studies of human umbilical vein endothelial cells. Mar Drugs 11:12

    Article  CAS  Google Scholar 

  236. Rouhi AM (2003) Betting on natural products for cures. Chem Eng News 81:10

    Google Scholar 

  237. Hee SW, Tsai SH, Chang YC et al (2012) The role of nocturnin in early adipogenesis and modulation of systemic insulin resistance in human. Obesity (Silver Spring, MD) 20(8):1558–1565. https://doi.org/10.1038/oby.2012.37

    Article  CAS  Google Scholar 

  238. Stubblefield JJ, Terrien J, Green CB (2012) Nocturnin: at the crossroads of clocks and metabolism. Trends Endocrinol Metab 23(7):326–333. https://doi.org/10.1016/j.tem.2012.03.007

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  239. Ivatt RJ (1984) The biology of glycoproteins. Plenum Press, New York, NY

    Book  Google Scholar 

  240. Gottschalk A (1972) Glycoproteins: their composition, structure and function. Elsevier, New York, NY

    Google Scholar 

  241. Senaratne LS, Park PJ, Kim SK (2006) Isolation and characterization of collagen from brown backed toadfish (Lagocephalus gloveri) skin. Bioresour Technol 97(2):191–197. https://doi.org/10.1016/j.biortech.2005.02.024

    Article  PubMed  CAS  Google Scholar 

  242. Pettit RK (2011) Culturability and secondary metabolite diversity of extreme microbes: expanding contribution of deep sea and deep-sea vent microbes to natural product discovery. Mar Biotechnol 13(1):1–11. https://doi.org/10.1007/s10126-010-9294-y

    Article  CAS  Google Scholar 

  243. Harvey A (2000) Strategies for discovering drugs from previously unexplored natural products. Drug Discov Today 5(7):294–300. https://doi.org/10.1016/S1359-6446(00)01511-7

    Article  PubMed  CAS  Google Scholar 

  244. Lallier LE, McMeel O, Greiber T, Vanagt T, Dobson ADW, Jaspars M (2014) Access to and use of marine genetic resources: understanding the legal framework. Nat Prod Rep 31(5):612–616. https://doi.org/10.1039/c3np70123a

    Article  PubMed  CAS  Google Scholar 

  245. Dias DA, Urban S, Roessner U (2012) A historical overview of natural products in drug discovery. Metabolites 2(2):303–336

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  246. Cragg GM, Katz F, David JNA, Rosenthal J (2012) The impact of the United Nations Convention on Biological Diversity on natural products research. Nat Prod Rep 29(12):1407–1423. https://doi.org/10.1039/c2np20091k

    Article  PubMed  CAS  Google Scholar 

  247. Aruoma OI (2006) The impact of food regulation on the food supply chain. Toxicology 221(1):119–127. https://doi.org/10.1016/j.tox.2005.12.024

    Article  PubMed  CAS  Google Scholar 

  248. Byrne D (2003) Health nutrition and labeling. Food Sci Technol 17:26–28

    Google Scholar 

  249. David B, Wolfender JL, Dias DA (2015) The pharmaceutical industry and natural products: historical status and new trends. Phytochem Rev 14(2):299–315. https://doi.org/10.1007/s11101-014-9367-z

    Article  CAS  Google Scholar 

  250. Hughes JP, Rees S, Kalindjian SB, Philpott KL (2011) Principles of early drug discovery. Br J Pharmacol 162(6):1239–1249. https://doi.org/10.1111/j.1476-5381.2010.01127.x

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  251. Kingston DGI (2011) Modern natural products drug discovery and its relevance to biodiversity conservation. J Nat Prod 74(3):496–511. https://doi.org/10.1021/np100550t

    Article  PubMed  CAS  Google Scholar 

  252. Carter GT (2011) Natural products and Pharma 2011: strategic changes spur new opportunities. Nat Prod Rep 28(11):1783–1789. https://doi.org/10.1039/c1np00033k

    Article  PubMed  CAS  Google Scholar 

  253. Perez-Victoria I, Martin J, Reyes F (2016) Combined LC/UV/MS and NMR strategies for the dereplication of marine natural products. Planta Med 82(9–10):857–871. https://doi.org/10.1055/s-0042-101763

    Article  PubMed  CAS  Google Scholar 

  254. Radjasa OK, Vaske YM, Navarro G et al (2011) Highlights of marine invertebrate-derived biosynthetic products: their biomedical potential and possible production by microbial associants. Bioorg Med Chem 19(22):6658–6674. https://doi.org/10.1016/j.bmc.2011.07.017

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  255. Hubert J, Nuzillard J-M, Renault J-H (2015) Dereplication strategies in natural product research: how many tools and methodologies behind the same concept? Phytochem Rev. https://doi.org/10.1007/s11101-015-9448-7

  256. Gaudêncio SP, Pereira F (2015) Dereplication: racing to speed up the natural products discovery process. Nat Prod Rep 32(6):779–810. https://doi.org/10.1039/c4np00134f

    Article  PubMed  CAS  Google Scholar 

  257. Michel T, Halabalaki M, Skaltsounis AL (2013) New concepts, experimental approaches, and dereplication strategies for the discovery of novel phytoestrogens from natural sources. Planta Med 79(7):514–532. https://doi.org/10.1055/s-0032-1328300

    Article  PubMed  CAS  Google Scholar 

  258. Petersen F, Amstutz R (2008) Natural compounds as drugs. Birkhäuser, Basel

    Book  Google Scholar 

Download references

Acknowledgements

This work was financed by national funds through the Portuguese Foundation for Science and Technology (Fundação para a Ciência e Tecnologia) within the scope of projects UID/Multi/04046/2013 (BioISI, Biosystems and Integrative Sciences Institute) UID/Multi/00612/2013 (CQB, Chemistry and Biochemistry Centre) and UID/MAR/04292/2013 (MARE—Marine and Environmental Sciences Centre). Thanks are also due to FCT/MEC through national funds and the co-funding by the FEDER, within the PT2020 Partnership Agreement and Compete 2020 for the financial support to CESAM (UID/AMB/50017/2013).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Helena Vieira .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG, part of Springer Nature

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Calado, R. et al. (2018). How to Succeed in Marketing Marine Natural Products for Nutraceutical, Pharmaceutical and Cosmeceutical Markets. In: Rampelotto, P., Trincone, A. (eds) Grand Challenges in Marine Biotechnology. Grand Challenges in Biology and Biotechnology. Springer, Cham. https://doi.org/10.1007/978-3-319-69075-9_9

Download citation

Publish with us

Policies and ethics