Skip to main content

The Promise of Personalized Medicine

  • Chapter
  • First Online:
  • 979 Accesses

Abstract

There is substantial potential for the human intestinal microbiota to be mined for its therapeutic potential. Absent exogenous perturbations, the intestinal microbiota tends to be highly stable throughout adult life, and it appears to be associated with a variety of rheumatic diseases. Furthermore, the microbiota can influence the metabolism of many medicines used to treat inflammatory diseases, thus potentially playing a role in the disease process even in the absence of specific abnormalities in its composition. We have multiple tools at hand to alter the microbiota, and the future may hold targeted approaches to remove specific bacteria associated with a particular condition. Finally, there is evidence that an individual’s baseline microbiota might predict his or her response to dietary therapy, thus ushering in an era of individualized medicine targeting the human intestinal microbiota.

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

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   199.99
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

Learn about institutional subscriptions

Abbreviations

AID:

Anti-inflammatory diet

CGR:

Cardiac glycoside reductase

EEN:

Exclusive enteral nutrition

FMT:

Fecal microbial transplantation

HMP:

Human Microbiome Project

IBD:

Inflammatory bowel disease

JIA:

Juvenile idiopathic arthritis

RA:

Rheumatoid arthritis

ReA:

Reactive arthritis

SCFA:

Short-chain fatty acids

SpA:

Spondyloarthritis

References

  1. Clunie GP, Lennard L. Relevance of thiopurine methyltransferase status in rheumatology patients receiving azathioprine. Rheumatology (Oxford). 2004;43(1):13–8.

    Article  CAS  Google Scholar 

  2. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Brewerton DA, Hart FD, Nicholls A, Caffrey M, James DC, Sturrock RD. Ankylosing spondylitis and HL-A 27. Lancet. 1973;1(7809):904–7.

    Article  CAS  PubMed  Google Scholar 

  4. Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut Microbiome during the first year of life. Cell Host Microbe. 2015;17(6):852.

    Article  CAS  PubMed  Google Scholar 

  5. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A. 2010;107(26):11971–5.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hollister EB, Riehle K, Luna RA, Weidler EM, Rubio-Gonzales M, Mistretta TA, et al. Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome. 2015;3:36.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009;326(5960):1694–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, et al. The long-term stability of the human gut microbiota. Science. 2013;341(6141):1237439.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Franzosa EA, Huang K, Meadow JF, Gevers D, Lemon KP, Bohannan BJ, et al. Identifying personal microbiomes using metagenomic codes. Proc Natl Acad Sci U S A. 2015;112(22):E2930–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Taurog JD, Richardson JA, Croft JT, Simmons WA, Zhou M, Fernandez-Sueiro JL, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med. 1994;180(6):2359–64.

    Article  CAS  PubMed  Google Scholar 

  12. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32(6):815–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Berer K, Mues M, Koutrolos M, Rasbi ZA, Boziki M, Johner C, et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 2011;479(7374):538–41.

    Article  CAS  PubMed  Google Scholar 

  14. Vieira AT, Macia L, Galvao I, Martins FS, Canesso MC, Amaral FA, et al. A role for gut Microbiota and the metabolite-sensing receptor GPR43 in a murine model of gout. Arthritis Rheumatol. 2015;67(6):1646–56.

    Article  CAS  PubMed  Google Scholar 

  15. Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009;15(8):1183–9.

    Article  CAS  PubMed  Google Scholar 

  16. Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2013;63(8):1275–83.

    Article  CAS  PubMed  Google Scholar 

  17. Stoll ML, Kumar R, Morrow CD, Lefkowitz EJ, Cui X, Genin A, et al. Altered microbiota associated with abnormal humoral immune responses to commensal organisms in enthesitis-related arthritis. Arthritis Res Ther. 2014;16(6):486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Shaw KA, Bertha M, Hofmekler T, Chopra P, Vatanen T, Srivatsa A, et al. Dysbiosis, inflammation, and response to treatment: a longitudinal study of pediatric subjects with newly diagnosed inflammatory bowel disease. Genome Med. 2016;8(1):75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, et al. Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheumatol. 2016;68(11):2646–61.

    Article  CAS  PubMed  Google Scholar 

  20. Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tejesvi MV, Arvonen M, Kangas SM, Keskitalo PL, Pirttila AM, Karttunen TJ, et al. Faecal microbiome in new-onset juvenile idiopathic arthritis. Eur J Clin Microbiol Infect Dis. 2015;35(3):363–70.

    Article  CAS  PubMed  Google Scholar 

  22. Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 2011;5(1):82–91.

    Article  CAS  PubMed  Google Scholar 

  23. Murri M, Leiva I, Gomez-Zumaquero JM, Tinahones FJ, Cardona F, Soriguer F, et al. Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Med. 2013;11:46.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zhou Y, Zhi F. Lower level of Bacteroides in the gut Microbiota is associated with inflammatory bowel disease: a meta-analysis. Biomed Res Int. 2016;2016:5828959.

    PubMed  PubMed Central  Google Scholar 

  25. Durban A, Abellan JJ, Jimenez-Hernandez N, Salgado P, Ponce M, Ponce J, et al. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome. Environ Microbiol Rep. 2012;4(2):242–7.

    Article  CAS  PubMed  Google Scholar 

  26. Scher JU, Ubeda C, Artacho A, Attur M, Isaac S, Reddy SM, et al. Decreased bacterial diversity characterizes the altered gut microbiota in patients with psoriatic arthritis, resembling dysbiosis in inflammatory bowel disease. Arthritis Rheumatol. 2015;67(1):128–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Michail S, Durbin M, Turner D, Griffiths AM, Mack DR, Hyams J, et al. Alterations in the gut microbiome of children with severe ulcerative colitis. Inflamm Bowel Dis. 2012;18(10):1799–808.

    Article  PubMed  Google Scholar 

  28. Taur Y, Jenq RR, Perales MA, Littmann ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124(7):1174–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stoll ML. Gut microbes, immunity, and spondyloarthritis. Clin Immunol. 2015;159(2):134–42.

    Article  CAS  PubMed  Google Scholar 

  30. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74.

    Article  CAS  PubMed  Google Scholar 

  31. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332(6032):974–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Farkas AM, Panea C, Goto Y, Nakato G, Galan-Diez M, Narushima S, et al. Induction of Th17 cells by segmented filamentous bacteria in the murine intestine. J Immunol Methods. 2015;421:104–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hussaarts L, Yazdanbakhsh M, Guigas B. Priming dendritic cells for th2 polarization: lessons learned from helminths and implications for metabolic disorders. Front Immunol. 2014;5:499.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Summers RW, Elliott DE, Urban JF Jr, Thompson R, Weinstock JV. Trichuris suis therapy in Crohn’s disease. Gut. 2005;54(1):87–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Summers RW, Elliott DE, Urban JF Jr, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005;128(4):825–32.

    Article  PubMed  Google Scholar 

  36. Spanogiannopoulos P, Bess EN, Carmody RN, Turnbaugh PJ. The microbial pharmacists within us: a metagenomic view of xenobiotic metabolism. Nat Rev Microbiol. 2016;14(5):273–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jourova L, Anzenbacher P, Anzenbacherova E. Human gut microbiota plays a role in the metabolism of drugs. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2016;160(3):317–26.

    Article  PubMed  Google Scholar 

  38. Morgan SL, Baggott JE, Vaughn WH, Austin JS, Veitch TA, Lee JY, et al. Supplementation with folic acid during methotrexate therapy for rheumatoid arthritis. A double-blind, placebo-controlled trial. Ann Intern Med. 1994;121(11):833–41.

    Article  CAS  PubMed  Google Scholar 

  39. Graham LD, Myones BL, Rivas-Chacon RF, Pachman LM. Morbidity associated with long-term methotrexate therapy in juvenile rheumatoid arthritis. J Pediatr. 1992;120(3):468–73.

    Article  CAS  PubMed  Google Scholar 

  40. Nayak RR, O’Loughlin C, Fischbach M, Turnbaugh PJ. Methotrexate is an antibacterial drug metabolized by human gut Bacteria [abstract]. Arthritis Rheum. 2016;68(suppl 10).

    Google Scholar 

  41. Robertson LW, Chandrasekaran A, Reuning RH, Hui J, Rawal BD. Reduction of digoxin to 20R-dihydrodigoxin by cultures of Eubacterium lentum. Appl Environ Microbiol. 1986;51(6):1300–3.

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Greenblatt DJ, Smith TW, Koch-Weser J. Bioavailability of drugs: the digoxin dilemma. Clin Pharmacokinet. 1976;1(1):36–51.

    Article  CAS  PubMed  Google Scholar 

  43. Lindenbaum J, Rund DG, Butler VP Jr, Tse-Eng D, Saha JR. Inactivation of digoxin by the gut flora: reversal by antibiotic therapy. N Engl J Med. 1981;305(14):789–94.

    Article  CAS  PubMed  Google Scholar 

  44. Saha JR, Butler VP Jr, Neu HC, Lindenbaum J. Digoxin-inactivating bacteria: identification in human gut flora. Science. 1983;220(4594):325–7.

    Article  CAS  PubMed  Google Scholar 

  45. Haiser HJ, Gootenberg DB, Chatman K, Sirasani G, Balskus EP, Turnbaugh PJ. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science. 2013;341(6143):295–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sokol H. Probiotics and antibiotics in IBD. Dig Dis. 2014;32(Suppl 1):10–7.

    Article  PubMed  Google Scholar 

  47. Carter JD. Reactive arthritis: defined etiologies, emerging pathophysiology, and unresolved treatment. Infect Dis Clin N Am. 2006;20(4):827–47.

    Article  Google Scholar 

  48. Barber CE, Kim J, Inman RD, Esdaile JM, James MT. Antibiotics for treatment of reactive arthritis: a systematic review and metaanalysis. J Rheumatol. 2013;40(6):916–28.

    Article  CAS  PubMed  Google Scholar 

  49. Smieja M, MacPherson DW, Kean W, Schmuck ML, Goldsmith CH, Buchanan W, et al. Randomised, blinded, placebo controlled trial of doxycycline for chronic seronegative arthritis. Ann Rheum Dis. 2001;60(12):1088–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Derikx LA, Dieleman LA, Hoentjen F. Probiotics and prebiotics in ulcerative colitis. Best Pract Res Clin Gastroenterol. 2016;30(1):55–71.

    Article  PubMed  Google Scholar 

  51. Jenks K, Stebbings S, Burton J, Schultz M, Herbison P, Highton J. Probiotic therapy for the treatment of spondyloarthritis: a randomized controlled trial. J Rheumatol. 2010;37(10):2118–25.

    Article  PubMed  Google Scholar 

  52. Brophy S, Burrows CL, Brooks C, Gravenor MB, Siebert S, Allen SJ. Internet-based randomised controlled trials for the evaluation of complementary and alternative medicines: probiotics in spondyloarthropathy. BMC Musculoskelet Disord. 2008;9:4.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Shukla A, Gaur P, Aggarwal A. Effect of probiotics on clinical and immune parameters in enthesitis-related arthritis category of juvenile idiopathic arthritis. Clin Exp Immunol. 2016;185(3):301–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kristensen NB, Bryrup T, Allin KH, Nielsen T, Hansen TH, Pedersen O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials. Genome Med. 2016;8(1):52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Shen TC, Albenberg L, Bittinger K, Chehoud C, Chen YY, Judge CA, et al. Engineering the gut microbiota to treat hyperammonemia. J Clin Invest. 2015;125(7):2841–50.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–63.

    Article  CAS  PubMed  Google Scholar 

  58. Bindels LB, Neyrinck AM, Salazar N, Taminiau B, Druart C, Muccioli GG, et al. Non digestible oligosaccharides modulate the gut Microbiota to control the development of Leukemia and associated Cachexia in mice. PLoS One. 2015;10(6):e0131009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lukens JR, Gurung P, Vogel P, Johnson GR, Carter RA, McGoldrick DJ, et al. Dietary modulation of the microbiome affects autoinflammatory disease. Nature. 2014;516(7530):246–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. McAllan L, Skuse P, Cotter PD, O’Connor P, Cryan JF, Ross RP, et al. Protein quality and the protein to carbohydrate ratio within a high fat diet influences energy balance and the gut microbiota in C57BL/6J mice. PLoS One. 2014;9(2):e88904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Le Leu RK, Young GP, Hu Y, Winter J, Conlon MA, et al. Dig Dis Sci. 2013;58(12):3475–82.

    Article  CAS  PubMed  Google Scholar 

  62. Sprong RC, Schonewille AJ, van der Meer R. Dietary cheese whey protein protects rats against mild dextran sulfate sodium-induced colitis: role of mucin and microbiota. J Dairy Sci. 2010;93(4):1364–71.

    Article  CAS  PubMed  Google Scholar 

  63. Kakodkar S, Farooqui AJ, Mikolaitis SL, Mutlu EA. The specific carbohydrate diet for inflammatory bowel disease: a case series. J Acad Nutr Diet. 2015;115(8):1226–32.

    Article  PubMed  Google Scholar 

  64. Ruemmele FM. Role of diet in inflammatory bowel disease. Ann Nutr Metab. 2016;68(Suppl 1):33–41.

    Article  PubMed  Google Scholar 

  65. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, de Roos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Cao Y, Shen J, Ran ZH. Association between Faecalibacterium prausnitzii reduction and inflammatory bowel disease: a meta-analysis and systematic review of the literature. Gastroenterol Res Pract. 2014;2014:872725.

    PubMed  PubMed Central  Google Scholar 

  67. Brotherton CS, Taylor AG, Bourguignon C, Anderson JG. A high-fiber diet may improve bowel function and health-related quality of life in patients with Crohn disease. Gastroenterol Nurs. 2014;37(3):206–16.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Olendzki BC, Silverstein TD, Persuitte GM, Ma Y, Baldwin KR, Cave D. An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report. Nutr J. 2014;13:5.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Sigall-Boneh R, Pfeffer-Gik T, Segal I, Zangen T, Boaz M, Levine A. Partial enteral nutrition with a Crohn’s disease exclusion diet is effective for induction of remission in children and young adults with Crohn’s disease. Inflamm Bowel Dis. 2014;20(8):1353–60.

    Article  PubMed  Google Scholar 

  70. Whitten KE, Rogers P, Ooi CY, Day AS. International survey of enteral nutrition protocols used in children with Crohn’s disease. J Dig Dis. 2012;13(2):107–12.

    Article  PubMed  Google Scholar 

  71. Heuschkel RB, Menache CC, Megerian JT, Baird AE. Enteral nutrition and corticosteroids in the treatment of acute Crohn’s disease in children. J Pediatr Gastroenterol Nutr. 2000;31(1):8–15.

    Article  CAS  PubMed  Google Scholar 

  72. Berntson L, Hedlund-Treutiger I, Alving K. Anti-inflammatory effect of exclusive enteral nutrition in patients with juvenile idiopathic arthritis. Clin Exp Rheumatol. 2016;34(5):941–5.

    PubMed  Google Scholar 

  73. Berntson L, Agback P, Dicksved J. Changes in fecal microbiota and metabolomics in a child with juvenile idiopathic arthritis (JIA) responding to two treatment periods with exclusive enteral nutrition (EEN). Clin Rheumatol. 2016;35(6):1501–6.

    Article  PubMed  Google Scholar 

  74. Kang C, Zhang Y, Zhu X, Liu K, Wang X, Chen M, et al. Healthy subjects differentially respond to dietary capsaicin correlating with the specific gut enterotypes. J Clin Endocrinol Metab. 2016;101(12):4681–9. jc20162786.

    Article  CAS  PubMed  Google Scholar 

  75. Stapel SO, Asero R, Ballmer-Weber BK, Knol EF, Strobel S, Vieths S, et al. Testing for IgG4 against foods is not recommended as a diagnostic tool: EAACI task force report. Allergy. 2008;63(7):793–6.

    Article  CAS  PubMed  Google Scholar 

  76. Shivappa N, Steck SE, Hurley TG, Hussey JR, Hebert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–96.

    Article  PubMed  Google Scholar 

  77. Russell GH, Kaplan JL, Youngster I, Baril-Dore M, Schindelar L, Hohmann E, et al. Fecal transplant for recurrent Clostridium difficile infection in children with and without inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2014;58(5):588–92.

    Article  CAS  PubMed  Google Scholar 

  78. Zhang FM, Wang HG, Wang M, Cui BT, Fan ZN, Ji GZ. Fecal microbiota transplantation for severe enterocolonic fistulizing Crohn’s disease. World J Gastroenterol. 2013;19(41):7213–6.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Suskind DL, Brittnacher MJ, Wahbeh G, Shaffer ML, Hayden HS, Qin X, et al. Fecal microbial transplant effect on clinical outcomes and fecal microbiome in active Crohn’s disease. Inflamm Bowel Dis. 2015;21(3):556–63.

    Article  PubMed  Google Scholar 

  80. Suskind DL, Singh N, Nielson H, Wahbeh G. Fecal microbial transplant via nasogastric tube for active pediatric ulcerative colitis. J Pediatr Gastroenterol Nutr. 2015;60(1):27–9.

    Article  PubMed  Google Scholar 

  81. Quera R, Espinoza R, Estay C, Rivera D. Bacteremia as an adverse event of fecal microbiota transplantation in a patient with Crohn’s disease and recurrent Clostridium difficile infection. J Crohns Colitis. 2014;8(3):252–3.

    Article  PubMed  Google Scholar 

  82. Kuntz TM, Gilbert JA. Introducing the Microbiome into precision medicine. Trends Pharmacol Sci. 2016;38(1):81–91.

    Article  CAS  PubMed  Google Scholar 

  83. Guo L, McLean JS, Yang Y, Eckert R, Kaplan CW, Kyme P, et al. Precision-guided antimicrobial peptide as a targeted modulator of human microbial ecology. Proc Natl Acad Sci USA. 2015;112(24):7569–74.

    Article  CAS  Google Scholar 

  84. Kutter E, De Vos D, Gvasalia G, Alavidze Z, Gogokhia L, Kuhl S, et al. Phage therapy in clinical practice: treatment of human infections. Curr Pharm Biotechnol. 2010;11(1):69–86.

    Article  CAS  PubMed  Google Scholar 

  85. Stoll ML, Kumar R, Lefkowitz EJ, Cron RQ, Morrow CD, Barnes S. Fecal metabolomics in pediatric spondyloarthritis implicate decreased metabolic diversity and altered tryptophan metabolism as pathogenic factors. Genes Immun. 2016;17(7):400–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Matthew L. Stoll .

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

Stoll, M.L. (2018). The Promise of Personalized Medicine. In: Ragab, G., Atkinson, T., Stoll, M. (eds) The Microbiome in Rheumatic Diseases and Infection. Springer, Cham. https://doi.org/10.1007/978-3-319-79026-8_35

Download citation

  • DOI: https://doi.org/10.1007/978-3-319-79026-8_35

  • Published:

  • Publisher Name: Springer, Cham

  • Print ISBN: 978-3-319-79025-1

  • Online ISBN: 978-3-319-79026-8

  • eBook Packages: MedicineMedicine (R0)

Publish with us

Policies and ethics