Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases

Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases

Gastroenterology 2019;-:1–16 Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Disease...

3MB Sizes 0 Downloads 27 Views

Gastroenterology 2019;-:1–16

Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases Q4

Louis J. Cohen,1 Judy H. Cho,1,2,3 Dirk Gevers,4 and Hiutung Chu5,6 1

Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; 3The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; 4Janssen Human Microbiome Institute, Janssen Research & Development, Cambridge, Massachusetts; 5Department of Pathology, University of California–San Diego, La Jolla, California; and 6Chiba University and University of California–San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, California 2

The intestinal microbiota is a dynamic community of bacteria, fungi, and viruses that mediates mucosal homeostasis and physiology. Imbalances in the microbiome and aberrant immune responses to gut bacteria can disrupt homeostasis and are associated with inflammatory bowel diseases (IBDs) in humans and colitis in mice. We review genetic variants associated with IBD and their effects on the intestinal microbiome, the immune response, and disease pathogenesis. The intestinal microbiome, which includes microbial antigens, adjuvants, and metabolic products, affects the development and function of the intestinal mucosa and inflammatory responses in the gut. Therefore, strategies to manipulate the microbiome might be used in treatment of IBD. We review microbe-based therapies for IBD and the potential to engineer patients’ intestinal microbiota. We discuss how studies of patients with IBD and mouse models have advanced our understanding of the interactions between genetic factors and the gut microbiome and challenges to the development of microbe-based therapies for IBD. Keywords: Microbiome; Inflammatory Bowel Disease; Host Genetics; Mucosal Immunity; Preclinical Therapies.

I

nflammatory bowel diseases (IBDs) include Crohn disease (CD) and ulcerative colitis (UC), chronic diseases that develop through complex interactions among genetic, immune, environmental, and microbial factors.1–3 Dysregulation of any components of this network can result in intestinal inflammation and IBD. Genetic studies have identified regulators of this network that are altered in patients with IBD—many of these control the immune response to microbes.4,5 Variants associated with risk for IBD have been identified in NOD2, ATG16L1, CARD9, and CLEC7A.6 Variants in genes that control immune detection of and response to microbes can perturb intestinal homeostasis and promote intestinal inflammation. It is important to distinguish factors that mediate the immune response to pathogens from factors that control the overall microbial ecology, which also can be affected by environmental factors (diet, medications, and geography).7 However, as we study the mechanisms by which genetic variants associated with IBD affect responses to microbes, we might learn more

about environmental factors that also do so and identify new targets for diagnosis and treatment of IBD. Studies from model systems have indicated that the gut microbiome can be modified to increase or decrease the severity of intestinal inflammation. The gut microbiome can be altered by introduction of microbes or their effectors, such as lipids, small molecules, proteins, or sugars. During the past decade, interest in microbe-based therapies has increased due to the number and perceived safety of these therapies and the potential to correct one of the causes of a disease, rather than the symptoms. Increased interest in these therapies is due in part to insights from studies of antibiotics, probiotics, and more recently fecal microbial transplantation (FMT) for IBD and other disorders.8 Strategies to correct the microbiome or its functions in patients with UC or CD have produced inconsistent results, although antibiotics were found to be effective in patients with pouchitis, with an excellent safety profile.9–11 FMT was found to alleviate symptoms in some patients with UC, although outcomes varied. Further studies are required to optimize selection of donors, determine the ability of the donated microbiota to engraft, and determine whether FMT might be better as an induction or maintenance therapy. Studies also are needed to determine how variants in genes whose products function in microbe-sensing pathways (such as NOD2) would affect microbial therapies. We review interactions between IBD-associated gene variants and the microbiome and strategies to therapeutically target specific microbiome functions.9,12 The growth of microbebased therapies presents new challenges to drug development and regulatory approval.

Abbreviations used in this paper: AHR, aryl hydrocarbon receptor; AIEC, adherent invasive Escherichia coli; CARD9, caspase recruitment domain containing protein 9; CD, Crohn disease; FDA, Food and Drug Administration; FMT, fecal microbial transplantation; GPCR, G-protein–coupled receptor; HLA, human leukocyte antigen; IBD, inflammatory bowel disease; Ig, immunoglobulin; IL, interleukin; IND, investigational new drug; LBP, live bio-therapeutic product; LPS, lipopolysaccharide; MHC, major histocompatibility complex; PSA, polysaccharide A; SCFA, short-chain fatty acid; Th, T-helper; Treg, T-regulatory; UC, ulcerative colitis. © 2019 by the AGA Institute 0016-5085/$36.00 https://doi.org/10.1053/j.gastro.2019.03.017

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120

REVIEWS AND PERSPECTIVES

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

2

REVIEWS AND PERSPECTIVES

121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180

Cohen et al

Gastroenterology Vol.

Genes That Regulate the Microbiome Genes encode many proteins that microbes are exposed to, the availability of nutrients, and the level of the immune response to microbes.13–16 Genome-wide association studies of patients with IBD have identified variants in genes that affect the intestinal response to microbes (Figure 1).6,17–25

Variants in NOD2 Variants in NOD2 were the first to be associated with risk of CD. NOD2 encodes an intracellular pattern recognition receptor, which interacts with peptidoglycan motifs of bacteria.26–28 NOD2 helps control pathogenic bacteria through hematopoietic and non-hematopoietic cells. It was initially believed that individuals with some variants of NOD2 could not efficiently clear bacterial pathogens, leading to IBD pathology.29 This hypothesis is supported by the association in patient cohorts between gastrointestinal pathogens and IBD onset.30 However, it became apparent that NOD2 also mediates the immune response to nonpathogenic commensal microbes. Patients with variants in NOD2 have microbiomes that are distinct from individuals without these variants, characterized by increased abundance of Escherichia species and decreased Faecalibacterium species, although this pattern also can be independent of NOD2 variants.15,31–33 Nod2/ mice have intestinal dysbiosis, which increase their susceptibility to colitis, compared with wild-type mice.32,34–39 Researchers have identified commensal bacteria that are pathogenic in Nod2/ mice (pathobionts), such as Bacteroides vulgatus. Mucosal barrier defects observed in Nod2/ mice were linked to B vulgatus, including abnormalities in goblet cells, expression of inflammatory genes, and increased numbers of intraepithelial lymphocytes that express interferon g.37 Depletion of B vulgatus reversed the mucosal barrier defects in Nod2/ mice, so targeted removal of organisms that exacerbate NOD2 signaling defects might restore intestinal barrier functions in patients with IBD.

Variants in ATG16L1 and Autophagy Several variants associated with risk of CD are in genes that regulate the autophagy pathway (such as ATG16L1, IGRM, and LRRK2). Autophagy has many functions, but one of its effects is to mediate lysosomal degradation and clearance of intracellular bacteria.18,19,40 Several studies have demonstrated that NOD2 interacts with ATG16L1 and that expression of CD-associated variants disrupts the association between these proteins, impairing bacterial clearance and antigen presentation.41,42 The variant encoding the T300A substitution in ATG16L1 increases susceptibility of the gene product to caspase-3 cleavage and decreases its function.43 Similar to Nod2/ mice, mice hypomorphic for ATG16L1 have microbiota-dependent susceptibility to induction of colitis and defects in toll-like receptor signaling and production of antimicrobial peptides by Paneth cells. These abnormalities in toll-like receptor signaling and Paneth cell function also have been observed in patients with CD who are homozygous for the T300A

-,

No.

-

substitution in ATG16L1.44,45 In mice, disruption of the ATG16L1 gene affects CD4þ T cells, decreasing numbers of intestinal Foxp3þ T-regulatory (Treg) cells and T-helper (Th) type 2 cell-mediated responses. These impaired T-cell functions contribute to disruption of the mucosal barrier through loss of tolerance to intestinal antigens and increased production of immunoglobulin (Ig) G and IgA against commensal microbiota.46 Although many individuals carry IBD-associated variants in NOD2 and genes that regulate autophagy, only a small proportion develop IBD. Therefore, additional environmental factors and alterations to interactions between the intestinal epithelia and microbiota are likely to be required for development of IBD. Studies of mice and cells with deletion of ATG16L1 or NOD2 have found these proteins to mediate the effects of therapeutic microbes by blocking immunomodulatory signals. For example, the common human commensal Bacteroides fragilis decreases colitis in mice by converting CD4þ T cells into Foxp3þ Treg cells that produce interleukin (IL) 10.47,48 This effect of B fragilis is lost when dendritic cells are defective in NOD2 or ATG16L1 signaling.49 Human immune cells that express ATG16L1 T300A do not induce Treg cell development at exposure to B fragilis.

Variants in CLEC7A and CARD9 Bacteria are the most well-defined microbes in the intestinal microbiota, but fungal communities also are altered in microbiomes of patients with IBD. This should not be surprising considering the associations between IBD and an aberrant immune response to fungal antigens, based on detection of antibodies to Saccharomyces cerevisiae.50 Similar to the intestinal bacterial communities in patients with IBD, the diversity of fungal microbiota is decreased and certain atypical phyla dominate, such Ascomycota and Basidiomycota.51 Fungal members of the gut microbiota interact with pattern recognition receptors such as CLEC7A (also called DECTIN1), a glycoprotein expressed by cells in the innate immune system that recognizes b-1,3–linked and b-1,6–linked glucans from fungi. A single-nucleotide polymorphism in CLEC7A has been associated with IBD. Mice lacking DECTIN1 have increased susceptibility to colitis and an altered fungal community.52 Interestingly, some variants in CLEC7A have been associated with medically refractory UC; no other IBD-associated variants have been associated with response to therapy.52 DECTIN1 signals through the adaptor protein caspase recruitment domain containing protein 9 (CARD9).23 Card9/ mice also have an altered fungal community structure with increased susceptibility to colitis induced by dextran sodium sulfate.53 The fungal dysbiosis that results from loss of CARD9 in mice is associated with loss of Th17 cells, consistent with the importance of these cells to controlling fungal infections. Patients with a homozygous mutation in CARD9 (rs10781499) have severe mucocutaneous candiadiasis.53–56 DECTIN1 signaling through CARD9 might alter the immune response through changes in pathways regulated by nuclear factor kB, c-Jun N-terminal kinase, and mitogen-activated protein kinase.54

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240

Genetics and Microbe-Based Therapies for IBD

3

print & web 4C=FPO

241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300

2019

Q1

301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360

REVIEWS AND PERSPECTIVES

-

Figure 1. Genetic factors that affect the intestinal microbiome. Variants of genes that affect risk for IBD have been associated with alterations in the composition of the microbiome. Mutations in NOD2, ATG16L1, and LRRK2 decrease secretion of antimicrobial peptide by Paneth cells. Variants in CLEC7A and CARD9 have been associated with decreased abundance of Lactobacillus species, possibly owing to altered activities of dendritic cells and macrophages. Variants in NOD2 are associated with increased abundance of Escherichia species and Bacteroides vulgatus and decreases in Faecalibacterium species. Impaired ATG16L1 signaling has been associated with increased production of IgG and IgA against commensal microbiota, resulting in a loss of tolerance to intestinal microbes. Polymorphisms in MHC class II or HLA genes affect production of IgA in response to microbes. Defects in mucus production alter the intestinal microbiome and increase susceptibility to colitis. Gene names in red have variants associated with CD and UC; gene names in orange have variants associated with only CD; and gene names in purple have variants associated with only UC. AMP, antimicrobial peptide; IFN, interferon; TNF, tumor necrosis factor.

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

4

REVIEWS AND PERSPECTIVES

361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420

Cohen et al

Gastroenterology Vol.

CARD9 also has a role in response to bacteria through its interaction with NOD2.57 A study of Card9/ mice reported alterations in fungal and bacterial communities, but colitis susceptibility was dependent only on the bacterial community. Variations in phenotypes of knockout mice show the complexities of microbiome functions; many potential confounding variables affect host microbial interactions. In Card9/ mice, bacterial tryptophan metabolites were found to account for some variations in phenotypes.58 Bacterial tryptophan metabolites signal through human aryl hydrocarbon receptors (AHRs), which are important for mucosal tolerance. Impaired microbial tryptophan metabolism in Card9/ mice was associated with colitis susceptibility. Administration of Lactobacillus strains that metabolize tryptophan into AHR ligands was sufficient to decrease colitis in Card9/ mice. Fecal samples from patients with CD or UC with IBD-associated polymorphisms in CARD9 lacked AHR ligands.

Human Leukocyte Antigens Human leukocyte antigens (HLAs) are encoded by genes in the major histocompatibility complex (MHC), among the most polymorphic in humans. Proteins encoded by the MHC locus mediate antigen presentation and coordination of the immune response. The diversity of HLAs allows the immune system to respond to different pathogens, but certain polymorphisms increase risk for inappropriate responses to self-antigens.59 Variants in MHC class II genes have been associated with UC and CD and correlate with disease location.60 The heterozygosity of variants in class II genes is lower in patients with UC.61 These variants might weaken recognition of antigens on commensal microbes.61 Studies of mice and patients with other MHC class II-associated diseases, including rheumatoid arthritis, celiac disease, and ankylosing spondylitis,62–68 showed a correlation between specific HLA alleles and distinct communities of microbes, which increase numbers of Th17 cells and intestinal permeability.69,70 Studies of mice indicated that interactions between HLA polymorphisms and the intestinal microbiota are mediated by altered production of IgA and specific bacterial species, such as Bacteroides species, which are sufficient to induce colitis.64,71 Further research is needed into HLA variants and their associated microbial communities in patients with IBD.

Mucins The intestinal mucus layer serves as another physical barrier that separates luminal microbes from the intestinal epithelium. The mucus layer is composed of the glycoproteins MUC2, MUC5AC, MUC5B, and MUC6, which are secreted by goblet cells.72 Specific microbes degrade mucin glycoproteins, so microbial community structure corresponds with changes in mucin glycosylation. The diverse array of mucin glycans creates a specific niche for specific intestinal bacteria that have evolved to bind these glycoproteins and use them as a carbon source. Therefore, a disruption in the mucus barrier would change bacterial ecology and deplete an important intestinal barrier function.

-,

No.

-

In intestines of mice with colitis caused by administration of dextran sodium sulfate or disruption of the Il10 gene and in patients with UC, the mucus layer is thinner and highly penetrable to organisms that do not typically inhabit this niche.73,74 Patients with UC have aberrant expression of MUC5AC, MUC6, and MUC2.75–78 Mice with deletion of MUC2 have increased susceptibility to colitis.79–81 Although variants in mucin genes have not been associated with IBD, their altered expression patterns in intestinal tissues from patients with IBD indicate that their activity is important for maintenance of the microbiome and prevention of inflammation.

Other Genetic Variants Genome-wide association studies have identified more than 200 loci with significant associations with IBD, but only a minority of these loci can be mapped to the 1 or 2 most likely alleles.82 These loci are generally enriched for proteinaltering variants and proteins in cytokine pathways. For example, the IL23 signaling pathway includes IL23R, IL12B, TYK2, and JAK2. IL23 promotes development of Th17 cells, CD4þ T cells, Tc17 CD8þ T cells, and innate lymphoid cells type 3. Agents that block IL12 and IL23 signaling have been approved by the Food and Drug Administration (FDA) for treatment of CD. However, cytokines produced by Th17 cells, including IL17 and IL22, protect against IBD.83,84 In mice, IL17 prevents expansion of segmented filamentous bacteria, which promote colitis, through IL17 receptor– mediated epithelial cell signaling or by increasing neutrophil recruitment.85,86 Some anti-inflammatory effects of IL22 are mediated by increased control of intestinal pathobionts and/or increased production of mucus.87,88 Although many treatments for IBD aim to control cytokine production, dietary and bacterial metabolites can be ligands for G-protein–coupled receptors (GPCRs), which often activate anti-inflammatory signaling pathways.89,90 Variants in GPCR genes have been associated with IBD. The GPCR GPR35 is a receptor for the tryptophan intermediate kynurenic acid, which has been associated to IBD and primary sclerosing cholangitis.82,89,91

Interactions Between Genetic Variants and the Intestinal Microbiome Altered interactions between the intestinal epithelium and the microbiota are an important step in IBD pathogenesis.92 These defective interactions might be corrected with microbes or microbial products. Many variants in genes associated with IBD affect responses of immune or intestinal cells to microbes, but do not affect the overall microbial ecology. However, currently we can assess only large changes in the overall microbiome, and we might miss changes in specific microbial populations or niches. We need to better understand interactions between genetic alterations and changes in specific populations of microbes. It also is important to conduct experiments with appropriate controls, because many factors can perturb the microbiome.93 The most powerful evidence for the mechanisms

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480

481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540

2019

Genetics and Microbe-Based Therapies for IBD

by which genetic variants alter the intestinal microbiome has come from studies of adoptive fecal transfer with littermate controls and careful analyses of knockout or knockdown mice, such as in the studies of NOD2. Studies of mice have shown that alterations of the microbiota can promote colitis. Microbes interact with cellsurface proteins, secreted metabolites, and other environmental substrates (Figure 2). However, it is not clear whether it is alterations in the microbes or their effectors that promote development of IBD. Understanding how different intestinal microbes can cause different phenotypes of IBD could lead to development of microbe-based therapies.

Germ-Free and Gnotobiotic Mice Studies of germ-free and gnotobiotic mice have increased our understanding of interactions among intestinal cells, microbes, and development of the immune system. Mice raised under germ-free conditions have alterations in gutassociated lymphoid tissue, plasma cells, T cells, responses to microbial peptides, the crypt–villus architecture, and the mucus barrier.94,95 Germ-free mice also have decreased expression of NOD2, indicating that its expression is regulated in response to microbes.34 Colonization of germ-free mice with a healthy microbiota restored intestinal homeostasis, although individual mucosal functions can be restored by organisms such as Lactobacillus plantarum or Escherichia coli Nissle 1917, which reactivates NOD2 signaling.34 Interestingly, infection with norovirus can restore most mucosal barrier abnormalities observed in germ-free mice.96 Experiments with germ-free and gnotobiotic mice established the role of the microbiota in the development of colitis independent of genetic factors. In mice with T-cell transfer induced colitis, specific microbes promote development of colitis, whereas others do not.97 Microbes can induce or lessen the severity of colitis in IL10-knockout mice or in mice given dextran sodium sulfate.98–100 Hence, individual populations of microbes can promote or prevent intestinal inflammation, depending on genotype; in IL10 knockout mice, most bacteria elicit colitis. Therefore, microbe-based therapies might be selected based on a patient’s genotype, but not be effective in the entire population of patients with IBD. However, some microbiome therapies, such as those that increase mucosal barrier function, could have the widest applicability.

Interactions Between Microbes and Intestinal Cells Intestinal microbes can alter the immune response. For example, commensal Clostridia strains promote accumulation of Foxp3þ Treg cells in the gut by inducing production of transforming growth factor b and indoleamine 2,3dioxygenase.101,102 Treg cells down-regulate inflammatory responses and mice colonized with specific Clostridia species are resistant to induction of colitis.101 Although specific Clostridia species can induce development of RORgtþ Treg cells, Clostridium ramosum also has this function.103,104 Studies of gnotobiotic mice identified specific immunemodulatory effects of individual species of commensal

5

bacteria and showed that specific types of immune cells, such as Treg cells, can be induced by a wide range of bacteria, whereas others appear to require specific microbes.105 Some microbes activate populations of immune cells that promote intestinal inflammation, such as Th17 cells.106 Segmented filamentous bacteria activate Th17 cells in intestines of mice; adherent invasive E coli (AIEC) and Bifidobacteria adolescentis induce mucosal and systemic populations of Th17 cells in the gut.107–109 Decrease of Th17 cell-inducing bacteria can lessen the severity of colitis in mice. An organism does not have to change its abundance in the population to have significant effects. Segmented filamentous bacteria, which increase development of Th17 cells, also promote T-cell–dependent production of IgA.110 Coating of microbes by IgA has been proposed as a marker of immune activation by that microbe; IgA-coated bacteria, including certain Enterobacteriaceae, induce colitis in mice and have been associated with CD-associated spondyloarthritis.108,110,111 Because IBD-promoting pathobionts might be specific to a gene or an individual, understanding IgA responses to microbes might help prevent the emergence of pathobionts or help target pathobionts in specific individuals. The immune response to a microbe is likely specific to its niche. Alcaligenes, Achromobacter, Bordetella, and Ochrobactrum species specifically colonize lymphoid tissues, where they interact with innate lymphoid cells and dendritic cells to modulate IL10 production and intestinal repair mechanisms.112 The bioactivity of a microbe might require certain environmental signals, such as dietary metabolites, which can induce production or activity of bacterial effectors or serve as metabolic substrates. Some Alistipes, Clostridium, and Bilophila species can decrease production of tumor necrosis factor by immune cells only in the presence of a certain diet.113,114 Specific Candida species can cause colitis in mice; colonization can be inhibited by Bacteroides thetaiotaomicron, which induce the production of the antimicrobial peptide CRAMP.115 Interactions among organisms might affect the efficacy of microbiota-based therapies. Helminths are not considered commensals, but were prevalent during human history and are believed to have functions that affect microbiome development.116 Certain helminths induce responses of Th2 cells and increase IL10 secretion.117–119 Although helminths probably regulate these immune responses to promote their own infection of a patient, their functions might benefit patients with IBD, which has been demonstrated in phase 2 and 3 studies. The observation that the same organism can be beneficial or detrimental, depending on the patient or model, is not unique to helminths, but applies to many microbes. Viruses could have roles in IBD pathogenesis and norovirus is sufficient to restore mucosal barrier defects in germ-free mice, independent of microbiota.96,120

Microbe Effectors Studies of model systems have identified organisms that induce specific cell responses, but these responses vary. Many complex factors mediate these interactions and we

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600

REVIEWS AND PERSPECTIVES

-

6

Gastroenterology Vol.

-,

No.

-

print & web 4C=FPO

REVIEWS AND PERSPECTIVES

601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660

Cohen et al

Figure 2. Effects of the microbiome on intestinal and immune cells. The intestinal microbiome and its products modulate immune responses through induction of dendritic cells and lymphocytes (such as Th17 cells, Treg cells [Tregs in figure]), innate lymphoid cells, and cytokine production (IL10, IL22). Intestinal bacteria also can modulate immune signaling pathways, such as expression of NOD2, and epithelial repair. Specific microbes can increase susceptibility of mice to colitis. APC, antigen presenting cell; ILC, innate lymphoid cell.

know little about the mechanisms by which bacteria alter the intestinal environment. Microbes interact with the intestinal epithelium or each other through secreted or cellsurface effectors. Identifying these effectors could help us learn more about the pathogenesis of IBD and lead to

therapeutic strategies. Small molecules produced by microbes have been tested for their therapeutic effects for decades.121 Studies are needed to identify the effectors produced by microbes that act on intestinal and immune cells.122

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720

721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780

2019

Genetics and Microbe-Based Therapies for IBD

Short-chain fatty acids (SCFAs) decrease colitis, promote Treg-cell development, and down-regulate inflammatory signaling pathways.123,124 Clostridia species produce SCFAs, which reach millimolar concentrations in the intestine and can activate GPCRs, inhibit histone deacetylases, and provide an energy source for colon epithelial cells.125–127 Polyamines such as putrescine or spermidine are virulence factors but also enhance intestinal barrier functions, including mucus secretion, T-cell differentiation, and production of IgA.128–131 Bifidobacterium animalis increases polyamine levels, which correlates with decreased secretion of tumor necrosis factor and IL6 by myeloid cells.130 Interestingly, bacteria and human cells each produce polyamines, which might mediate some of their interactions. Bacteria and human cells also each produce long-chain N-acyl signaling molecules that signal through specific GPCRs to regulate immunity, inflammation, and metabolism.90,132 Structural similarity between human and bacterial signaling molecules is likely to be common, because bacteria also can synthesize the neurotransmitter g-aminobutyric acid and certain Bacteroides species metabolize tryptophan to tryptamine, a precursor to serotonin.133–135 Bacterial tryptophan metabolites are ligands for AHR, but the metabolism of tryptophan and other aromatic amino acids in Clostridia species has been linked to intestinal barrier functions through the production of indoleproprionic acid.136 Bacterial metabolites of bile acids, such as the generation of taurine, might regulate inflammasome functions and increase microbial diversity.137 Bacteria interact through cell-surface effectors, including through secretion of outer membrane vesicles.138 Zwitterionic polysaccharide A (PSA), on the surface of B fragilis, regulates activity of Foxp3þ Treg cells in the gut.47 Administration of purified PSA, or B fragilis, is sufficient to activate intestinal Treg cells and decrease colitis in mice.48,49,139 Lipopolysaccharide (LPS), probably the most well-studied bacterial cell-surface molecule, has countless variations in structure. Specific types of LPS, such as penta-acylated LPS produced by certain Bacteroides species, can inhibit immune responses, in contrast to hexa-acylated LPS from E coli, which stimulates the immune response.140 Interestingly, in a mouse model of diabetes, this LPS from E coli decreased autoimmunity and development of diabetes. Therefore, bacteria can have different effects in different model systems, so it is important to understand all the effects of a microbe before it is included in a therapeutic strategy. Sphingolipids isolated from B fragilis are similar to the human molecules and can regulate natural killer T cells.141

Challenges to Microbiome-Based Therapeutics Microbe-based therapeutic strategies can aim to alter the overall microbiome or its environment, introduce therapeutic microbes, or alter production of microbe effectors. Early studies focused on application of therapeutic microbes, despite the challenges of developing a drug that includes living organisms. Small-molecule development begins with basic research and discovery of bioactive

7

molecules, followed by preclinical studies (formulation, toxicity, and pharmacokinetic analyses), followed by trials of safety and efficacy in patients. Development of microbebased therapies has changed concepts of drug mechanisms, formulation, and monitoring, requiring new approaches for development and regulation (Figure 3). In 2012 the Center for Biologics Evaluation and Research issued guidelines to assist in therapeutic development of live organisms, which they classified as live bio-therapeutic products (LBPs). LBPs are defined as biologic products that contain live organisms, such as bacteria, and that might be used in the prevention, treatment, or cure of human diseases but are not vaccines. Development of prebiotics and bacterial effectors is likely to follow regular drug development pathways, but LBPs are the most pursued of the microbe-based therapies (Figure 3).

FDA Regulation In 2012 the FDA published regulations for development of LBPs, which were updated in 2016.142 It is important to distinguish LBPs from probiotics, which are organisms that have obtained the generally recognized as safe label and fall outside of this regulation. Trials of LBPs for treatment of diseases require an investigational new drug (IND) application. An IND application can be waived for an LBP that is available in conventional foods or dietary supplements, in consultation with the FDA. FMTs were first performed without an IND because feces were considered widely available, but in 2012, regulation by the FDA changed—now an IND is required for studies of FMT in patients with IBD. However, use of FMT for recurrent Clostridium difficile infection has discretionary regulation. The FDA does not require toxicity studies for trials of FMT, but it does require adequate characterization of microbe strains to be tested. Chemistry, manufacturing, and control data must include the historical context of the organism(s), the purity, and details about the presence of virulence factors, toxins, or antibiotic resistance genes and the potential to spread these genes. Whole-genome sequencing is performed for many LBPs to address these safety concerns. Genetically modified LBP require additional tests to ensure the stability of genetic modifications. Antibiotic resistance genes can be present but require justification and are not acceptable for LBPs that could cause opportunistic infections. Phenotype-based antibiotic resistance testing for each LBP is required in addition to traditional toxicologic profiling. Product release testing (the identity, viability, potency and purity of each LBP) is perhaps a more important step, relative to smallmolecule therapies, because LBPs are a challenge to quantify and can change during production. Potency assays (such as colony-forming units) should be used to calculate the dose or release of a predefined product when mechanisms are well defined. Later stages might require an assay that tests the agent’s mechanism, which might not be straightforward for a complex LBP. In addition, it is still not clear what level of evidence is needed to justify a specific selection of strains for an LBP. Regulations are likely to include standardization of these definitions among countries,

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840

REVIEWS AND PERSPECTIVES

-

8

Gastroenterology Vol.

-,

No.

-

print & web 4C=FPO

REVIEWS AND PERSPECTIVES

841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900

Cohen et al

Figure 3. Challenges to development of therapeutic LBPs. Compared with small molecules, LBPs have different challenges at each stage of drug development. Challenges are presented to drug discovery, preclinical studies, and clinical studies. (A) In contrast to small molecules, which usually target a specific protein or class of feature of proteins, LBPs are identified based on their association with a disease phenotype in humans or mouse models. (B) Small molecules require extensive toxicity studies, whereas LBPs are believed to be nontoxic but require assessments for virulence or antibiotic resistance. Preclinical studies of LBPs are not informative for bioavailability but focus on viability or bioactivity, which can require specific encapsulation methods, adjuvants, or genetic modifications. (C) Trials of LBPs require specific attention to adverse events related to transmission of the microbe, loss of its bioactivity, or off-target effects. Small molecules also can have off-target effects, but these might be easier to predict based on findings from preclinical studies. Early-phase studies of LPBs might be important for final formulation, because bioavailability and potential mechanisms can be assessed based on findings from small groups of patients.

because there is no equivalent definition of an LBP in the European Union, which has categorizations for therapeutic organisms not present in the United States.

Microbiota-Based Therapeutics in Preclinical and Clinical Development Because the guiding framework for LBP has been clarified during the past 6 years, there has been an increase in

companies developing discovery platforms and/or introducing candidate therapies into preclinical and clinical studies for the treatment of IBD (Table 1).

Modulators of the Microbiome The microbiome can be modified by improving mucosal barrier functions or depletion or enrichment of organisms linked with diseases, an increased or decreased immune response, or other outcomes. Zoenasa (Altheus Therapeutics,

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960

961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 -

Table 1.Microbiota-Based Therapies in Development for IBD

Ecology modulator

Microbial effectors

Company

Producta

Location

Deliverya

Development stage Phase 2 study of patients with CD Preclinical

Mechanism Prevents pathobiont invasion by blocking fimH

Enterome

USA

EB-8018: small molecule

Oral

Immuron

Australia

N/A

Oral

BiomX Eligo Intralytix

Israel France USA

BX002 N/A EcoActive

N/A N/A Oral

Artizan Biosciences Seres Therapeutics

USA USA

N/A SER-287: naturally derived community

N/A Oral

Seres Therapeutics Janssen (licensed to Vendanta Biosciences) Rebiotix (acquired by Ferring) 4D Pharma

USA

SER-301: defined community VE202: defined community

Oral Oral

USA

RBX2660

Enema

UK

Thetanix: Bacteroides thetaiotaomicron

Oral

4D Pharma Osel ImmuneBiotech ActBiotics

UK USA Sweden USA

Rosburix: Roseburia hominis CBM588: Clostridium butyricum IB002: lactobacilli AG-014: GMO

Oral Oral N/A Oral

Preclinical Preclinical Phase 1 and 2a studies of patients with CD Preclinical Phase 2 study of patients with UC Preclinical Phase 1 study of patients with UC Phase 1 in pediatric patients with UC Phase 1 study of patients with CD Preclinical Phase 1 Preclinical Phase 1

Rise Therapeutics

USA

R-3750: GMO

N/A

Preclinical

Nordisk Rebalance

Denmark

Profermin: Lactobacillus plantarum, oats, phosphatidylcholine

Oral

Finch Therapeutics

USA

FIN524, defined community

Oral

Allergan

Ireland

ABI-M201,301

Oral

Marketed as food for special medical purpose Preclinical studies of colitis Preclinical

NextBiotix/Exeliom Biosciences ViThera Chain Biotech

France

NBX-1650

N/A

Preclinical

USA USA

VT301 CHN-1, CHN-2

Oral Oral

Preclinical Preclinical

PanTheryx Host Therabiomics

USA UK/USA

PTX-400 L1173

Oral N/A

Preclinical Preclinical

Licensed from Assembly Biosciences and uses Gemicel coating Faecalibacterium prausnitzii to treat inflammation Modified lactobacilli strains Antimicrobial peptide with Clostridium Assisted Drug Delivery Medical food, prebiotic Platform to identify effectors

Second Genome Symbiotix Biotherapies

USA USA

SG-2-0776: protein SYMB-104: PSA

oral N/A

Preclinical Preclinical

Intestinal healing Bacteroides fragilis–derived immune modulator

Antibodies and adjuvant to boost immune response to pathobionts Bacteriophage to deplete pathobionts CRISPR-CAS to deplete pathobionts Bacteriophage to target AIEC

Subtractive therapy Not available Not available Induce Treg cells Restore microbiome composition Not available Not available Increase SCFAs Not available Heterologous expression of anti-TNF nano-body by Lactobacillus species Lactobacillus species for heterologous expression Phase 2 of patients with UC, a prebiotic that increases SCFAs Not available

Genetics and Microbe-Based Therapies for IBD

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

LBP

2019

Therapy type

9

1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080

REVIEWS AND PERSPECTIVES

Host target

N/A, not applicable because agent is in preclinical stage of development; GMO, genetically modified organism; TNF, tumor necrosis factor. a Companies were identified by the Janssen Human Microbiome Institute from public resources.

France USA USA UK Belgium USA USA USA

N/A plasmid EB110/EB220 Aquashell Phloral, Duocoat In vitro microbiome model GMO Gemicel capsule SGM-1019: Small molecule

Injection Oral Oral Oral N/A N/A Oral Oral

Preclinical Preclinical N/A N/A N/A N/A N/A Phase 1

Plasmids that produce heat shock proteins Microbial metabolite associated with CD pH release: polysaccharide pH release Predict in vivo conditions Not available pH release Modulates inflammasome

Gastroenterology Vol.

Formulation

Therapy type

Table 1. Continued

Company

Location

Producta

Deliverya

Development stage

Mechanism

REVIEWS AND PERSPECTIVES

1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140

Cohen et al

Alma Bio Therapeutics Enterome Finch Therapeutics Intract Pharma Prodigest Synlogic Therapeutics Assembly Biosciences Second Genome

10

-,

No.

-

Oklahoma City, OK) is a formulation of N-acetyl cysteine, phosphatidylcholine, and mesalamine that is believed to strengthen the mucosal barrier. Zoenasa has been formulated as a rectal gel or oral tablet and tested in a phase 2 study of patients with distal UC. Specific microbes or communities of microbes could be pathogens (pathobionts), and depletion of these microbes might be beneficial. A challenge to modulating the microbiome by depleting pathobionts is that these microbes might be specific to an individual based on that individual’s pathophysiology and/or genetics. Production of IgA in response to bacteria might be measured to identify specific pathobionts in individual patients. Immuron (Blackburn North, VIC, Australia) developed an oral immunotherapy (antigens, adjuvants, and antibodies) designed to decrease or increase specific microbiota. IMM124E (Immuron) is an oral formulation of antibody against LPS and glycosphingolipid adjuvants that is in preclinical studies but might be used to treat UC. BiomX (Ness Ziona, Israel) developed a therapy that directly targets pathobionts using bacteriophages and Eligo Biosciences (Paris, France) developed a therapy to deplete bacteria using CRISPR.143 The companies that are developing these agents have not stated their specific target species, but Eligo has a platform to allow for an individualized assessment of potential pathobionts. EcoActive (Intralytix, Inc, Baltimore, MD) has just entered phase 1 and 2 trials of patients with CD—it is an oral bacteriophage cocktail that depletes AIEC. AIEC is enriched by defects in NOD2 signaling in patients with CD.144 EB-8018 was designed to decrease AIEC by blocking fimH. EB-8018 (Enterome SA, Paris, France) is entering phase 2 studies of patients with CD and was found to be safe in a phase 1 study. A diagnostic assay (IBD-210) has been designed to measure fimH in fecal samples to identify patients likely to respond to EB-8018.

Live Bio-Therapeutic Products Studies of patients and animal models have led to the discovery of many LBPs. LBP are being developed using different formulations, including naturally derived communities, defined communities, individual organisms, and genetically modified organisms. SER-287 (Seres Therapeutics, Cambridge, MA) is a naturally derived community and SER-301 (Seres Therapeutics) is a defined community based on human cohort studies, whereas VE202 (Janssen, Beerse, Belgium; licensed to Vedanta Biosciences, Cambridge, MA) was developed as a synthetic community in mice, based on a targeted increases in Treg cells.101 In contrast to community LBP, Thetanix (4D Pharma, Leeds, UK) is a single strain of B thetaiotaomicron that is in a phase 1 trial of children with CD. Bacteroides thetaiotaomicron might have multiple mechanisms, including modulation of fungi, although there also is a negative association between B thetaiotaomicron and infection with pathogenic strains of E coli. The pleiotropic effects of LBP will warrant specific safety attention. A strain of Clostridium butyricum in preclinical studies for IBD was found to be safe for treatment of C difficile infection. Clostridium butyricum is believed to act specifically by increasing SCFAs, although other LBPs are

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200

1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260

2019

Genetics and Microbe-Based Therapies for IBD

11

believed to increase SCFA.145 ImmuneBiotech (Lund, Sweden) has a narrow focus on a proprietary panel of lactobacilli, to which they assign immunomodulatory functions. Many lactobacilli carry generally recognized as safe designations, which will facilitate their approval process and are easy to manipulate in the laboratory for the development of genetically modified organisms. AG-014 (ActBiotics, Venice, CA) is a lactobacillus engineered to produce a nano-body against tumor necrosis factor that is in phase 1 studies of patients with IBD. Countries outside the United States have an additional regulatory category, independent of dietary supplement and LBP, which is a food for special medical purposes. A food for special medical purposes is naturally found in the diet but can be marketed for the treatment of a disease. In Denmark, Profermin (Nordisk Rebalance, Hillerød, Denmark) is a food for special medical purposes—it is a combination of Lactobacillus plantarum, oats, and phosphatidylcholine. Each component of this pill has a separate effect as a prebiotic (oats), barrier modulator (phosphatidylcholine), and LBP (L plantarum), although it is unclear if there is interaction among components. Profermin is on the market and has been studied in small trials of patients with UC, in which it had moderate efficacy but without endoscopic end points.146,147 Helminths, specifically Trichuris suis, showed efficacy in phase 2 studies of patients with CD but had limited efficacy in phase 3 studies.148–151

Companies have focused on formulations for better delivery an LBP to a niche. The Gemicel capsule (Assembly Biosciences, Carmel, IN) was developed for colonic release of LBP, incorporating 2 separate pH-dependent mechanisms. Aquashell (Finch Therapeutics, Somerville, MA) is a pH-sensitive formulation for colonic delivery that incorporates a separate polysaccharide coating that is digested by colon microbes. Both formulation strategies have been used to encapsulate LBPs that are in trials of patients with IBD. Duocoat (Intract Pharma, London, UK) is optimized for duodenal release, using a pH-sensitive coat, and Phloral (Intract Pharma) is optimized for colon release. Each of these encapsulation strategies necessitates specific attention to a strict anaerobic process that is unique for each LBP, because previous dietary probiotics (such as Lactobacillus or Bifidobacterium species) were microaerophilic.153 Genetically modified organisms for treatment of IBD might be viewed as a type of formulation strategy to increase therapeutic effect and mitigate variation. Genetic tools to manipulate human microbes are required. Synlogic Therapeutics (Cambridge, MA) programs internal circuits in E coli Nissle strains to induce expression of effector genes in response to specific environmental signals. This technology is used to produce microbial effectors only in the correct environment. Synlogic Therapeutics also has developed chromosome markers for in vivo monitoring of LBPs, which will facilitate bioavailability studies.

Microbe Effectors

Targeting Interactions Between Intestinal Cells and Microbes

Microbe effectors have specific effects on cells and follow a traditional drug development strategy. Approval of microbial effectors by regulatory agencies might be straightforward, but there are few products in early stages of development, because we understand so little about them. EB110 (Enterome, Paris, France) is a microbe-derived metabolite identified in humans that has been associated with development of CD through unknown mechanisms. SG2-0776 (Second Genome, South San Francisco, CA) is a microbe effector (protein) that promotes intestinal healing and is in preclinical studies for treatment of IBD. PSA has been one of the most extensively studied bacterial effectors and has a number of immune-regulatory properties.47,152 However, IBD-associated variants in NOD2 and ATG16L1 could mitigate the effect of PSA and be used to identify patients not likely to respond.49 PSA is in preclinical development and it is unclear whether clinical trials will compare effects in patients with different genotypes.

Formulation Strategies Formulation is an important challenge for microbiomebased therapies. Formulation aims for reproducible effects among individuals and delivery of viable organisms to their niche. Many LBPs in early-stage trials are administered daily, because it is likely that LBPs do not incorporate into the microbiome and expand. However, they might be given in intermittent or even single doses, if our understanding of microbiome homeostasis improves, and we can more carefully select LBPs or use of adjuvants.

We have begun to identify cell signaling pathways that are regulated by bacteria and might be therapeutically manipulated. SGM-1019 (Second Genome) is a small molecule that affects the inflammasome, identified using a discovery platform. SGM-1019 has progressed through phase 1 studies and is being developed for treatment of IBD and nonalcoholic steatohepatitis, for which it is entering phase 2 studies.

Future Directions Microbe-based therapies are becoming more diverse and effective as our understanding of the interactions between the microbiome and human cells increases. We have begun to better understand the effects of genetic and environmental factors on the microbiome and its products or effectors. Formulation strategies can be refined to address the primary challenge of diversity in the microbiomes among individuals and for treatment of specific diseases. Additional challenges to development and use of microbe-based therapies involve issues regarding intellectual property. A full discussion of this topic is beyond the scope of this review, but one of the major problems is how to enforce patent laws for the composition of a natural product. LBPs that can be defined as natural products include isolated microbes or their effectors (such as metabolites or proteins). Natural products can be protected by method patents, which state their use for treatment of IBD, but these patents are not as easily enforced, which

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320

REVIEWS AND PERSPECTIVES

-

12

REVIEWS AND PERSPECTIVES

1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380

Cohen et al

Gastroenterology Vol.

dissuades companies from developing these types of products. One strategy has been to genetically modify microbes or use them in combination with other microbes, as a genetically modified organism, or as a defined community that is not found in nature (not a natural product). Companies have obtained composition of matter patents for LBPs that are natural communities, so it will be important to see how these patents are enforced if these products come to market. Patent law protection is critical for development of microbe-based therapies; it is likely that the legal framework will change as it has in the past.

References 1. Abraham C, Medzhitov R. Interactions between the host innate immune system and microbes in inflammatory bowel disease. Gastroenterology 2011;140:1729–1737. 2. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121–141. 3. Sun M, He C, Cong Y, et al. Regulatory immune cells in regulation of intestinal inflammatory response to microbiota. Mucosal Immunol 2015;8:969–978. 4. Hall AB, Tolonen AC, Xavier RJ. Human genetic variation and the gut microbiome in disease. Nat Rev Genet 2017; 18:690–699. 5. Luca F, Kupfer SS, Knights D, et al. Functional genomics of host–microbiome interactions in humans. Trends Genet 2018;34:30–40. 6. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature 2011;474:307–317. 7. Rothschild D, Weissbrod O, Barkan E, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018;555:210–215. 8. Hoffmann DE, Palumbo FB, Ravel J, et al. A proposed definition of microbiota transplantation for regulatory purposes. Gut Microbes 2017;8:208–213. 9. Lopez J, Grinspan A. Fecal microbiota transplantation for inflammatory bowel disease. Gastroenterol Hepatol (N Y) 2016;12:374–379. 10. Khan KJ, Ullman TA, Ford AC, et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011;106:661–673. 11. Singh S, Stroud AM, Holubar SD, et al. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev 2015;11:CD001176. 12. Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology 2017;152:327–339 e4. 13. Benson AK, Kelly SA, Legge R, et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci U S A 2010;107:18933–18938. 14. Goodrich JK, Waters JL, Poole AC, et al. Human genetics shape the gut microbiome. Cell 2014;159:789–799. 15. Knights D, Silverberg MS, Weersma RK, et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med 2014;6:107.

-,

No.

-

16. Blekhman R, Goodrich JK, Huang K, et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol 2015;16:191. 17. Parkes M, Barrett JC, Prescott NJ, et al. Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn’s disease susceptibility. Nat Genet 2007;39:830–832. 18. Hampe J, Franke A, Rosenstiel P, et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 2007;39:207–211. 19. Consortium WTCC. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007;447:661–678. 20. Franke A, McGovern DP, Barrett JC, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet 2010; 42:1118–1125. 21. Rivas MA, Beaudoin M, Gardet A, et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat Genet 2011;43:1066–1073. 22. Anderson CA, Boucher G, Lees CW, et al. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet 2011;43:246–252. 23. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491:119–124. 24. Liu JZ, van Sommeren S, Huang H, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015;47:979–986. 25. Ellinghaus D, Jostins L, Spain SL, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet 2016;48:510–518. 26. Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 2001;411:599–603. 27. Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 2001;411:603–606. 28. Philpott DJ, Sorbara MT, Robertson SJ, et al. NOD proteins: regulators of inflammation in health and disease. Nat Rev Immunol 2014;14:9–23. 29. Kobayashi KS, Chamaillard M, Ogura Y, et al. Nod2dependent regulation of innate and adaptive immunity in the intestinal tract. Science 2005;307:731–734. 30. Molodecky NA, Kaplan GG. Environmental risk factors for inflammatory bowel disease. Gastroenterol Hepatol (N Y) 2010;6:339–346. 31. Frank DN, Robertson CE, Hamm CM, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm Bowel Dis 2011;17:179–184. 32. Rehman A, Sina C, Gavrilova O, et al. Nod2 is essential for temporal development of intestinal microbial communities. Gut 2011;60:1354–1362.

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

Q2

1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440

1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500

2019

Genetics and Microbe-Based Therapies for IBD

33. Imhann F, Vich Vila A, Bonder MJ, et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018;67:108–119. 34. Petnicki-Ocwieja T, Hrncir T, Liu YJ, et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A 2009;106:15813–15818. 35. Mondot S, Barreau F, Al Nabhani Z, et al. Altered gut microbiota composition in immune-impaired Nod2(/) mice. Gut 2012;61:634–635. 36. Couturier-Maillard A, Secher T, Rehman A, et al. NOD2mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest 2013;123:700–711. 37. Ramanan D, Tang MS, Bowcutt R, et al. Bacterial sensor Nod2 prevents inflammation of the small intestine by restricting the expansion of the commensal Bacteroides vulgatus. Immunity 2014;41:311–324. 38. Alnabhani Z, Hugot JP, Montcuquet N, et al. Respective roles of hematopoietic and nonhematopoietic Nod2 on the gut microbiota and mucosal homeostasis. Inflamm Bowel Dis 2016;22:763–773. 39. Al Nabhani Z, Lepage P, Mauny P, et al. Nod2 deficiency leads to a specific and transmissible mucosa-associated microbial dysbiosis which is independent of the mucosal barrier defect. J Crohns Colitis 2016;10:1428–1436. 40. Rioux JD, Xavier RJ, Taylor KD, et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 2007;39:596–604. 41. Cooney R, Baker J, Brain O, et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med 2010;16:90–97. 42. Travassos LH, Carneiro LA, Ramjeet M, et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol 2010;11:55–62. 43. Murthy A, Li Y, Peng I, et al. A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3. Nature 2014;506:456–462. 44. Saitoh T, Fujita N, Jang MH, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 2008;456:264–268. 45. Cadwell K, Liu JY, Brown SL, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 2008;456:259–263. 46. Kabat AM, Harrison OJ, Riffelmacher T, et al. The autophagy gene Atg16l1 differentially regulates Treg and TH2 cells to control intestinal inflammation. Elife 2016; 5:e12444. 47. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008;453:620–625. 48. Round JL, Mazmanian SK. Inducible Foxp3þ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A 2010; 107:12204–12209. 49. Chu H, Khosravi A, Kusumawardhani IP, et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 2016;352:1116–1120.

13

50. Mitsuyama K, Niwa M, Takedatsu H, et al. Antibody markers in the diagnosis of inflammatory bowel disease. World J Gastroenterol 2016;22:1304–1310. 51. Sokol H, Leducq V, Aschard H, et al. Fungal microbiota dysbiosis in IBD. Gut 2017;66:1039–1048. 52. Iliev ID, Funari VA, Taylor KD, et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 2012;336:1314–1317. 53. Sokol H, Conway KL, Zhang M, et al. Card9 mediates intestinal epithelial cell restitution, T-helper 17 responses, and control of bacterial infection in mice. Gastroenterology 2013;145:591–601 e3. 54. Gross O, Gewies A, Finger K, et al. Card9 controls a nonTLR signalling pathway for innate anti-fungal immunity. Nature 2006;442:651–656. 55. LeibundGut-Landmann S, Gross O, Robinson MJ, et al. Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat Immunol 2007;8:630–638. 56. Glocker EO, Hennigs A, Nabavi M, et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N Engl J Med 2009;361:1727–1735. 57. Hsu YM, Zhang Y, You Y, et al. The adaptor protein CARD9 is required for innate immune responses to intracellular pathogens. Nat Immunol 2007;8:198–205. 58. Lamas B, Richard ML, Leducq V, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med 2016;22:598–605. 59. Doherty PC, Zinkernagel RM. A biological role for the major histocompatibility antigens. Lancet 1975;1:1406–1409. 60. Cleynen I, Boucher G, Jostins L, et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet 2016; 387:156–167. 61. Goyette P, Boucher G, Mallon D, et al. High-density mapping of the MHC identifies a shared role for HLADRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat Genet 2015;47:172–179. 62. Bolnick DI, Snowberg LK, Caporaso JG, et al. Major Histocompatibility Complex class IIb polymorphism influences gut microbiota composition and diversity. Mol Ecol 2014;23:4831–4845. 63. Lin P, Bach M, Asquith M, et al. HLA-B27 and human beta2-microglobulin affect the gut microbiota of transgenic rats. PLoS One 2014;9:e105684. 64. Kubinak JL, Stephens WZ, Soto R, et al. MHC variation sculpts individualized microbial communities that control susceptibility to enteric infection. Nat Commun 2015; 6:8642. 65. Silverman M, Kua L, Tanca A, et al. Protective major histocompatibility complex allele prevents type 1 diabetes by shaping the intestinal microbiota early in ontogeny. Proc Natl Acad Sci U S A 2017;114:9671–9676. 66. De Palma G, Capilla A, Nadal I, et al. Interplay between human leukocyte antigen genes and the microbial colonization process of the newborn intestine. Curr Issues Mol Biol 2010;12:1–10.

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560

REVIEWS AND PERSPECTIVES

-

14

REVIEWS AND PERSPECTIVES

1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 1584 1585 1586 1587 1588 1589 1590 1591 1592 1593 1594 1595 1596 1597 1598 1599 1600 1601 1602 1603 1604 1605 1606 1607 1608 1609 1610 1611 1612 1613 1614 1615 1616 1617 1618 1619 1620

Cohen et al

Gastroenterology Vol.

67. Rosenbaum JT, Davey MP. Time for a gut check: evidence for the hypothesis that HLA-B27 predisposes to ankylosing spondylitis by altering the microbiome. Arthritis Rheum 2011;63:3195–3198. 68. Olivares M, Neef A, Castillejo G, et al. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing coeliac disease. Gut 2015;64:406–417. 69. Scher JU, Sczesnak A, Longman RS, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2013;2:e01202. 70. Gomez A, Luckey D, Yeoman CJ, et al. Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritisresistant 0402 mice. PLoS One 2012;7:e36095. 71. Rath HC, Herfarth HH, Ikeda JS, et al. Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. J Clin Invest 1996; 98:945–953. 72. Johansson ME, Hansson GC. Immunological aspects of intestinal mucus and mucins. Nat Rev Immunol 2016; 16:639–649. 73. Pullan RD, Thomas GA, Rhodes M, et al. Thickness of adherent mucus gel on colonic mucosa in humans and its relevance to colitis. Gut 1994;35:353–359. 74. Johansson ME, Gustafsson JK, Holmen-Larsson J, et al. Bacteria penetrate the normally impenetrable inner colon mucus layer in both murine colitis models and patients with ulcerative colitis. Gut 2014;63:281–291. 75. Van Klinken BJ, Van der Wal JW, Einerhand AW, et al. Sulphation and secretion of the predominant secretory human colonic mucin MUC2 in ulcerative colitis. Gut 1999;44:387–393. 76. Forgue-Lafitte ME, Fabiani B, Levy PP, et al. Abnormal expression of M1/MUC5AC mucin in distal colon of patients with diverticulitis, ulcerative colitis and cancer. Int J Cancer 2007;121:1543–1549. 77. Corfield AP, Myerscough N, Bradfield N, et al. Colonic mucins in ulcerative colitis: evidence for loss of sulfation. Glycoconj J 1996;13:809–822. 78. Larsson JM, Karlsson H, Crespo JG, et al. Altered Oglycosylation profile of MUC2 mucin occurs in active ulcerative colitis and is associated with increased inflammation. Inflamm Bowel Dis 2011;17:2299–2307. 79. Van der Sluis M, De Koning BA, De Bruijn AC, et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 2006;131:117–129. 80. Johansson ME, Phillipson M, Petersson J, et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A 2008;105:15064–15069. 81. Wenzel UA, Magnusson MK, Rydstrom A, et al. Spontaneous colitis in Muc2-deficient mice reflects clinical and cellular features of active ulcerative colitis. PLoS One 2014;9:e100217. 82. Huang H, Fang M, Jostins L, et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 2017;547:173–178.

-,

No.

-

83. O’Connor W Jr, Kamanaka M, Booth CJ, et al. A protective function for interleukin 17A in T cellmediated intestinal inflammation. Nat Immunol 2009; 10:603–609. 84. Zenewicz LA, Yancopoulos GD, Valenzuela DM, et al. Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity 2008;29:947–957. 85. Kumar P, Monin L, Castillo P, et al. Intestinal interleukin17 receptor signaling mediates reciprocal control of the gut microbiota and autoimmune inflammation. Immunity 2016;44:659–671. 86. Flannigan KL, Ngo VL, Geem D, et al. IL-17A–mediated neutrophil recruitment limits expansion of segmented filamentous bacteria. Mucosal Immunol 2017;10:673–684. 87. Hasegawa M, Yada S, Liu MZ, et al. Interleukin-22 regulates the complement system to promote resistance against pathobionts after pathogen-induced intestinal damage. Immunity 2014;41:620–632. 88. Sugimoto K, Ogawa A, Mizoguchi E, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest 2008;118:534–544. 89. Thorburn AN, Macia L, Mackay CR. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity 2014;40:833–842. 90. Cohen LJ, Esterhazy D, Kim SH, et al. Commensal bacteria make GPCR ligands that mimic human signalling molecules. Nature 2017;549:48–53. 91. Ellinghaus D, Folseraas T, Holm K, et al. Genome-wide association analysis in primary sclerosing cholangitis and ulcerative colitis identifies risk loci at GPR35 and TCF4. Hepatology 2013;58:1074–1083. 92. Jostins L, Ripke S, Weersma RK, et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491:119–124. 93. Stappenbeck TS, Virgin HW. Accounting for reciprocal host–microbiome interactions in experimental science. Nature 2016;534:191–199. 94. Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol 2004;4:478–485. 95. Maynard CL, Elson CO, Hatton RD, et al. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012;489:231–241. 96. Kernbauer E, Ding Y, Cadwell K. An enteric virus can replace the beneficial function of commensal bacteria. Nature 2014;516:94–98. 97. Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007;131:33–45. 98. Kiesler P, Fuss IJ, Strober W. Experimental models of inflammatory bowel diseases. Cell Mol Gastroenterol Hepatol 2015;1:154–170. 99. Cominelli F, Arseneau KO, Rodriguez-Palacios A, et al. Uncovering pathogenic mechanisms of inflammatory bowel disease using mouse models of Crohn’s diseaselike ileitis: what is the right model? Cell Mol Gastroenterol Hepatol 2017;4:19–32. 100. Keubler LM, Buettner M, Hager C, et al. A multihit model: colitis lessons from the interleukin-10–deficient mouse. Inflamm Bowel Dis 2015;21:1967–1975.

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

1621 1622 1623 1624 1625 1626 1627 1628 1629 1630 1631 1632 1633 1634 1635 1636 1637 1638 1639 1640 1641 1642 1643 1644 1645 1646 1647 1648 1649 1650 1651 1652 1653 1654 1655 1656 1657 1658 1659 1660 1661 1662 1663 1664 1665 1666 1667 1668 1669 1670 1671 1672 1673 1674 1675 1676 1677 1678 1679 1680

1681 1682 1683 1684 1685 1686 1687 1688 1689 1690 1691 1692 1693 1694 1695 1696 1697 1698 1699 1700 1701 1702 1703 1704 1705 1706 1707 1708 1709 1710 1711 1712 1713 1714 1715 1716 1717 1718 1719 1720 1721 1722 1723 1724 1725 1726 1727 1728 1729 1730 1731 1732 1733 1734 1735 1736 1737 1738 1739 1740

2019

Genetics and Microbe-Based Therapies for IBD

101. Atarashi K, Tanoue T, Shima T, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011;331:337–341. 102. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013;500:232–236. 103. Yissachar N, Zhou Y, Ung L, et al. An intestinal organ culture system uncovers a role for the nervous system in microbe– immune crosstalk. Cell 2017;168:1135–1148 e12. 104. Sefik E, Geva-Zatorsky N, Oh S, et al. Mucosal immunology. Individual intestinal symbionts induce a distinct population of RORgamma(þ) regulatory T cells. Science 2015;349:993–997. 105. Geva-Zatorsky N, Sefik E, Kua L, et al. Mining the human gut microbiota for immunomodulatory organisms. Cell 2017;168:928–943 e11. 106. Atarashi K, Tanoue T, Ando M, et al. Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell 2015;163:367–380. 107. Tan TG, Sefik E, Geva-Zatorsky N, et al. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc Natl Acad Sci U S A 2016;113:E8141–E8150. 108. Viladomiu M, Kivolowitz C, Abdulhamid A, et al. IgAcoated E. coli enriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation. Sci Transl Med 2017;9(376). 109. Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009;139:485–498. 110. Bunker JJ, Flynn TM, Koval JC, et al. Innate and adaptive humoral responses coat distinct commensal bacteria with immunoglobulin A. Immunity 2015;43:541–553. 111. Palm NW, de Zoete MR, Cullen TW, et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 2014;158:1000–1010. 112. Fung TC, Bessman NJ, Hepworth MR, et al. Lymphoid– tissue–resident commensal bacteria promote members of the IL-10 cytokine family to establish mutualism. Immunity 2016;44:634–646. 113. Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 2016;167:1125–1136 e8. 114. Devkota S, Wang Y, Musch MW, et al. Dietary-fat–induced taurocholic acid promotes pathobiont expansion and colitis in Il10/ mice. Nature 2012;487:104–108. 115. Fan D, Coughlin LA, Neubauer MM, et al. Activation of HIF1alpha and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat Med 2015;21:808–814. 116. Helmby H. Human helminth therapy to treat inflammatory disorders—where do we stand? BMC Immunol 2015;16:12. 117. Schnoeller C, Rausch S, Pillai S, et al. A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10–producing macrophages. J Immunol 2008;180:4265–4272. 118. Elliott DE, Summers RW, Weinstock JV. Helminths as governors of immune-mediated inflammation. Int J Parasitol 2007;37:457–464. 119. Figueiredo CA, Barreto ML, Rodrigues LC, et al. Chronic intestinal helminth infections are associated with

15

immune hyporesponsiveness and induction of a regulatory network. Infect Immun 2010;78:3160–3167. 120. Norman JM, Handley SA, Baldridge MT, et al. Diseasespecific alterations in the enteric virome in inflammatory bowel disease. Cell 2015;160:447–460. 121. Eder J, Sedrani R, Wiesmann C. The discovery of first-inclass drugs: origins and evolution. Nat Rev Drug Discov 2014;13:577–587. 122. Koppel N, Balskus EP. Exploring and understanding the biochemical diversity of the human microbiota. Cell Chem Biol 2016;23:18–30. 123. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16:341–352. 124. Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341:569–573. 125. Shimazu T, Hirschey MD, Newman J, et al. Suppression of oxidative stress by beta-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science 2013;339:211–214. 126. Samuel BS, Shaito A, Motoike T, et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proceedings of the National Academy of Sciences of the United States of America 2008; 105:16767–16772. 127. Kim MH, Kang SG, Park JH, et al. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013;145:396–406 e1–10. 128. Chen J, Rao JN, Zou T, et al. Polyamines are required for expression of Toll-like receptor 2 modulating intestinal epithelial barrier integrity. Am J Physiol Gastrointest Liver Physiol 2007;293:G568–G576. 129. Dubin K, Callahan MK, Ren B, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 2016;7:10391. 130. Kibe R, Kurihara S, Sakai Y, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep 2014;4:4548. 131. Liu L, Guo X, Rao JN, et al. Polyamines regulate Ecadherin transcription through c-Myc modulating intestinal epithelial barrier function. Am J Physiol Cell Physiol 2009;296:C801–C810. 132. Cohen LJ, Kang HS, Chu J, et al. Functional metagenomic discovery of bacterial effectors in the human microbiome and isolation of commendamide, a GPCR G2A/132 agonist. Proc Natl Acad Sci U S A 2015; 112:E4825–E4834. 133. Pokusaeva K, Johnson C, Luk B, et al. GABA-producing Bifidobacterium dentium modulates visceral sensitivity in the intestine. Neurogastroenterol Motil 2017;29(1). 134. Williams BB, Van Benschoten AH, Cimermancic P, et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe 2014;16:495–503. 135. Strandwitz P, Kim KH, Terekhova D, et al. GABAmodulating bacteria of the human gut microbiota. Nat Microbiol 2019;4:396–403.

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:45 pm  ce

1741 1742 1743 1744 1745 1746 1747 1748 1749 1750 1751 1752 1753 1754 1755 1756 1757 1758 1759 1760 1761 1762 1763 1764 1765 1766 1767 1768 1769 1770 1771 1772 1773 1774 1775 1776 1777 1778 1779 1780 1781 1782 1783 1784 1785 1786 1787 1788 1789 1790 1791 1792 1793 1794 1795 1796 1797 1798 1799 1800

REVIEWS AND PERSPECTIVES

-

16

REVIEWS AND PERSPECTIVES

1801 1802 1803 1804 1805 1806 1807 1808 1809 1810 1811 1812 1813 1814 1815 1816 1817 1818 1819 1820 1821 1822 1823 1824 1825 1826 1827 1828 1829 1830 1831 1832 1833 1834 1835 1836 1837 1838 1839 1840 1841 1842 1843 1844 1845 1846 1847 1848 1849 1850 1851 1852 1853 1854 1855 1856 1857 1858 1859 1860

Cohen et al

Gastroenterology Vol.

136. Dodd D, Spitzer MH, Van Treuren W, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017;551:648–652. 137. Levy M, Thaiss CA, Zeevi D, et al. Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell 2015; 163:1428–1443. 138. Kaparakis-Liaskos M, Ferrero RL. Immune modulation by bacterial outer membrane vesicles. Nat Rev Immunol 2015;15:375–387. 139. Shen Y, Giardino Torchia ML, Lawson GW, et al. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 2012;12:509–520. 140. Vatanen T, Kostic AD, d’Hennezel E, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell 2016;165:842–853. 141. Donia Mohamed S, Cimermancic P, Schulze Christopher J, et al. A systematic analysis of biosynthetic gene clusters in the human microbiome reveals a common family of antibiotics. Cell 2014;158:1402–1414. 142. Dreher-Lesnick SM, Stibitz S, Carlson PE Jr. U.S. regulatory considerations for development of live biotherapeutic products as drugs. Microbiol Spectr 2017;5(5). 143. Bikard D, Euler CW, Jiang W, et al. Exploiting CRISPRCas nucleases to produce sequence-specific antimicrobials. Nat Biotechnol 2014;32:1146–1150. 144. Paixao AC, Ferreira AC, Fontes M, et al. Detection of virulence-associated genes in pathogenic and commensal avian Escherichia coli isolates. Poult Sci 2016;95:1646–1652. 145. Yasueda A, Mizushima T, Nezu R, et al. The effect of Clostridium butyricum MIYAIRI on the prevention of pouchitis and alteration of the microbiota profile in patients with ulcerative colitis. Surg Today 2016;46:939–949. 146. Krag A, Israelsen H, von Ryberg B, et al. Safety and efficacy of Profermin(R) to induce remission in ulcerative colitis. World J Gastroenterol 2012;18:1773–1780.

-,

No.

-

147. Krag A, Munkholm P, Israelsen H, et al. Profermin is efficacious in patients with active ulcerative colitis—a randomized controlled trial. Inflamm Bowel Dis 2013; 19:2584–2592. 148. Scholmerich J, Fellermann K, Seibold FW, et al. A randomised, double-blind, placebo-controlled trial of Trichuris suis ova in active Crohn’s disease. J Crohns Colitis 2017;11:390–399. 149. Summers R. Trichuris suis seems to be safe and possibly effective in the treatment of inflammatory bowel disease. Am J Gastroenterol 2003;98:2034–2041. 150. Summers RW, Elliott DE, Urban JF, et al. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology 2005;128:825–832. 151. Summers RW, Elliott DE, Urban JF Jr, et al. Trichuris suis therapy in Crohn’s disease. Gut 2005;54:87–90. 152. Mazmanian SK, Liu CH, Tzianabos AO, et al. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005; 122:107–118. 153. Olle B. Medicines from microbiota. Nat Biotechnol 2013; 31:309–315.

Received March 9, 2018. Accepted March 2, 2019. Reprint requests Address requests for reprint to: Louis J. Cohen, MD, Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, 10029. e-mail: [email protected]; or Hiutung Chu, PhD, Department of Pathology, University of California–San Diego, La Jolla, California 92093. e-mail: [email protected]. Conflicts of interest Dirk Gevers is employed by Janssen, which has invested or licensed products from Vedanta, BiomX and Enterome. Funding This work was supported by the National Institutes of Health (grants K08 Q3 DK109287-01 [Louis J. Cohen], U01 DK62429, U01 DK062422, R01 DK092235, R01 DK106593, and R00 DK110534 [Hiutung Chu]) and the Sanford J. Grossman Center for Integrative Studies in Crohn’s Disease (Judy H. Cho).

REV 5.5.0 DTD  YGAST62526_proof  17 April 2019  2:46 pm  ce

1861 1862 1863 1864 1865 1866 1867 1868 1869 1870 1871 1872 1873 1874 1875 1876 1877 1878 1879 1880 1881 1882 1883 1884 1885 1886 1887 1888 1889 1890 1891 1892 1893 1894 1895 1896 1897 1898 1899 1900 1901 1902 1903 1904 1905 1906 1907 1908 1909 1910 1911 1912 1913 1914 1915 1916 1917 1918 1919 1920